-
1Academic Journal
المؤلفون: A. S. Potapov, A. E. Pushkareva, A. V. Vinokurova, M. O. Prokhorenkova, A. E. Krasnovidova, А. С. Потапов, А. Е. Пушкарева, А. В. Винокурова, М. О. Прохоренкова, А. Е. Красновидова
المصدر: Meditsinskiy sovet = Medical Council; № 13 (2024); 124-129 ; Медицинский Совет; № 13 (2024); 124-129 ; 2658-5790 ; 2079-701X
مصطلحات موضوعية: упадацитиниб, Janus kinase inhibitor, targeted immunosuppressants, selective immunosuppressants, small molecules, biological therapy, upadacitinib, ингибитор янус-киназ, таргетные иммуносупрессоры, селективные иммунодепрессанты, малые молекулы, биологическая терапия
وصف الملف: application/pdf
Relation: https://www.med-sovet.pro/jour/article/view/8506/7477; Harbord M, Eliakim R, Bettenworth D, Karmiris K, Katsanos K, Kopylov U et al. Third European Evidence-based Consensus on Diagnosis and Management of Ulcerative Colitis. Part 2: Current Management. J Crohns Colitis. 2017;11(7):769-784. https://doi.org/10.1093/ecco-jcc/jjx009.; Cohen BL, Sachar DB. Update on anti-tumor necrosis factor agents and other new drugs for inflammatory bowel disease. BMJ. 2017;357:j2505. https://doi.org/10.1136/bmj.j2505.; Levin AD, Wildenberg ME, van den Brink GR. Mechanism of Action of Anti-TNF Therapy in Inflammatory Bowel Disease. J Crohns Colitis. 2016;10(8):989-997. https://doi.org/10.1093/ecco-jcc/jjw053.; Roda G, Jharap B, Neeraj N, Colombel JF. Loss of Response to Anti-TNFs: Definition, Epidemiology, and Management. Clin Transl Gastroenterol. 2016;7(1):e135. https://doi.org/10.1038/ctg.2015.63.; Papamichael K, Cheifetz AS. Therapeutic drug monitoring in inflammatory bowel disease: for every patient and every drug? Curr Opin Gastroenterol. 2019;35(4):302-310. https://doi.org/10.1097/MOG.0000000000000536.; Dulai PS, Mosli M, Khanna R, Levesque BG, Sandborn WJ, Feagan BG. Vedolizumab for the treatment of moderately to severely active ulcerative colitis. Pharmacotherapy. 2015;35(4):412-423. https://doi.org/10.1002/phar.1561.; Hajjat TM, Mosha M, Whaley KG, Rosen MJ, Suppa C, Markowitz J et al. Vedolizumab Experience in Children and Adolescents With Inflammatory Bowel Disease: A Multicenter Observational Study. Crohns Colitis 360. 2021;3(3):otab039. https://doi.org/10.1093/crocol/otab039.; Teng MW, Bowman EP, McElwee JJ, Smyth MJ, Casanova JL, Cooper AM, Cua DJ. IL-12 and IL-23 cytokines: from discovery to targeted therapies for immune-mediated inflammatory diseases. Nat Med. 2015;21(7):719-729. https://doi.org/10.1038/nm.3895.; Kim JW, Kim SY. The Era of Janus Kinase Inhibitors for Inflammatory Bowel Disease Treatment. Int J Mol Sci. 2021;22(21):11322. https://doi.org/10.3390/ijms222111322.; De Vries LCS, Wildenberg ME, De Jonge WJ, D'Haens GR. The Future of Janus Kinase Inhibitors in Inflammatory Bowel Disease. J Crohns Colitis. 2017;11(7):885-893. https://doi.org/10.1093/ecco-jcc/jjx003.; Olivera P, Danese S, Peyrin-Biroulet L. JAK inhibition in inflammatory bowel disease. Expert Rev Clin Immunol. 2017;13(7):693-703. https://doi.org/10.1080/1744666X.2017.1291342.; Coskun M, Salem M, Pedersen J, Nielsen OH. Involvement of JAK/STAT signaling in the pathogenesis of inflammatory bowel disease. Pharmacol Res. 2013;76:1-8. https://doi.org/10.1016/j.phrs.2013.06.007.; Núñez P, Quera R, Yarur AJ. Safety of Janus Kinase Inhibitors in Inflammatory Bowel Diseases. Drugs. 2023;83(4):299-314. https://doi.org/10.1007/s40265-023-01840-5.; Garrido I, Lopes S, Macedo G. Hit the Road JAK! The Role of New Oral Treatment in Inflammatory Bowel Disease. Inflamm Bowel Dis. 2021;27(12):2010-2022. https://doi.org/10.1093/ibd/izab037.; Sandborn WJ, Feagan BG, Loftus EV Jr, Peyrin-Biroulet L, Van Assche G, D'Haens G et al. Efficacy and Safety of Upadacitinib in a Randomized Trial of Patients With Crohn's Disease. Gastroenterology. 2020;158(8):2123-2138.e8. https://doi.org/10.1053/j.gastro.2020.01.047.; Irani M, Fan C, Glassner K, Abraham BP. Clinical Evaluation of Upadacitinib in the Treatment of Adults with Moderately to Severely Active Ulcerative Colitis (UC): Patient Selection and Reported Outcomes. Clin Exp Gastroenterol. 2023;16:21-28. https://doi.org/10.2147/CEG.S367086.; Danese S, Vermeire S, Zhou W, Pangan AL, Siffledeen J, Greenbloom S et al. Upadacitinib as induction and maintenance therapy for moderately to severely active ulcerative colitis: results from three phase 3, multicentre, double-blind, randomised trials. Lancet. 2022;399(10341):2113-2128. https://doi.org/10.1016/S0140-6736(22)00581-5.; D'Haens G, Panes J, Louis E, Lacerda A, Zhou Q, Liu J, Loftus EV Jr. Upadacitinib Was Efficacious and Well-tolerated Over 30 Months in Patients With Crohn's Disease in the CELEST Extension Study. Clin Gastroenterol Hepatol. 2022;20(10):2337-2346.e3. https://doi.org/10.1016/j.cgh.2021.12.030.; Chugh R, Braga-Neto MB, Fredrick TW, Ramos GP, Terdiman J, El-Nachef N et al. Multicentre Real-world Experience of Upadacitinib in the Treatment of Crohn's Disease. J Crohns Colitis. 2023;17(4):504-512. https://doi.org/10.1093/ecco-jcc/jjac157.; Miller M, Patel AS, Pasternak B. Rescue therapy with upadacitinib in medically refractory pediatric ulcerative colitis. JPGN Rep. 2024;5(2):197-199. https://doi.org/10.1002/jpr3.12067.; Kontaki E, Merchant A, Gaynor E, Kiparissi F. P494 Efficacy and safety of Upadacitinib in moderate to severe paediatric Crohn's disease and ulcerative colitis in a tertiary Paediatric IBD (PIBD) centre - A case series. J Crohns Colitis. 2024;18(1 Suppl.):i986. https://doi.org/10.1093/ecco-jcc/jjad212.0624.; Spencer EA, Bergstein S, Dolinger M, Pittman N, Kellar A, Dunkin D, Dubinsky MC. Single-center Experience With Upadacitinib for Adolescents With Refractory Inflammatory Bowel Disease. Inflamm Bowel Dis. 2023:izad300. https://doi.org/10.1093/ibd/izad300.; Collen LV. Rapid Clinical Remission With Upadacitinib in a Pediatric Patient With Refractory Crohn's Disease. Inflamm Bowel Dis. 2023;29(7):1175-1176. https://doi.org/10.1093/ibd/izad048.; Martinez-Vinson C, Lengliné H, Viala J. G-PP225 Upadacitinib: The Robert Debre Experience. JPGN Rep. 2024;5(1 Suppl.):S767. https://doi.org/10.1002/jpr3.12073.; Nedelkopoulou N, Sharma S. G-EPV158 Upadacitinib For Difficult-To-Treat Paediatric Ibd; A Single Centre Experience. JPGN Rep. 2024;5(1 Suppl.):S519. https://doi.org/10.1002/jpr3.12073.; https://www.med-sovet.pro/jour/article/view/8506
-
2Academic Journal
المؤلفون: G. V. Volynets, A. I. Khavkin, A. S. Potapov, A. V. Nikitin, Г. В. Волынец, А. И. Хавкин, А. С. Потапов, А. В. Никитин
المصدر: Rossiyskiy Vestnik Perinatologii i Pediatrii (Russian Bulletin of Perinatology and Pediatrics); Том 68, № 6 (2023); 5-14 ; Российский вестник перинатологии и педиатрии; Том 68, № 6 (2023); 5-14 ; 2500-2228 ; 1027-4065
مصطلحات موضوعية: Helicobacter pylori, autoimmune gastritis, Epstein–Barr virus, аутоиммунный гастрит, вирус Эпштейна–Барр
وصف الملف: application/pdf
Relation: https://www.ped-perinatology.ru/jour/article/view/1900/1429; Hall S.N., Appelman H.D. Autoimmune Gastritis. Arch Pathol Lab Med. 2019; 143(11): 1327–1331. DOI:10.5858/arpa.2019–0345-RA; Strickland R.G., Mackay I.R. A reappraisal of the nature and significance of chronic atrophic gastritis. Am J Dig Dis. 1973; 18(5): 426–440. DOI:10.1007/BF01071995; Kamada T., Maruyama Y., Monobe Y., Haruma K. Clinical features and endoscopic findings of autoimmune gastritis and resultant gastric tumor. Dig Endosc. 2022; 34(4): 700–713. DOI:10.1111/den.14175; Malik T.H., Sayahan M.Y., Al Ahmed H.A., Hong X. Gastric intestinal metaplasia: an intermediate precancerous lesion in the cascade of gastric carcinogenesis. J Coll Physicians Surg Pak. 2017; 27(3): 166–172; Massironi S., Zilli A., Elvevi A., Invernizzi P. The changing face of chronic autoimmune atrophic gastritis: an updated comprehensive perspective. Autoimmun Rev. 2019; 18(3): 215–222. DOI:10.1016/j.autrev.2018.08.011; Song J.H., Kim S.G., Jin E.H., Lim J.H., Yang S.Y. Risk factors for gastric tumorigenesis in underlying gastric mucosal atrophy. Gut Liver. 2017; 11(5): 612–619. DOI:10.5009/gnl16488; Saglietti C., Sciarra A., Abdelrahman K., Schneider V., Karpate A., Nydegger A. et al. Autoimmune gastritis in the pediatric age: an underestimated condition report of two cases and review. Front Pediatr. 2018; 6: 123. DOI:10.3389/fped.2018.00123; Rugge M., Genta R.M., Fassan M., Valentini E., Coati I., Guzzinati S. et al. OLGA gastritis staging for the prediction of gastric cancer risk: a long-term follow-up study of 7436 patients. Am J Gastroenterol. 2018; 113(11): 1621–1628. DOI:10.1038/s41395–018–0353–8; Rugge M., Meggio A., Pravadelli C., Barbareschi M., Fassan M., Gentilini M. et al. Gastritis staging in the endoscopic follow-up for the secondary prevention of gastric cancer: a 5-year prospective study of 1755 patients Gut. 2019; 68(1): 11–17. DOI:10.1136/gutjnl-2017–314600; Ikuse T., Blanchard T.G., Czinn S.J. Inflammation, Immunity, and Vaccine Development for the Gastric Pathogen Helicobacter pylori. Curr Top Microbiol Immunol. 2019; 421: 1–19. DOI:10.1007/978–3–030–15138–6_1; Ding S.Z. Global whole family based- Helicobacter pylori eradication strategy to prevent its related diseases and gastric cancer. World J Gastroenterol. 2020; 26(10): 995–1004. DOI:10.3748/wjg.v26.i10.995; Eusebi L.H., Telese A., Marasco G., Bazzoli F., Zagari R.M. Gastric cancer prevention strategies: a global perspective. J Gastroenterol Hepatol. 2020; 35(9): 1495–1502. DOI:10.1111/jgh.15037; Malfertheiner P., Camargo M.C., El-Omar E., Liou J.M., Peek R., Schulz C. et al. Helicobacter pylori infection. Nat Rev Dis Primers. 2023;9(1):19. DOI:10.1038/s41572–023–00431–8; Ihara T., Ihara N., Kushima R., Haruma K. Rapid Progression of Autoimmune Gastritis after Helicobacter pylori Eradication Therapy: A Case Report. Intern Med. 2023; 62(11): 1603–1609. DOI:10.2169/internalmedicine.0533–22; Kotera T., Nishimi Y., Kushima R., Haruma K. Regression of Autoimmune Gastritis after Eradication of Helicobacter pylori. Case Rep Gastroenterol. 2023; 17(1): 34–40. DOI:10.1159/000528388; Sumi N., Haruma K., Urata N. Autoimmune gastritis with rapid development of corporal atrophy found after H. pylori eradication therapy, report of a case. I to Chou (Stomach and Intestine) 2019; 54: 1053–1057 (in Japanese, Abstract in English). DOI:10.11477/mf.1403201789; Zhang Y., Weck M.N., Schöttker B., Rothenbacher D., Brenner H. Gastric parietal cell antibodies, Helicobacter pylori infection, and chronic atrophic gastritis: evidence from a large population-based study in Germany. Cancer Epidemiol Biomarkers Prev. 2013; 22(5): 821–6. DOI:10.1158/1055–9965. EPI-12–1343; Demir A.M., Berberoğlu Ateş B., Hızal G., Yaman A., Tuna Kırsaçlıoğlu C., Oğuz A.S. et al. Autoimmune atrophic gastritis: The role of Helicobacter pylori infection in children. Helicobacter. 2020; 25(5): e12716. DOI:10.1111/hel.12716; Moreira-Silva H., Silva G., Costa E., Guerra I., Santos-Silva E., Tavares M. et al. Insights Into Pediatric Autoimmune Gastritis: Is There a Role for Helicobacter pylori Infection? J Pediatr Gastroenterol Nutr. 2019; 68(6): e99–e104. DOI:10.1097/MPG.0000000000002278; Волынец Г.В. Этиологические факторы хронических га- стритов у детей. Вопросы современной педиатрии. 2006; 5(3): 15–21.; Sarshari B., Mohebbi S.R., Ravanshad M., Shahrokh S., Aghdaei H.A., Zali M.R. Detection and quantification of Epstein– Barr virus, cytomegalovirus, and human herpesvirus-6 in stomach frozen tissue of chronic gastritis and gastric cancer patients. Microbiol Immunol. 2022; 66(8): 379–385. DOI:10.1111/1348–0421.13013; Zhao K., Zhang Y., Xia S., Feng L., Zhou W., Zhang M. et al. Epstein–Barr Virus is Associated with Gastric Cancer Precursor: Atrophic Gastritis. Int J Med Sci. 2022; 19(5): 924–931. DOI:10.7150/ijms.71820; Suzuki Y., Ito S., Nomura K., Matsui A., Kikuchi D., Hoteya S. Multiple Epstein–Barr Virus-associated Gastric Cancers Arising in a Patient with Autoimmune Gastritis. Intern Med. 2023; 62(10): 1459–1466. DOI:10.2169/internalmedicine.0673–22; Yu H., Robertson E.S. Epstein–Barr Virus History and Pathogenesis. Viruses. 2023;15(3):714. DOI:10.3390/v15030714; Rusak E., Chobot A., Krzywicka A., Wenzlau J. Anti-parietal cell antibodies — diagnostic significance. Adv Med Sci. 2016; 61(2): 175–179. DOI:10.1016/j.advms.2015.12.004; Toh B.H. Pathophysiology and laboratory diagnosis of pernicious anemia. Immunol Res. 2017; 65(1): 326–330. DOI:10.1007/s12026–016–8841–7; Волынец Г.В. Хронический гастрит у детей. LAP LAMBERT, Германия, 2013; 356.; Volynets G.V. Chronic gastritis in children. LAP LAMBERT, Germany, 2013; 356. (in Russ.) ISBN: 978–3–659–35473–1; Shah S.C., Piazuelo M.B., Kuipers E.J., Li D. AGA clinical practice update on the diagnosis and management of atrophic gastritis: expert review. Gastroenterology. 2021; 161(4): 1325–1332.e7. DOI:10.1053/j.gastro.2021.06.078; Kulak O., Gurram B., Montgomery E.A., Park J.Y. Pediatric autoimmune gastritis: clinical correlates and histologic features. Hum Pathol. 2021; 116: 31–38. DOI:10.1016/j.humpath.2021.07.002; Волынец Г.В., Хавкин А.И., Никонов Е.Л., Мурашкин В.Ю., Блат С.Ф. Эндоскопически визуализируемые изменения слизистой оболочки верхнего отдела пищеварительного тракта у детей в зависимости от инфекций Helicobacter pylori и Эпштейна–Барр. Доказательная гастроэнтерология. 2018; 7(2): 4–9.; Волынец Г.В., Хавкин А.И., Никонов Е.Л., Мурашкин В.Ю. Особенности морфологических изменений слизистой оболочки желудка у детей в зависимости от инфекции Helicobacter pylori и Эпштейна–Барр-вирусной инфекции. Вопросы детской диетологии. 2018; 16(4): 5–12.; Волынец Г.В., Беляев Д.Л., Виноградова Т.В., Мураш- кин В.Ю., Бабаянц А.А., Шаповалова Т.Г., Семенов А.В. Подходы к лечению аутоиммунного гастрита у детей. Российский вестник перинатологии и педиатрии. 2007; 52(6): 33–39.; https://www.ped-perinatology.ru/jour/article/view/1900
-
3Academic Journal
المؤلفون: Natalia A. Averkina, Madlena E. Bagaeva, Aleksander A. Baranov, Nato D. Vashakmadze, Elena A. Vishneva, Olga S. Gundobina, Nataliya V. Zhurkova, Elena V. Kaitukova, Elena V. Komarova, Tea V. Margieva, Leyla S. Namazova-Baranova, Valeria P. Novikova, Elena E. Petryaykina, Mariya M. Platonova, Aleksander S. Potapov, Olga Ya. Smirnova, Tatiana V. Strokova, Andrey N. Surkov, Nataliya N. Taran, Marina V. Fedoseenko, Nataliya A. Semenova, Inga V. Anisimova, Svetlana A. Repina, Dmitriy M. Subbotin, Valeria V. Sviridova, Anatoliy I. Havkin, Ekaterina A. Yablokova, Galina V. Volynets, Irina V. Sadovnikova, Elena L. Tumanova, Н. А. Аверкина, М. Э. Багаева, А. А. Баранов, Н. Д. Вашакмадзе, Е. А. Вишнева, О. С. Гундобина, Н. В. Журкова, Е. В. Кайтукова, Е. В. Комарова, Т. В. Маргиева, Л. С. Намазова-Баранова, В. П. Новикова, Е. Е. Петряйкина, М. М. Платонова, А. С. Потапов, О. Я. Смирнова, Т. В. Строкова, А. Н. Сурков, Н. Н. Таран, М. В. Федосеенко, Н. А. Семёнова, И. В. Анисимова, С. А. Репина, Д. М. Субботин, В. В. Свиридова, А. И. Хавкин, Е. А. Яблокова, Г. В. Волынец, И. В. Садовникова, Е. Л. Туманова
المساهمون: Not specified., Отсутствует
المصدر: Pediatric pharmacology; Том 21, № 3 (2024); 263-288 ; Педиатрическая фармакология; Том 21, № 3 (2024); 263-288 ; 2500-3089 ; 1727-5776
مصطلحات موضوعية: дети, glycogen-storage disease, glycogenosis, clinical guidelines, children, болезни накопления гликогена, гликогеноз, клинические рекомендации
وصف الملف: application/pdf
Relation: https://www.pedpharma.ru/jour/article/view/2467/1608; Ellingwood SS, Cheng A. Biochemical and Clinical Aspects of Glycogen Storage Diseases. J Endocrinol. 2018;238(3):131–141. https://doi.org/10.1530/JOE-18-0120; Lei KJ, Chen YT, Chen H, et al. Genetic basis of glycogen storage disease type 1a: prevalent mutations at the glucose-6-phosphatase locus. Am J Hum Genet. 1995;57(4):766–771.; Chen YT. Glycogen storage diseases. In: The Metabolic Bases of Inherited Disease. 8th edn. Scriver CR, Beaudet AL, Sly WS, Valle D, eds. New York: McGraw-Hill; 2000. pp. 1521–1551.; Зайчик А.Ш., Чурилов Л.П. Основы общей патологии. Ч. 2. Основы патохимии: учебное пособие для студентов медицинских вузов. — СПб.: ЭЛБИ; 2000. — 688 с.; Brody LC, Abel KJ, Castilla LH, et al. Construction of a transcription map surrounding the BRCA1 locus of human chromosome 17. Genomics. 1995;25(1):238–247. https://doi.org/10.1016/0888-7543(95)80131-5; Цыгин А.Н. Сочетанные заболевания печени и почек у детей // Клиническая нефрология. — 2009. — № 3. — С. 47–51.; Chen YT. Type I glycogen storage disease: kidney involvement, pathogenesis and its treatment. Pediatr Nephrol. 1991;5(1):71–76. https://doi.org/10.1007/BF00852851; Ueno N, Tomita M, Ariga T, et al. Impaired monocyte function in glycogen storage disease type Ib. Eur J Pediatr. 1986;145(4):312–314. https://doi.org/10.1007/BF00439409; Leuzzi R, Banhegyi G, Kardon T, et al. Inhibition of microsomal glucose-6-phosphate transport in human neutrophils results in apoptosis: a potential explanation for neutrophil dysfunction in glycogen storage disease type 1b. Blood. 2003;101(6):2381–2387. https://doi.org/10.1182/blood-2002-08-2576; Shen J, Bao Y, Liu HM, et al. Mutations in exon 3 of the glycogen debranching enzyme gene are associated with glycogen storage disease type III that is differentially expressed in liver and muscle. J Clin Invest. 1996;98(2):352–357. https://doi.org/10.1172/JCI118799; Li XH, Gong QM, Ling Y, et al. Inherent lipid metabolic dysfunction in glycogen storage disease IIIa. Biochem Biophys Res Commun. 2014;455(1-2):90–97. https://doi.org/10.1016/j.bbrc.2014.10.096; Moses SW, Parvari R. The variable presentations of glycogen storage disease type IV: a review of clinical, enzymatic and molecular studies. Curr Mol Med. 2002;2(2):177–188. https://doi.org/10.2174/1566524024605815; Burwinkel B, Bakker HD, Herschkovitz E, et al. Mutations in the liver glycogen phosphorylase gene (PYGL) underlying glycogenosis type VI (Hers disease). Am J Hum Genet. 1998;62(4):785–791. doi: https://doi.org/10.1086/301790; Chang S, Rosenberg MJ, Morton H, et al. Identification of a mutation in liver glycogen phosphorylase in glycogen storage disease type VI. Hum Molec Genet. 1998;7(5):865–870. doi: https://doi.org/10.1093/hmg/7.5.865; Hug G, Schubert WK, Chuck G. Deficient activity of dephoshophosphorylase kinase and accumulation of glycogene in the liver. J Clin Invest. 1969;48(4):704–715. https://doi.org/10.1172/JCI106028; Lerner A, Iancu TC, Bashan N, et al. A new variant of glycogen storage disease. Type IXc. Am J Dis Child. 1982;136(5):406–410. https://doi.org/10.1001/archpedi.1982.03970410024004; Stone WL, Basit H, Adil A. Glycogen Storage Disease. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2024. Available online: https://www.ncbi.nlm.nih.gov/books/NBK459277. Accessed on June 01, 2024.; Ozen H. Glycogen storage diseases: new perspectives. World J Gastroenterol. 2007;13(18):2541–2553. https://doi.org/10.3748/wjg.v13.i18.2541; Kishnani PS, Austin SL, Arn P, et al. Glycogen storage disease type III diagnosis and management guidelines. Genet Med. 2010;12(7):446–463. https://doi.org/10.1097/GIM.0b013e3181e655b6; Hendrickx J, Willems PJ. Genetic deficiencies of the glycogen phosphorylase system. Hum Genet. 1996;97(5):551–556. doi: https://doi.org/10.1007/BF02281858; Краснопольская К.Д. Наследственные болезни обмена веществ: справочное пособие для врачей. — М.: РОО «Центр социальной адаптации и реабилитации детей «Фохат»; 2005. — 364 с.; Rake JP, Visser G, Labrune P, et al. Guidelines for management of glycogen storage disease type I — European Study on Glycogen Storage Disease Type I (ESGSD I). Eur J Pediatr. 2002;161(Suppl 1):S112–S119. https://doi.org/10.1007/s00431-002-1016-7; Kishnani PS, Austin SL, Abdenur JE, et al. Diagnosis and management of glycogen storage disease type I: a practice guideline of the American College of Medical Genetics and Genomics. Genet Med. 2014;16(11):e1. https://doi.org/10.1038/gim.2014.128; Humbert M, Labrune P, Simonneau G. Severe pulmonary arterial hypertension in type1 glycogen storage disease. Eur J Pediatr. 2002;161(Suppl 1):S93–S96. https://doi.org/10.1007/s00431-002-1012-y; Лобзин В.С., Сайкова Л.А., Шиман А.Г. Нервно-мышечные болезни. — СПб.: Гиппократ; 1998. — 224 с. [Lobzin VS, Saikova LA, Shiman AG. Nervno-myshechnye bolezni. St. Petersburg: Gippokrat; 1998. 224 p. (In Russ).]; Visser G, Rake JP, Labrune P, et al. Consensus guidelines for management of glycogen storage disease type 1b — European Study on Glycogen Storage Disease Type 1. Eur J Pediatr. 2002;161(Suppl 1):S120–S123. https://doi.org/10.1007/s00431-002-1017-6; Visser G, Rake JP, Fernandes J, et al. Neutropenia, neutrophil dysfunction, and infl ammatory bowel disease in glycogen storage disease type Ib: results of the European Study on Glycogen Storage Disease type I. J Pediatr. 2000;137;(2):187–191. https://doi.org/10.1067/mpd.2000.105232; Kure S, Hou DC, Suzuki Y, et al. Glycogen storage disease type Ib without neutropenia. J Pediatr. 2000;137(2):253–256. https://doi.org/10.1067/mpd.2000.107472; Melis D, Fulceri R, Parenti G, et al. Genotype/phenotype correlation in glycogen storage disease type 1b: a multicentre study and review of the literature. Eur J Pediatr. 2005;164(8):501–508. https://doi.org/10.1007/s00431-005-1657-4; Shen JJ, Chen YT. Molecular characterization of glycogen storage disease type III. Curr Mol Med. 2002;2(2):167–175. https://doi.org/10.2174/1566524024605752; L’Herminé-Coulomb A, Beuzen F, Bouvier R, et al. Fetal type IV glycogen storage disease: clinical, enzymatic, and genetic data of a pure muscular form with variable and early antenatal manifestations in the same family. Am J Med Genet. 2005;139A(2):118–122. https://doi.org/10.1002/ajmg.a.30945; Szymańska E, Szymańska S, Truszkowska G, et al. Variable clinical presentation of glycogen storage disease type IV: from severe hepatosplenomegaly to cardiac insufficiency. Some discrepancies in genetic and biochemical abnormalities. Arch Med Sci. 2018;14(1):237–247. https://doi.org/10.5114/aoms.2018.72246; Malfatti E, Barnérias C, Hedberg-Oldfors C, et al. A novel neuromuscular form of glycogen storage disease type IV with arthrogryposis, spinal stiffness and rare polyglucosan bodies in muscle. Neuromuscul Disord. 2016;26(10):681–687. https://doi.org/10.1016/j.nmd.2016.07.005; Roscher A, Patel J, Hewson S, et al. The natural history of glycogen storage disease types VI and IX: Long-term outcome from the largest metabolic center in Canada. Mol Genet Metab. 2014;113(3):171–176. https://doi.org/10.1016/j.ymgme.2014.09.005285; Hodax JK, Uysal S, Quintos JB, Phornphutkul C. Glycogen storage disease type IX and growth hormone deficiency presenting as severe ketotic hypoglycemia. J Pediatr Endocrinol Metab. 2017;30(2):247–251. https://doi.org/10.1515/jpem-2016-0342; Сурков А.Н. Гликогеновая болезнь у детей: новые аспекты патогенеза, современные подходы к диагностике, оптимизация ведения пациентов: автореф. дис. … докт. мед. наук. — М.; 2019. — 47 с.; Баранов А.А., Намазова-Баранова Л.С., Сурков А.Н. и др. Гликогеновая болезнь у детей: учебное пособие / Союз педиатров России. — М.: ПедиатрЪ; 2012. — 128 с. — (Болезни детского возраста от А до Я).; Kishnani PS, Goldstein J, Austin SL, et al. Diagnosis and Management of Glycogen Storage Diseases Type VI and IX: a clinical practice resource of the American College of Medical Genetics and Genomics (ACMG). Genet Med. 2019;21(4):772–789. https://doi.org/10.1038/s41436-018-0364-2; Anastasopoulou C. Glycogen Storage Diseases Types I–VII. In: Medscape. Updated: Dec 01, 2022. Available online: https://emedicine.medscape.com/article/1116574-overview. Accessed on June 01, 2024.; Steunenberg TAH, Peeks F, Hoogeveen IJ, et. al. Safety issues associated with dietary management in patients with hepatic glycogen storage disease. Mol Genet Metab. 2018;125(Issues 1-2):79–85. https://doi.org/10.1016/j.ymgme.2018.07.004; Ng K, Mogul DB. Pediatric Liver Tumors. Clin Liver Dis. 2018;22(4):753–772. https://doi.org/10.1016/j.cld.2018.06.008; Laumonier H, Bioulac-Sage P, Laurent C, et al. Hepatocellular adenomas: magnetic resonance imaging features as a function of molecular pathological classification. Hepatology. 2008;48:808–818. https://doi.org/10.1002/hep.22417; Khanna R, Verma SK. Pediatric hepatocellular carcinoma. World J Gastroenterol. 2018;24(35):3980–3999. https://doi.org/10.3748/wjg.v24.i35.3980; Bali DS, Chen YT, Austin S, Goldstein JL. Glycogen Storage Disease Type I. In: GeneReviews® [Internet]. Adam MP, Feldman J, Mirzaa GM, et al., eds. Seattle (WA): University of Washington, Seattle; 1993–2020. Available online: https://www.ncbi.nlm.nih.gov/books/NBK1312. Accessed on June 01, 2024.; Sharari S, Abou-Alloul M, Hussain K, Ahmad Khan F. FanconiBickel Syndrome: A Review of the Mechanisms That Lead to Dysglycaemia. Int J Mol Sci. 2020;21(17):628. https://doi.org/10.3390/ijms21176286; Gupta N, Nambam B, Weinstein DA, Shoemaker LR. Late Diagnosis of Fanconi-Bickel Syndrome: Challenges With the Diagnosis and Literature Review. J Inborn Errors Metab Screen. 2016;4:1–6. https://doi.org/10.1177/2326409816679430; Cori GT. Glycogen structure and enzyme dificiencies in Glycogen Storage Disease. Harvey Lect. 1954;48:145–171.; Дворяковская Г.М., Уварова Е.В., Дворяковский И.В. и др. Роль ультразвуковой диагностики при обследовании детей с печеночной формой гликогенозов // Ультразвуковая и функциональная диагностика. — 2002. — № 4. — С. 53–59.; Pereira NL, Grogan M, Dec GW. Spectrum of Restrictive and Infiltrative Cardiomyopathies: Part 1 of a 2-Part Series. J Am Coll Cardiol. 2018;71(10):1130–1148. https://doi.org/10.1016/j.jacc.2018.01.016; Schreuder AB, Rossi A, Grünert SC, Derks TGJ. Glycogen Storage Disease Type III. In: GeneReviews® [Internet]. Adam MP, Feldman J, Mirzaa GM, et al., eds. Seattle (WA): University of Washington, Seattle; 1993–2020. Available online: https://www.ncbi.nlm.nih.gov/books/NBK26372. Accessed on June 01, 2024.; Mogahed EA, Girgis MY, Sobhy R, et al. Skeletal and cardiac muscle involvement in children with glycogen storage disease type III. Eur J Pediatr. 2015;174(11):1545–1548. https://doi.org/10.1007/s00431-015-2546-0; Magoulas PL, El-Hattab AW. Glycogen Storage Disease Type IV. In: GeneReviews® [Internet]. Adam MP, Feldman J, Mirzaa GM, et al., eds. Seattle (WA): University of Washington, Seattle; 1993–2023. Available online: https://www.ncbi.nlm.nih.gov/books/NBK115333. Accessed on June 01, 2024.; Попович Ю.Г., Чибисов И.В., Потапова-Виноградова И.Н. и др. Клинико-биохимические и морфологические особенности печеночной формы гликогенозов у детей // Педиатрия. Журнал им. Г.Н. Сперанского. — 1988. — № 1. — С. 35–39.; Pathology of the liver. MacSween RNM, Burt AD, Portmann BC, et al., eds. 4th edn. London: Churchill Livingstone; 2001. 982 p.; Клиническая диетология детского возраста: руководство для врачей / под ред. Т.Э. Боровик, К.С. Ладодо. — М.: ООО «Медицинское информационное агентство»; 2008. — 614 с.; Уварова Е.В. Течение гликогеновой болезни печени у детей в условиях комплексной терапии: автореф. дис. … канд. мед. наук. — М.; 2005. — 28 с.; Heller S, Worona L, Consuelo A. Nutritional therapy for glycogen storage diseases. J Pediatr Gastroenterol Nutr. 2008;47(Suppl 1):15–21. https://doi.org/10.1097/MPG.0b013e3181818ea5; Melis D, Della Casa R, Parini R, et al. Vitamin E supplementation improves neutropenia and reduces the frequency of infections in patients with glycogen storage disease type 1b. Eur J Pediatr. 2009;168(9):1069–1074. https://doi.org/10.1007/s00431-008-0889-5; Hicks J, Wartchow E, Mierau G. Glycogen storage diseases: a brief review and update on clinical features, genetic abnormalities, pathologic features, and treatment. Ultrastruct Pathol. 2011;35(5):183–196. https://doi.org/10.3109/01913123.2011.601404; Adeva-Andany MM, Calvo-Castro I, Fernández-Fernández C, et al. Significance of l-carnitine for human health. IUBMB Life. 2017;69(8):578–559. https://doi.org/10.1002/iub.1646; Parikh NS, Ahlawat R. Glycogen Storage Disease Type I. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2024. Available online: https://www.ncbi.nlm.nih.gov/books/NBK534196. Accessed on June 06, 2024.; Ozen H, Ciliv G, Koçak N, et al. Short-term effect of captopril on microalbuminuria in children with glycogen storage disease type Ia. J Inherit Metab Dis. 2000;23(5):459–463. https://doi.org/10.1023/a:1005608113270; Quinlivan R, Martinuzzi A, Schoser B. Pharmacological and nutritional treatment for McArdle disease (Glycogen Storage Disease type V). Cochrane Database Syst Rev. 2014;2014(11):CD003458. https://doi.org/10.1002/14651858.CD003458.pub5; Martinuzzi A, Liava A, Trevisi E, et al. Randomized, placebocontrolled, double-blind pilot trial of ramipril in McArdle’s disease. Muscle Nerve. 2008;37(3):350–357. https://doi.org/10.1002/mus.20937; Wicker C, Roda C, Perry A, et al. Infectious and digestive complications in glycogen storage disease type Ib: Study of a French cohort. Mol Genet Metab Rep. 2020;23:100581. https://doi.org/10.1016/j.ymgmr.2020.100581; Visser G, Rake JP, Labrune P, et al. Granulocyte colonystimulating factor in glycogen storage disease type 1b. Results of the European Study on Glycogen Storage Disease Type 1. Eur J Pediatr. 2002;161(Suppl 1):S83–S87. https://doi.org/10.1007/s00431-002-1010-0; Dale DC, Bolyard AA, Marrero T, et al. Neutropenia in glycogen storage disease Ib: outcomes for patients treated with granulocyte colony-stimulating factor. Curr Opin Hematol. 2019;26(1):16–21. https://doi.org/10.1097/MOH.0000000000000474; Melis D, Parenti G, Della Casa R, et al. Crohn’s-like ileo-colitis in patients affected by glycogen storage disease Ib: two years’ follow-up of patients with a wide spectrum of gastrointestinal signs. Acta Paediatr. 2003;92(12):1415–1421. https://doi.org/10.1080/08035250310007033; Melis D, Della Casa R, Balivo F, et al. Involvement of endocrine system in a patient affected by glycogen storage disease 1b: speculation on the role of autoimmunity. Ital J Pediatr. 2014;40(1):30. https://doi.org/10.1186/1824-7288-40-30; Gong YZ, Zhong XM, Zou JZ. Infliximab treatment of glycogenosis Ib with Crohn’s-like enterocolitis: A case report. World J Clin Cases. 2021;9(19):5280–5286. https://doi.org/10.12998/wjcc.v9.i19.5280; Davis MK, Rufo PA, Polyak SF, Weinstein DA. Adalimumab for the treatment of Crohn-like colitis and enteritis in glycogen storage disease type Ib. J Inherit Metab Dis. 2008;31(Suppl 3):505–509. https://doi.org/10.1007/s10545-007-0774-9; Готье С.В., Цирульникова О.М., Мнацаканян Д.С. и др. Трансплантация печени у детей с болезнями накопления гликогена: оценка риска и необходимость ее проведения // Вестник трансплантологии и искусственных органов. — 2013. — Т. 15. — № 1. — С. 67–74. — https://doi.org/10.15825/1995-1191-2013-1-67-74; Филин А .В., С еменков А .В., Короте ева Н. А . и др. Родственная пересадка фрагментов печени при гликогенозах I типа: первый российский опыт // Трансплантология. — 2011 . — № 2–3. — С. 24–28 . — doi: ht tps://doi.org/10.23873/20740506-20110-2-3-24-28; Boers SJ, Visser G, Smit PG, Fuchs SA. Liver transplantation in glycogen storage disease type I. Orphanet J Rare Dis. 2014;9:47. https://doi.org/10.1186/1750-1172-9-47; Oshita A, Itamoto T, Amano H, et al. Perioperative management of benign hepatic tumors in patients with glycogen storage disease type Ia. J Hepatobiliary Pancreat Surg. 2008;15(2):200–203. https://doi.org/10.1007/s00534-007-1244-3; Brady MT. Immunization recommendations for children with metabolic disorders: more data would help. Pediatrics. 2006;118(2):810–813. https://doi.org/10.1542/peds.2006-0846.; Cerutti M, De Lonlay P, Menni F, et al. Vaccination coverage of patients with inborn errors of metabolism and the attitudes of their parents towards vaccines. Vaccine. 2015;33(48):6520–6524. https://doi.org/10.1016/j.vaccine.2015.10.073; https://www.pedpharma.ru/jour/article/view/2467
-
4Academic Journal
المؤلفون: G. V. Volynets, A. S. Potapov, A. V. Nikitin, L. G. Danilov, T. A. Skvortsova, V. V. Dudurich, Г. В. Волынец, А. С. Потапов, А. В. Никитин, Л. Г. Данилов, Т. А. Скворцова, В. В. Дудурич
المصدر: Rossiyskiy Vestnik Perinatologii i Pediatrii (Russian Bulletin of Perinatology and Pediatrics); Том 68, № 4 (2023); 66-76 ; Российский вестник перинатологии и педиатрии; Том 68, № 4 (2023); 66-76 ; 2500-2228 ; 1027-4065
مصطلحات موضوعية: иммуносупрессивная терапия, intestinal microbiota, chronic liver disease, autoimmune liver disease, non-autoimmune liver disease, immunosuppressive therapy, кишечная микробиота, хронические болезни печени, аутоиммунные болезни печени, неаутоиммунные болезни печени
وصف الملف: application/pdf
Relation: https://www.ped-perinatology.ru/jour/article/view/1850/1396; Mieli-Vergani G., Vergani D., Baumann U., Czubkowski P., Debray D., Dezsofi A. et al. Diagnosis and Management of Pediatric Autoimmune Liver Disease: ESPGHAN Hepatology Committee Position Statement. J Pediatr Gastroenterol Nutr 2018; 66(2): 345–360. DOI:10.1097/MPG.0000000000001801; Webb G.J., Hirschfield G.M., Krawitt E.L., Gershwin M.E. Cellular and Molecular Mechanisms of Autoimmune Hepatitis. Annu Rev Pathol 2018; 13: 247–292. DOI:10.1146/annurev-pathol-020117–043534; Mieli-Vergani G., Vergani D., Czaja A.J., Manns M.P., Krawitt E.L., Vierling J.M. et al. Autoimmune hepatitis. Nat Rev Dis Primers 2018; 4: 18017. DOI:10.1038/nrdp.2018.17; European Association for the Study of the Liver. EASL Clinical Practice Guidelines: Autoimmune hepatitis. J Hepatol 2015; 63(4): 971–1004. DOI:10.1016/j.jhep.2015.06.030; Corrigan M., Hirschfield G.M., Oo Y.H., Adams D.H. Autoimmune hepatitis: an approach to disease understanding and management. Br Med Bull 2015; 114(1): 181–91. DOI:10.1093/bmb/ldv021; Manns M.P., Lohse A.W., Vergani D. Autoimmune hepatitis — Update 2015. J Hepatol 2015; 62(1 Suppl): S100–11. DOI:10.1016/j.jhep.2015.03.005; de Boer Y.S., van Gerven N.M., Zwiers A., Verwer B.J., van Hoek B., van Erpecum K.J. et al.; Dutch Autoimmune Hepatitis Study Group; LifeLines Cohort Study; Study of Health in Pomerania. Genome-wide association study identifies variants associated with autoimmune hepatitis type 1. Gastroenterology 2014; 147(2): 443–52.e5. DOI:10.1053/j.gastro.2014.04.022; Webb G.J., Hirschfield G.M. Using GWAS to identify genetic predisposition in hepatic autoimmunity. J Autoimmun 2016; 66: 25–39. DOI:10.1016/j.jaut.2015.08.016; Adolph T.E., Grander C., Moschen A.R., Tilg H. Liver-Microbiome Axis in Health and Disease. Trends Immunol 2018; 39(9): 712–723. DOI:10.1016/j.it.2018.05.002; Kummen M., Holm K., Anmarkrud J.A., Nygård S., Vesterhus M., Høivik M.L. et al. The gut microbial profile in patients with primary sclerosing cholangitis is distinct from patients with ulcerative colitis without biliary disease and healthy controls. Gut 2017; 66(4): 611–619. DOI:10.1136/gutjnl-2015–310500; Sabino J., Vieira-Silva S., Machiels K., Joossens M., Falony G., Ballet V. et al. Primary sclerosing cholangitis is characterised by intestinal dysbiosis independent from IBD. Gut 2016; 65(10): 1681–1689. DOI:10.1136/gutjnl-2015–311004; Tang R., Wei Y., Li Y., Chen W., Chen H., Wang Q. et al. Gut microbial profile is altered in primary biliary cholangitis and partially restored after UDCA therapy. Gut 2018; 67(3): 534–541. DOI:10.1136/gutjnl-2016–313332; Tripathi A., Debelius J., Brenner D.A., Karin M., Loomba R., Schnabl B. et al. The gut-liver axis and the intersection with the microbiome. Nat Rev Gastroenterol Hepatol 2018; 15(7): 397–411. DOI:10.1038/s41575–018–0011-z; Yuksel M., Wang Y., Tai N., Peng J., Guo J., Beland K. et al. A novel «humanized mouse» model for autoimmune hepatitis and the association of gut microbiota with liver inflammation. Hepatology 2015; 62(5): 1536–1550. DOI:10.1002/hep.27998; Manfredo Vieira S., Hiltensperger M., Kumar V., Zegarra-Ruiz D., Dehner C., Khan N. et al. Translocation of a gut pathobiont drives autoimmunity in mice and humans. Science 2018; 359(6380): 1156–1161. DOI:10.1126/science.aar7201; Abe K., Takahashi A., Fujita M., Imaizumi H., Hayashi M., Okai K. et al. Dysbiosis of oral microbiota and its association with salivary immunological biomarkers in autoimmune liver disease. PLoS One 2018; 13(7): e0198757. DOI:10.1371/journal.pone.0198757; Lv L., Jiang H., Chen X., Wang Q., Wang K., Ye J. et al. The Salivary Microbiota of Patients With Primary Biliary Cholangitis Is Distinctive and Pathogenic. Front Immunol 2021; 12: 713647. DOI:10.3389/fimmu.2021.713647; He Y., Wu W., Zheng H.M., Li P., McDonald D., Sheng H.F. et al. Regional variation limits applications of healthy gut microbiome reference ranges and disease models. Nat Med 2018; 24(10): 1532–1535. DOI:10.1038/s41591–018–0164-x; Huttenhower C., Gevers D., Knight R., Abubucker S., Badger J.H., Chinwalla A.T. et al. Human Microbiome Project Consortium. Structure, function and diversity of the healthy human microbiome. Nature 2012; 486(7402): 207–14. DOI:10.1038/nature11234; David L.A., Maurice C.F., Carmody R.N., Gootenberg D.B., Button J.E., Wolfe B.E. et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature 2014; 505(7484): 559–563. DOI:10.1038/nature12820; Sonnenburg E.D., Smits S.A., Tikhonov M., Higginbottom S.K., Wingreen N.S., Sonnenburg J.L. Diet-induced extinctions in the gut microbiota compound over generations. Nature 2016; 529(7585): 212–215. DOI:10.1038/nature16504; Modi S.R., Collins J.J., Relman D.A. Antibiotics and the gut microbiota. Clin Invest 2014; 124(10): 4212–4128. DOI:10.1172/JCI72333; Maurice C.F., Haiser H.J., Turnbaugh P.J. Xenobiotics shape the physiology and gene expression of the active human gut microbiome. Cell 2013; 152(1–2): 39–50. DOI:10.1016/j.cell.2012.10.052; Gabarre P., Loens C., Tamzali Y., Barrou B., Jaisser F., Tourret J. Immunosuppressive therapy after solid organ transplantation and the gut microbiota: Bidirectional interactions with clinical consequences. Am J Transplant 2022; 22(4): 1014–1030. DOI:10.1111/ajt.16836; Sonnenburg J.L., Backhed F. Diet-microbiota interactions as moderators of human metabolism. Nature 2016; 535(7610): 56–64. DOI:10.1038/nature18846; Callahan B.J., McMurdie P.J., Rosen M.J., Han A.W., Johnson A.J., Holmes S.P. DADA2: High-resolution sample inference from Illumina amplicon data. Nat Methods 2016; 13(7): 581–583. DOI:10.1038/nmeth.3869; Wei Y., Li Y., Yan L., Sun C., Miao Q., Wang Q. et al. Alterations of gut microbiome in autoimmune hepatitis. Gut 2020; 69(3): 569–577. DOI:10.1136/gutjnl-2018–317836; Zhao L. The gut microbiota and obesity: from correlation to causality. Nat Rev Microbiol 2013; 11: 639–647. DOI:10.1038/nrmicro3089; Imajo K., Fujita K., Yoneda M., Nozaki Y., Ogawa Y., Shinohara Y. et al. Hyperresponsivity tolow-dose endotoxin during progression to nonalcoholic steatohepatitis isregulated by leptin-mediated signaling. Cell Metab 2012; 16: 44–54. DOI:10.1016/j.cmet.2012.05.012; Gevers D., Kugathasan S., Denson L.A., Vázquez-Baeza Y., Van Treuren W., Ren B. et al. The treatment-naive microbiome in new-onset Crohn’s disease. Cell Host Microbe 2014; 15: 382–392. DOI:10.1016/j.chom.2014.02.005; Akberova D., Kiassov A.P., Abdulganieva D. Serum Cytokine Levels and Their Relation to Clinical Features in Patients with Autoimmune Liver Diseases. J Immunol Res 2017; 2017: 9829436. DOI:10.1155/2017/9829436; Yoshida N., Emoto T., Yamashita T., Watanabe H., Hayashi T., Tabata T. et al. Bacteroides vulgatus and Bacteroides dorei Reduce Gut Microbial Lipopolysaccharide Production and Inhibit Atherosclerosis. Circulation 2018; 138(22): 2486–2498. DOI:10.1161/CIRCULATIONAHA.118.033714; Koh A., De Vadder F., Kovatcheva-Datchary P., Bäckhed F. From Dietary Fiber to Host Physiology: Short-Chain Fatty Acids as Key Bacterial Metabolites. Cell 2016; 165(6): 1332–1345. DOI:10.1016/j.cell.2016.05.041; Kriss M., Hazleton K.Z., Nusbacher N.M., Martin C.G., Lozupone C.A. Low diversity gut microbiota dysbiosis: drivers, functional implications and recovery. Curr Opin Microbiol 2018; 44: 34–40. DOI:10.1016/j.mib.2018.07.003; Kelly C.J., Zheng L., Campbell E.L., Saeedi B., Scholz C.C., Bayless A.J. et al. Crosstalk between Microbiota-Derived Short-Chain Fatty Acids and Intestinal Epithelial HIF Augments Tissue Barrier Function. Cell Host Microbe 2015; 17(5): 662–671. DOI:10.1016/j.chom.2015.03.005; Wu T., Yang L., Jiang J., Ni Y., Zhu J., Zheng X. et al. Chronic glucocorticoid treatment induced circadian clock disorder leads to lipid metabolism and gut microbiota alterations in rats. Life Sci 2018; 192: 173–182. DOI:10.1016/j.lfs.2017.11.049; Tourret J., Willing B.P., Dion S., MacPherson J., Denamur E., Finlay B.B. Immunosuppressive Treatment Alters Secretion of Ileal Antimicrobial Peptides and Gut Microbiota, and Favors Subsequent Colonization by Uropathogenic Escherichia coli. Transplantation 2017; 101(1): 74–82. DOI:10.1097/TP.0000000000001492; He Z., Kong X., Shao T., Zhang Y., Wen C. Alterations of the Gut Microbiota Associated With Promoting Efficacy of Prednisone by Bromofuranone in MRL/lpr Mice. Front Microbiol 2019; 10: 978. DOI:10.3389/fmicb.2019.00978; Huang E.Y., Inoue T., Leone V.A., Dalal S., Touw K., Wang Y. et al. Using corticosteroids to reshape the gut microbiome: implications for inflammatory bowel diseases. Inflamm Bowel Dis 2015; 21(5): 963–972. DOI:10.1097/MIB.0000000000000332; Steiner R.W., Awdishu L. Steroids in kidney transplant patients. Semin Immunopathol 2011; 33(2): 157–167. DOI:10.1007/s00281–011–0259–7; Rodríguez-Piñeiro A.M., Johansson M.E. The colonic mucus protection depends on the microbiota. Gut Microbes 2015; 6(5): 326–330. DOI:10.1080/19490976.2015.1086057; Bunker J.J., Flynn T.M., Koval J.C., Shaw D.G., Meisel M., McDonald B.D. et al. Innate and Adaptive Humoral Responses Coat Distinct Commensal Bacteria with Immunoglobulin A. Immunity 2015; 43(3): 541–553. DOI:10.1016/j.immuni.2015.08.007; Sommer F., Anderson J.M., Bharti R., Raes J., Rosenstiel P. The resilience of the intestinal microbiota influences health and disease. Nat Rev Microbiol 2017; 15(10): 630–638. DOI:10.1038/nrmicro.2017.58; Kim H.B., Wang Y., Sun X. A Detrimental Role of Immunosuppressive Drug, Dexamethasone, During Clostridium difficile Infection in Association with a Gastrointestinal Microbial Shift. J Microbiol Biotechnol 2016; 26(3): 567–571. DOI:10.4014/jmb.1512.12017; Liu F., Ma R., Riordan S.M., Grimm M.C., Liu L., Wang Y. et al. Azathioprine, Mercaptopurine, and 5-Aminosalicylic Acid Affect the Growth of IBD-Associated Campylobacter Species and Other Enteric Microbes. Front Microbiol 2017; 8: 527. DOI:10.3389/fmicb.2017.00527; Swidsinski A., Loening-Baucke V., Bengmark S., Lochs H., Dörffel Y. Azathioprine and mesalazine-induced effects on the mucosal flora in patients with IBD colitis. Inflamm Bowel Dis 2007; 13(1): 51–56. DOI:10.1002/ibd.20003; Mousa O.Y., Juran B.D., McCauley B.M., Vesterhus M.N., Folseraas T., Turgeon C.T. et al. Bile Acid Profiles in Primary Sclerosing Cholangitis and Their Ability to Predict Hepatic Decompensation. Hepatology 2021; 74(1): 281–295. DOI:10.1002/hep.31652; Watanabe Y., Nagai F, Morotomi M. Characterization of Phascolarctobacterium succinatutens sp. nov., an asaccharolytic, succinate-utilizing bacterium isolated from human feces. Appl Environ Microbiol 2012; 78(2): 511–518. DOI:10.1128/AEM.06035–11; Morgan X.C., Tickle T.L., Sokol H., Gevers D., Devaney K.L., Ward D.V. et al. Dysfunction of the intestinal microbiome in inflammatory bowel disease and treatment. Genome Biol 2012; 13(9): R79. DOI:10.1186/gb-2012–13–9-r79; Horvath T.D., Ihekweazu F.D., Haidacher S.J., Ruan W., Engevik K.A., Fultz R. et al. Bacteroides ovatus colonization influences the abundance of intestinal short chain fatty acids and neurotransmitters. Science 2022; 25(5): 104158. DOI:10.1016/j.isci.2022.104158; https://www.ped-perinatology.ru/jour/article/view/1850
-
5Academic Journal
المؤلفون: G. V. Volynets, A. V. Nikitin, T. A. Skvortsova, A. S. Potapov, V. V. Dudurich, L. G. Danilov, Г. В. Волынец, А. В. Никитин, Т. А. Скворцова, А. С. Потапов, В. В. Дудурич, Л. Г. Данилов
المصدر: Rossiyskiy Vestnik Perinatologii i Pediatrii (Russian Bulletin of Perinatology and Pediatrics); Том 68, № 2 (2023); 69-73 ; Российский вестник перинатологии и педиатрии; Том 68, № 2 (2023); 69-73 ; 2500-2228 ; 1027-4065
مصطلحات موضوعية: хронические болезни печени, gut microbiota, chronic liver disease, кишечная микробиота
وصف الملف: application/pdf
Relation: https://www.ped-perinatology.ru/jour/article/view/1803/1363; Xu X.R., Liu C.Q., Feng B.S., Liu Z.J. Dysregulation of mucosal immune response in pathogenesis of inflammatory bowel disease. World J Gastroenterol 2014; 20(12): 3255–3264. DOI:10.3748/wjg.v20.i12.3255; Carrière J., Darfeuille-Michaud A., Nguyen H.T. Infectious etiopathogenesis of Crohn’s disease. World J Gastroenterol 2014; 20(34): 12102–12117. DOI:10.3748/wjg.v20.i34.12102; Abraham C., Cho J.H. Inflammatory bowel disease. N Engl J Med 2009; 361(21): 2066–2078. DOI:10.1056/NEJMra0804647; Kaser A., Zeissig S., Blumberg R.S. Inflammatory bowel disease. Annu Rev Immunol 2010; 28: 573–621. DOI:10.1146/annurev-immunol-030409–101225; David L.A., Maurice C.F., Carmody R.N., Gootenberg D.B., Button J.E., Wolfe B.E. et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature 2014; 505(7484): 559–563. DOI:10.1038/nature12820; Sonnenburg E.D., Smits S.A., Tikhonov M., Higginbottom S.K., Wingreen N.S., Sonnenburg J.L. Diet-induced extinctions in the gut microbiota compound over generations. Nature 2016; 529(7585): 212–215. DOI:10.1038/nature16504; Modi S.R., Collins J.J., Relman D.A. Antibiotics and the gut microbiota. Clin Invest 2014; 124(10): 4212–4218. DOI:10.1172/JCI72333; Maurice C.F., Haiser H.J., Turnbaugh P.J. Xenobiotics shape the physiology and gene expression of the active human gut microbiome. Cell 2013; 152(1–2): 39–50. DOI:10.1016/j.cell.2012.10.052; Sonnenburg J.L., Backhed F. Diet-microbiota interactions as moderators of human metabolism. Nature 2016; 535(7610): 56–64. DOI:10.1038/nature18846; Huttenhower C., Gevers D., Knight R., Abubucker S., Badger J.H., Chinwalla A.T. et al. Human Microbiome Project Consortium. Structure, function and diversity of the healthy human microbiome. Nature 2012; 486(7402): 207–214. DOI:10.1038/nature11234; Gevers D., Kugathasan S., Denson L.A., Vázquez-Baeza Y., Van Treuren W., Ren B. et al. The treatment-naive microbiome in new-onset Crohn’s disease. Cell Host Microbe 2014; 15(3): 382–392. DOI:10.1016/j.chom.2014.02.005; Kummen M., Hov J.R. The gut microbial influence on cholestatic liver disease. Liver Int 2019; 39(7): 1186–1196. DOI:10.1111/liv.14153; Nakamoto N., Sasaki N., Aoki R., Miyamoto K., Suda W., Teratani T. et al. Gut pathobionts underlie intestinal barrier dysfunction and liver T helper 17 cell immune response in primary sclerosing cholangitis. Nat Microbiol 2019; 4(3): 492– 503. DOI:10.1038/s41564–018–0333–1; Liao L., Schneider K.M., Galvez E.J.C., Frissen M., Marschall H.U., Su H. et al. Intestinal dysbiosis augments liver disease progression via NLRP3 in a murine model of primary sclerosing cholangitis. Gut 2019; 68(8): 1477–1492. DOI:10.1136/gutjnl-2018–316670; Волынец Г.В., Никитин А.В., Скворцова Т.А. Кишечный микробном и современные методы его исследования у детей. Российский вестник перинатологии и педиатрии 2022: 67 (4): 5–13. DOI:10.21508/1027–4065–2022–67–4–5–13; https://www.ped-perinatology.ru/jour/article/view/1803
-
6Academic Journal
المؤلفون: O. V. Kurbatova, S. V. Petrichuk, D. G. Kuptsova, G. B. Movsisyan, T. V. Radygina, A. D. Komarova, A. O. Anushenko, E. V. Freidlin, E. L. Semikina, A. S. Potapov, A. P. Fisenko, О. В. Курбатова, С. В. Петричук, Д. Г. Купцова, Г. Б. Мовсисян, Т. В. Радыгина, А. Д. Комарова, А. О. Анушенко, Е. В. Фрейдлин, Е. Л. Семикина, А. С. Потапов, А. П. Фисенко
المصدر: Medical Immunology (Russia); Том 25, № 5 (2023); 1205-1212 ; Медицинская иммунология; Том 25, № 5 (2023); 1205-1212 ; 2313-741X ; 1563-0625
مصطلحات موضوعية: NF-kB, Wilson-Konovalov disease, lymphocytes, Th17, Tc17, flow cytometry with visualization, болезнь Вильсона-Коновалова, лимфоциты, проточная цитометрия с визуализацией
وصف الملف: application/pdf
Relation: https://www.mimmun.ru/mimmun/article/view/2799/1747; https://www.mimmun.ru/mimmun/article/downloadSuppFile/2799/11727; https://www.mimmun.ru/mimmun/article/downloadSuppFile/2799/11728; https://www.mimmun.ru/mimmun/article/downloadSuppFile/2799/11729; https://www.mimmun.ru/mimmun/article/downloadSuppFile/2799/11731; https://www.mimmun.ru/mimmun/article/downloadSuppFile/2799/11732; https://www.mimmun.ru/mimmun/article/downloadSuppFile/2799/11733; https://www.mimmun.ru/mimmun/article/downloadSuppFile/2799/11735; https://www.mimmun.ru/mimmun/article/downloadSuppFile/2799/12159; https://www.mimmun.ru/mimmun/article/downloadSuppFile/2799/12160; https://www.mimmun.ru/mimmun/article/downloadSuppFile/2799/12171; https://www.mimmun.ru/mimmun/article/downloadSuppFile/2799/12172; https://www.mimmun.ru/mimmun/article/downloadSuppFile/2799/12173; https://www.mimmun.ru/mimmun/article/downloadSuppFile/2799/12177; Barteneva N.S., Vorobjev I.A. Imaging Flow Cytometry Methods and protocols. Methods Mol. Biol., 2017, pp. 178-188.; Deng H., Zhu S., Yang H., Cui H., Guo H., Deng J., Ren Z., Geng Y., Ouyang P., Xu Z., Deng Y., Zhu Y. Dysregulation of inflammatory pathways triggered by copper exposure. Biol. Trace Elem. Res., 2023, Vol. 201, no. 2, pp. 539-548.; Fisenko A.P., Surkov N.N., Potapov A.S. Wilson's disease in children. Moscow: National Medical Research Center of Children's Health, 2019. Р. 83. (In Russ.); Kuptsova D.G., Petrichuk S.V., Murashkin N.N., Kurbatova O.V., Radygina T.V., Khotko A.A., Ivanov R.A. Activity of nuclear factor kB in lymphocyte populations of children with psoriasis. Bulletin of the Russian State Medical University, 2022, no. 2, pp. 28-35. (In Russ.); Kurbatova O.V., Kuptsova D.G., Bezrukavnikova L.M., Radygina T.V., Movsisyan G.B., Anushenko A.O., Komarova A.D., Potapov A.S., Petrichuk S.V, Fisenko A.P. Features of immune status in children with Wilson-Konovalov disease at different stages of liver fibrosis. Russian Journal of Immunology, 2022, Vol. 25, no. 4, pp. 453-460. (In Russ.); Lucena-Valera A., Ruz-Zafra P., Ampuero J. Wilson's disease: overview. Enfermedad de Wilson. Med. Clin. (Barc.), 2023, Vol. 160, no. 6, pp. 261-267.; Luedde T., Schwabe R.F. NF-KB in the liver-linking injury, fibrosis and hepatocellular carcinoma. Nat. Rev. Gastroenterol. Hepatol., 2011, Vol. 8, no. 2, pp. 108-118.; Mehto S., Kundu S., Chauhan S., Chauhan S. RIPosomes are targets of IRGM-SQSTM1-dependent autophagy. Autophagy, 2023, Vol. 19, no. 3, pp. 1045-1047.; Oakley F., Mann J., Nailard S., Smart D., Mungalsingh N., Constandinou C., Ali S., Wilson S.J., Millward- Sadler H., Iredale J.P., Mann D.A. Nuclear factor-kappaB1 (p50) limits the inflammatory and fibrogenic responses to chronic injury. Am. J. Pathol., 2005, Vol. 166, no. 3, pp. 695-708.; Petrichuk S.V., Radygina T.V., Kuptsova D.G., Kurbatova O.V., Semikina E.L., Murashkin N.N., Potapov A.S., Fisenko A.P. Evaluation of anti-TNF treatment efficiency in children with immune-dependent diseases by means of testing the NF-kB activity in lymphocyte populations. Russian Journal of Immunology, 2022, Vol. 25, no. 4, pp. 491-498. (In Russ.); Petrichuk S., Radigina T., Gerasimova D., Illarionov A., Anushenko A., Erlikh-Fox T., Potapov A., P070 The level of nuclear factor kappa B (NF-kB) translocation during infliximab therapy in children with IBD. J. Crohns Colitis, 2019, Vol.13, Iss. suppl. 1, pp. S120-S121.; Przybylkowski A., Szeligowska J., Januszewicz M., Raszeja-Wyszomirska J., Szczepankiewicz B., Nehring P, Gornicka B., Litwin T., Czlonkowska A. Evaluation of liver fibrosis in patients with Wilson's disease. Eur. J. Gastroenterol. Hepatol., 2021, Vol. 33, no. 4, pp. 535-540.; Zhang T., Hu J., Wang X., Zhao X., Li Z., Niu J., Steer C.J Zheng G., Song G. MicroRNA-378 promotes hepatic inflammation and fibrosis via modulation of the NF-KB-TNFa pathway. J Hepatol., 2019, Vol. 70, no. 1, pp. 87-96.; https://www.mimmun.ru/mimmun/article/view/2799
-
7Academic Journal
المؤلفون: T. V. Radygina, E. G. Sorokina, S. V. Petrichuk, D. G. Kuptsova, O. V. Kurbatova, A. S. Potapov, S. A. Afanasyeva, Т. В. Радыгина, Е. Г. Сорокина, С. В. Петричук, Д. Г. Купцова, О. В. Курбатова, А. С. Потапов, С. А. Афанасьева
المصدر: Medical Immunology (Russia); Том 25, № 2 (2023); 415-422 ; Медицинская иммунология; Том 25, № 2 (2023); 415-422 ; 2313-741X ; 1563-0625
وصف الملف: application/pdf
Relation: https://www.mimmun.ru/mimmun/article/view/2616/1655; https://www.mimmun.ru/mimmun/article/downloadSuppFile/2616/10343; https://www.mimmun.ru/mimmun/article/downloadSuppFile/2616/10344; https://www.mimmun.ru/mimmun/article/downloadSuppFile/2616/10345; https://www.mimmun.ru/mimmun/article/downloadSuppFile/2616/10346; https://www.mimmun.ru/mimmun/article/downloadSuppFile/2616/10347; https://www.mimmun.ru/mimmun/article/downloadSuppFile/2616/10348; https://www.mimmun.ru/mimmun/article/downloadSuppFile/2616/10349; https://www.mimmun.ru/mimmun/article/downloadSuppFile/2616/10350; https://www.mimmun.ru/mimmun/article/downloadSuppFile/2616/10351; Горелов А.В., Каннер Е.В. Воспалительные заболевания кишечника у детей: особенности течения и терапии // Медицинский совет, 2018. № 2. C. 140-145.; Организация медицинской помощи пациентам с воспалительными заболеваниями кишечника в РФ в 2015-2021 гг.: информационный материал рабочей группы Экспертного совета по здравоохранению Комитета Совета Федерации по социальной политике //Медицинские технологии. Оценка и выбор, 2021 № 4. С. 9-15.; Петричук C.В., Мирошкина Л.В., Семикина E.Л., Топтыгина А.П., Потапов А.С., Цимбалова E.Г., Радыгина Т.В. Показатели популяционного состава лимфоцитов как предикторы эффективности терапии ингибитором TNFα у детей с воспалительными заболеваниями кишечника // Медицинская иммуноло гия, 2018. T. 20, № 5. С. 721-730. doi:10.15789/1563-0625-2018-5-721-730.; Álvarez-Sánchez N., Cruz-Chamorro I., Díaz-Sánchez M., Lardone P.J., Guerrero J.M., Carrillo-Vico A. Peripheral CD39-expressing T regulatory cells are increased and associated with relapsing-remitting multiple sclerosis in relapsing patients. Sci. Rep., 2019, Vol. 9, 2302. doi:10.1038/s41598-019-38897-w.; Budd S.L., Nicholls D.G. Mitochondria, calcium regulation, and acute glutamate excitotoxicity in cultured cerebellar granule cells. J. Neurochem., 1996, Vol. 67, no. 6, pp. 2282-2291.; Di Virgilio F., Sarti A.C., Silva R.C. Purinergic signaling, DAMPs, and inflammation. Am. J. Physiol. Cell Physiol., 2020, Vol. 318, no. 5, pp. 832-835.; Gibson D.J., Elliott L., McDermott E., Tosetto M., Keegan D., Byrne K., Martin S.T., Rispens T., Cullen G., Mulcahy H.E., Cheifetz A.S., Moss A.C., Robson S.C., Dohert G.A., Ryan E.J. Heightened еxpression of CD39 by regulatory T lymphocytes is associated with therapeutic remission in Inflammatory Bowel Disease. Inflamm. Bowel Dis., 2015, Vol. 21, pp. 2806-2814.; Grant C.R., Liberal R., Holder B.S., Cardone J., Ma Y., Robso S.C., Mieli-Vergani G., Vergani D., Longhi M.S. Dysfunctional CD39(POS) regulatory T cells and aberrant control of T-helper type 17 cells in autoimmune hepatitis. Hepatology (Baltimore, Md.), 2014, Vol. 59, no. 3, pp. 1007-1015.; Guo J., Wang L.-Y., Wu J., Xu L.F., Sun M. The JAK2 inhibitor AG490 regulates the Treg/Th17 balance and alleviates DSS-induced intestinal damage in IBD rats. Clin. Exp. Pharm. Physiol., 2020, Vol. 47, no. 8, pp. 1374-1381.; Moncrieffe H., Nistala K., Kamhieh Y., Evans J., Eddaoudi A., Eaton S., Wedderburn L.R. High expression of the ectonucleotidase CD39 on T cells from the inflamed site identifies two distinct populations, one regulatory and one memory T cell population. J. Immunol., 2010, Vol. 185, no. 1, pp. 134-143.; Negi S., Saini S., Tandel N., Sahu K., Mishr R., Tyagi R.K. Translating Treg therapy for inflammatory bowel disease in humanized mice. Cells. 2021, Vol. 10, no. 8, 1847. doi:10.3390/ cells10081847.; Sorokina E.G., Reutov V.P., Senilova Y.E., Khodorov B.I., Pinelis V.G. Changes in ATP content in cerebellar granule cells during hyperstimulation of glutamate receptors: possible role of NO and nitrite ions. Bull. Exp. Biol. Med., 2007, Vol. 143, no. 4, pp. 442-445.; Vuerich M, Harshe R.P., Robson S.C., Longhi M.S. Dysregulation of adenosinergic signaling in systemic and organ-specific autoimmunity. Int. J. Mol. Sci., 2019, Vol. 20, no. 3, 528. doi:10.3390/ijms20030528.; Yan J.B., Luo M.M., Chen Z.Y., He B.H. The function and role of the Th17/Treg cell balance in Inflammatory bowel disease. J. Immunol. Res., 2020, 8813558. doi:10.1155/2020/8813558.; Zeng J., Ning Z., Wang Y., Xiong H. Implications of CD39 in immune-related diseases. Int. Immunopharmacol., 2020, Vol. 89, Pt A, 107055. doi:10.1016/j.intimp.2020.107055.; https://www.mimmun.ru/mimmun/article/view/2616
-
8Academic Journal
المؤلفون: Olga V. Usoltseva, Andrey N. Surkov, Goar B. Movsisyan, Alexander S. Potapov, Elena A. Kulebina, Кirill A. Kulikov, Vladislav V. Chernikov, О. В. Усольцева, А. Н. Сурков, Г. Б. Мовсисян, А. С. Потапов, Е. А. Кулебина, К. А. Куликов, В. В. Черников
المساهمون: Not specified., Не указан.
المصدر: Current Pediatrics; Том 21, № 2 (2022); 105-112 ; Вопросы современной педиатрии; Том 21, № 2 (2022); 105-112 ; 1682-5535 ; 1682-5527
مصطلحات موضوعية: первичный склерозирующий холангит, liver fibrosis, liver cirrhosis, inflammatory bowel disease, autoimmune hepatitis, primary sclerosing cholangitis, фиброз печени, цирроз печени, воспалительные заболевания кишечника, аутоиммунный гепатит
وصف الملف: application/pdf
Relation: https://vsp.spr-journal.ru/jour/article/view/2920/1170; Carroll MW, Kuenzig ME, Mack DR, et al. The Impact of Inflammatory Bowel Disease in Canada 2018: Children and Adolescents with IBD. J Can Assoc Gastroenterol. 2019;2(Suppl 1): S49–S67. doi: https://doi.org/10.1093/jcag/gwy056; Hyams JS. Extraintestinal manifestations of inflammatory bowel disease in children. J Pediatr Gastroenterol Nutr. 1994;19(1):7–21. doi: https://doi.org/10.1097/00005176-199407000-00003; Яблокова Е.А., Горелов А.В., Ратникова М.А. и др. Первичносклерозирующий холангит при воспалительных заболеваниях кишечника у детей: течение, диагностика и лечение // Вопросы детской диетологии. — 2009. — Т. 7. — № 1. — С. 12–17.; Ricciuto A. Hepatobiliary pathology in paediatric inflammatory bowel disease. [dissertation]. Toronto; 2019.; Cardile S, Alterio T, Candusso M, et al. Autoimmune liver diseases and inflammatory bowel diseases in children: current issues and future perspectives. Scand J Gastroenterol. 2017;52(6-7):662–667. doi: https://doi.org/10.1080/00365521.2017.1298833; Bailey J, Sreepati G, Love J, et al. Autoimmune hepatitis with inflammatory bowel disease is distinct and may be more refractory to traditional treatment. Am J Gastroenterol. 2014;109:S149.; Perdigoto R, Carpenter HA, Czaja AJ. Frequency and significance of chronic ulcerative colitis in severe corticosteroid-treated autoimmune hepatitis. J Hepatol. 1992;14(2-3):325–331. doi: https://doi.org/10.1016/0168-8278(92)90178-r; Ludwig J, Barham SS, LaRusso NF, et al. Morphologic features of chronic hepatitis associated with primary sclerosing cholangitis and chronic ulcerative colitis. Hepatology. 1981;1(6):632–640. doi: https://doi.org/10.1002/hep.1840010612; MacCarty RL, LaRusso NF, May GR, et al. Cholangiocarcinoma complicating primary sclerosing cholangitis: cholangiographic appearances. Radiology. 1985;156(1):43–46. doi: https://doi.org/10.1148/radiology.156.1.2988012; Broomé U, Olsson R, Lööf L, et al. Natural history and prognostic factors in 305 Swedish patients with primary sclerosing cholangitis. Gut. 1996;38(4):610–615. doi: https://doi.org/10.1136/gut.38.4.610; Усольцева О.В., Сурков А.Н., Мовсисян Г.Б. и др. Первичный склерозирующий холангит у детей с воспалительными заболеваниями кишечника // Российский педиатрический журнал. — 2021. — Т. 24. — № 6. — С. 395–404. doi: https://doi.org/10.46563/1560-9561-2021-24-6-395-404; Brunt EM. Grading and staging the histopathological lesions of chronic hepatitis: the Knodell histology activity index and beyond. Hepatology. 2000;31(1):241–246. doi: https://doi.org/10.1002/hep.510310136; Buzzetti E, Hall A, Ekstedt M, et al. Collagen proportionate area is an independent predictor of long term 132 outcome in patients with non-alcoholic fatty liver disease. Aliment Pharmacol Ther. 2019;49(9):1214–1222. doi: https://doi.org/10.1111/apt.15219; Calvaruso V, Di Marco V, Bavetta MG, et al. Quantification of fibrosis by collagen proportionate area predicts hepatic decompensation in hepatitis C cirrhosis. Aliment Pharmacol Ther. 2015;41(5):477–486. doi: https://doi.org/10.1111/apt.13051; Sandrin L, Fourquet B, Hasquenoph J, et al. Transient elastography: a new noninvasive method for assessment of hepatic fibrosis. Ultrasound Med Biol. 2003;29(12):1705–1713. doi: https://doi.org/10.1016/j.ultrasmedbio.2003.07.001; Veidal SS, Karsdal MA, Nawrocki A, et al. Assessment of proteolytic degradation of the basement membrane: a fragment of type IV collagen as a biochemical marker for liver fibrosis. Fibrogenesis Tissue Repair. 2011;4:22. doi: https://doi.org/10.1186/1755-1536-4-22; Кулебина Е.А. Совершенствование неинвазивной диагностики фиброза при хронических болезнях печени у детей: дис. . канд. мед. наук. — М., 2021 — 144 с.; Евлюхина Н. Н. Разработка системы определения структурно-функциональных нарушений печени при ее хронических болезнях у детей: дис. … канд. мед. наук. — М.; 2015. — 132 c.; Cassinotto C, Boursier J, de Lédinghen V, et al. Liver stiffness in nonalcoholic fatty liver disease: A comparison of supersonic shear imaging, FibroScan, and ARFI with liver biopsy. Hepatology. 2016;63(6):1817–1827. doi: https://doi.org/10.1002/hep.28394; Vos MB, Abrams SH, Barlow SE, et al. NASPGHAN Clinical Practice Guideline for the Diagnosis and Treatment of Nonalcoholic Fatty Liver Disease in Children: Recommendations from the Expert Committee on NAFLD (ECON) and the North American Society of Pediatric Gastroenterology, Hepatology and Nutrition (NASPGHAN). J Pediatr Gastroenterol Nutr. 2017;64(2):319–334. doi: https://doi.org/10.1097/MPG.0000000000001482; Rockey DC, Caldwell SH, Goodman ZD, et al. Liver biopsy. Hepatology. 2009;49(3):1017–1044. doi: https://doi.org/10.1002/hep.22742; Mieli-Vergani G, Heller S, Jara P, et al. Autoimmune hepatitis. J Pediatr Gastroenterol Nutr. 2009;49(2):158–164. doi: https://doi.org/10.1097/MPG.0b013e3181a1c265; Lim TS, Kim JK. Is liver biopsy still useful in the era of noninvasive tests? Clin Mol Hepatol. 2020;26(3):302–304. doi: https://doi.org/10.3350/cmh.2020.0081; Abdelaal UM, Morita E, Nouda S, et al. Evaluation of portal hypertensive enteropathy by scoring with capsule endoscopy: is transient elastography of clinical impact? J Clin Biochem Nutr. 2010; 47(1):37–44. doi: https://doi.org/10.3164/jcbn.10-14; Patanwala I, Sweeney E. Transient Elastography may overestimate risk of severe fibrosis in patients with primary sclerosing cholangitis. Gut. 2021;70(Suppl):A150.; Ziol M, Handra-Luca A, Kettaneh A, et al. Noninvasive assessment of liver fibrosis by measurement of stiffness in patients with chronic hepatitis C. Hepatology. 2005;41(1):48–54. doi: https://doi.org/10.1002/hep.20506; Chon YE, Choi EH, Song KJ, et al. Performance of transient elastography for the staging of liver fibrosis in patients with chronic hepatitis B: a meta-analysis. PLoS One. 2012;7(9):e44930. doi: https://doi.org/10.1371/journal.pone.0044930; Heidari G, Motamed F, Heirati B, et al. P. Diagnostic Performance of Noninvasive Methods for Liver Biopsy by Fibroscan in Pediatric. Journal of Child Science. 2021);11(1):e55–e59. doi: https://doi.org/10.1055/s-0041-1725079; Longo-Santos LR, Teodoro WR, de Mello ES, et al. Early type I collagen deposition is associated with prognosis in biliary atresia. J Pediatr Surg. 2016;51(3):379–385. doi: https://doi.org/10.1016/j.jpedsurg.2015.08.061; Tulin-Silver S, Obi C, Kothary N, et al. Comparison of Transjugular Liver Biopsy and Percutaneous Liver Biopsy With Tract Embolization in Pediatric Patients. J Pediatr Gastroenterol Nutr. 2018;67(2): 180–184. doi: https://doi.org/10.1097/MPG.0000000000001951; Deneau MR, Mack C, Abdou R, et al. Gamma Glutamyltransferase Reduction Is Associated With Favorable Outcomes in Pediatric Primary Sclerosing Cholangitis. Hepatol Commun. 2018;2(11):1369–1378. doi: https://doi.org/10.1002/hep4.1251; Fickert P, Hirschfield GM, Denk G, et al. norUrsodeoxycholic acid improves cholestasis in primary sclerosing cholangitis. J Hepatol. 2017;67(3):549–558. doi: https://doi.org/10.1016/j.jhep.2017.05.009; Chandrakumar A, Loeppky R, Deneau M, El-Matary W. Inflammatory Bowel Disease in Children with Elevated Serum Gamma Glutamyltransferase Levels. J Pediatr. 2019;215:144–151.e3. doi: https://doi.org/10.1016/j.jpeds.2019.07.065; Cioffi M. Laboratory markers in ulcerative colitis: Current insights and future advances. World J Gastrointest Pathophysiol. 2015;1(6):13–22. doi: https://doi.org/10.4291/wjgp.v6.i1.13; https://vsp.spr-journal.ru/jour/article/view/2920
-
9Academic Journal
المؤلفون: A. P. Toptygina, E. L. Semikina, S. V. Petrichuk, A. S. Potapov, Andrey N. Surkov, А. П. Топтыгина, Е. Л. Семикина, С. В. Петричук, А. С. Потапов, А. Н. Сурков
المصدر: Medical Immunology (Russia); Том 22, № 4 (2020); 717-728 ; Медицинская иммунология; Том 22, № 4 (2020); 717-728 ; 2313-741X ; 1563-0625
مصطلحات موضوعية: воспалительные заболевания кишечника, antibody, Crohn’s disease, ulcerative colitis, inflammatory bowel disease, антитела, болезнь Крона, язвенный колит
وصف الملف: application/pdf
Relation: https://www.mimmun.ru/mimmun/article/view/1998/1290; https://www.mimmun.ru/mimmun/article/downloadSuppFile/1998/6045; https://www.mimmun.ru/mimmun/article/downloadSuppFile/1998/6046; https://www.mimmun.ru/mimmun/article/downloadSuppFile/1998/6047; https://www.mimmun.ru/mimmun/article/downloadSuppFile/1998/6048; https://www.mimmun.ru/mimmun/article/downloadSuppFile/1998/6049; https://www.mimmun.ru/mimmun/article/downloadSuppFile/1998/6050; https://www.mimmun.ru/mimmun/article/downloadSuppFile/1998/6051; https://www.mimmun.ru/mimmun/article/downloadSuppFile/1998/6052; https://www.mimmun.ru/mimmun/article/downloadSuppFile/1998/6053; https://www.mimmun.ru/mimmun/article/downloadSuppFile/1998/6054; https://www.mimmun.ru/mimmun/article/downloadSuppFile/1998/6055; Климович В.Б. Защитные и гомеостатические функции секреторных иммуноглобулинов (SIG) // Российский иммунологический журнал, 2008. Т. 2 (11), № 2. С. 2-7.; Петричук С.В., Мирошкина Л.В., Семикина Е.Л., Топтыгина А.П., Потапов А.С., Цимбалова Е.Г., Радыгина Т.В. Показатели популяционного состава лимфоцитов как предикторы эффективности терапии ингибитором TNF-α у детей с воспалительными заболеваниями кишечника // Медицинская иммунология, 2018. Т. 20, № 5. С. 721-730.; Топтыгина А.П., Семикина Е.Л., Бобылева Г.В., Мирошкина Л.В., Петричук С.В. Цитокиновый профиль у детей с воспалительными заболеваниями кишечника // Биохимия, 2014. Т. 79, № 12. С. 1673-1679.; Arumugam M., Raes J., Pelletier E., le Paslier D., Yamada T., Mende D.R., et al. Enterotypes of the human gut microbiome. Nature, 2011, Vol. 473, pp. 174-180.; Bogdanos D.P., Rigopoulou E.I., Smyk D.S., Roggenbuck D., Reinhold D., Forbes A., Laass M.W., Conrad K. Diagnostic value, clinical utility and pathogenic significance of reactivity to the molecular targets of Crohn’s disease specific-pancreatic autoantibodies. Autoimmun. Rev., 2011, Vol. 11, pp. 143-148.; Chao K., Zhang S., Yao J., He Y., Chen B., Zeng Z., Zhong B., Chen M. Imbalances of CD4(+) T-cell subgroups in Crohn’s disease and their relationship with disease activity and prognosis. J. Gastroenterol. Hepatol., 2014, Vol. 29, pp. 1808-1814.; Cobrin G.M., Abreu M.T. Defects in mucosal immunity leading to Crohn’s disease. Immunol. Rev, 2005, Vol. 206, pp. 277-295.; de Beéck K.O., Vermeire S., Rutgeerts P., Bossuyt X. Antibodies to GP2, the major zymogen granule membrane glycoprotein, in inflammatory bowel diseases. Gut, 2012, Vol. 61, pp. 162-164.; Fonseca-Camarillo G., Yamamoto-Furusho J.K. Immunoregulatory pathways involved in inflammatory bowel disease. Inflamm. Bowel Dis., 2015, Vol. 21, no. 9, pp. 2188-2193.; Fujino S., Andoh A., Bamba S., Ogawa A., Hata K., Araki Y., Bamba T., Fujiyama Y. Increased expression of interleukin 17 in inflammatory bowel disease. Gut, 2003, Vol. 52, pp. 65-70.; Gaboriau-Routhiau V., Rakotobe S., Lecuyer E., Mulder, I., Lan A., Bridonneau C., Rochet V., Pisi A., de Paepe M., Brandi G., Eberl G., Snel J., Kelly D., Cerf-Bensussan N. The key role of segmented filamentous bacteria in the coordinated maturation of gut helper T cell responses. Immunity, 2009, Vol. 31, no. 4, pp. 677-689.; Gullberg E., Söderholm J.D. Peyer’s patches and M cells as potential sites of the inflammatory onset in Crohn’s disease. Ann. NY Acad. Sci., 2006, Vol. 1072, pp. 218-232.; Hansen R., Russell R.K., Reiff C., Louis P., McIntosh F., Berry S.H., Mukhopadhya I., Bisset W.M., Barclay A.R., Bishop J., Flynn D.M., McGrogan P., Loganathan S., Mahdi G., Flint H.J., El-Omar E.M., Hold G.L. Microbiota of de-novo pediatric IBD: increased Faecalibacterium prausnitzii and reduced bacterial diversity in Crohn’s but not in ulcerative colitis. Am. J. Gastroenterol., 2012, Vol. 107, pp. 1913-1922.; Hase K., Kawano K., Nochi T., Pontes G.S., Fukuda S., Ebisawa M., Kadokura K., Tobe T., Fujimura Y., Kawano S., Yabashi A., Waguri S., Nakato G., Kimura S., Murakami T., Iimura M., Hamura K., Fukuoka S., Lowe A.W., Itoh K., Kiyono H., Ohno H. Uptake through glycoprotein 2 of FimH+ bacteria by M cells initiates mucosal immune response. Nature, 2009, Vol. 462, no. 7270, pp. 226-230.; Hold G.L., Smith M., Grange C., Watt E.R., El-Omar E.M., Mukhopadhya I. Role of the gut microbiota in inflammatory bowel disease pathogenesis: What have we learnt in the past 10 years? World J. Gastroenterol., 2014, Vol. 20, no. 5, pp. 1192-1210.; Lennard-Jones J.E. Classification of inflammatory bowel disease. Scand. J. Gastroenterol., Suppl., 1989, Vol. 170, pp. 2-6.; Liu J.Z., van Sommeren S., Huang H., Ng S.C., Alberts R., Takahashi A., Ripke S., Lee J.C., Jostins L., Shah T., Abedian S., Cheon J.H., Cho J., Dayani N.E., Franke L., Fuyuno Y., Hart A., Juyal R.C., Juyal G., Kim W.H., Morris A.P., Poustchi H., Newman W.G., Midha V., Orchard T.R., Vahedi H., Sood A., Sung J.Y., Malekzadeh R., Westra H.J., Yamazaki K., Yang S.K.; International Multiple Sclerosis Genetics Consortium; International IBD Genetics Consortium, Barrett J.C., Alizadeh B.Z., Parkes M., Bk T., Daly M.J., Kubo M., Anderson C.A., Weersma R.K. Association analyses identify 38 susceptibility loci for inflammatory bowel disease and highlight shared genetic risk across populations. Nat. Genet., 2015, Vol. 47, no. 9, pp. 979-986.; Mazmanian S.K., Round J.L., Kasper D.L. A microbial symbiosis factor prevents intestinal inflammatory disease. Nature, 2008, Vol. 453, pp. 620-625.; McGovern D., Kugathasan S., Cho J.H. Genetics of inflammatory bowel diseases. Gastroenterology, 2015, Vol. 149, no. 5, pp. 1163-1176.; Palm N.W., De Zoete M.R., Cullen T.W., Barry N.A., Stefanowski J., Hao L., Degnan P.H., Hu J., Peter I., Zhang W., Ruggiero E., Cho J.H., Goodman A.L., Flavell R.A. Immunoglobulin A coating identifies colitogenic bacteria in inflammatory bowel disease. Cell, 2014, Vol. 158, pp. 1000-1010.; Pavlidis P., Forbes A., Bogdanos D.P. Antibodies to glycoprotein 2 (GP2) in patients with inflammatory bowel diseases from UK. Clin. Chim. Acta, 2011, Vol. 412, no. 11-12, pp. 1163-1164.; Pavlidis P., Romanidou O., Roggenbuck D., Mytilinaiou M.G., Al-Sulttan F., Liaskos C., Smyk D.S., Koutsoumpas A.L., Rigopoulou E.I., Conrad K., Forbes A., Bogdanos D.P. Ileal inflammation may trigger the development of GP2-specific pancreatic autoantibodies in patients with crohn’s disease. Clin. Dev. Immunol., 2012, Vol. 2012, 640835. doi:10.1155/2012/640835.; Podolsky DK. The current future understanding of inflammatory bowel disease. Best Pract. Res. Clin. Gastroenterol., 2002, Vol. 16, pp. 933-943.; Roggenbuck D., Hausdorf G., Martinez-Gamboa L., Reinhold D., Büttner T., Jungblut P.R., Porstmann T., Laass M.W., Henker J., Büning C., Feist E., Conrad K. Identification of GP2, the major zymogen granule membrane glycoprotein, as the autoantigen of pancreatic antibodies in Crohn’s disease. Gut, 2009, Vol. 58, no. 12, pp. 1620-1628.; Roggenbuck D., Reinhold D., Wex T., Goihl A., von Arnim U., Malfertheiner P., Büttner T., Porstmann T., Porstmann S., Liedvogel B., Bogdanos D.P., Laass M.W., Conrad K. Autoantibodies to GP2, the major zymogen granule membrane glycoprotein, are new markers in Crohn’s disease. Clin. Chim. Acta, 2011, Vol. 412, no. 9-10, pp. 718-724.; Russell R. K., Drummond H. E., Nimmo E. E. Anderson N., Smith L., Wilson D.C., Gillett P.M., McGrogan P., Hassan K., Weaver L.T., Bisset M., Mahdi G., Satsangi J. Genotype-phenotype analysis in childhood-onset Crohn’s disease: NOD2/CARD15 variants consistently predict phenotypic characteristics of severe disease. Inflamm. Bowel Dis., 2005, Vol. 11, pp. 955-964.; Satsangi J., Silverberg M. S., Vermeire S., Colombel J.-F. The Montreal classification of inflammatory bowel disease: controversies, consensus, and implications. Gut, 2006, Vol. 55. no. 6, pp. 749-753.; Sha S., Xu B., Wang X., Zhang Y., Wang H., Kong X., Zhu H., Wu K. The biodiversity and composition of the dominant fecal microbiota in patients with inflammatory bowel disease. Diagn. Microbiol. Infect. Dis., 2013, Vol. 75, pp. 245-251.; Silverberg M.S., Satsangi J., Ahmad T., Arnott I.D., Bernstein C.N., Brant S.R., Caprilli R., Colombel J.F., Gasche C., Geboes K., Jewell D.P., Karban A., Loftus E.V. Jr, Peña A.S., Riddell R.H., Sachar D.B., Schreiber S., Steinhart A.H., Targan S.R., Vermeire S., Warren B.F. Toward an integrated clinical, molecular and serological classification of inflammatory bowel disease: report of a Working Party of the 2005 Montreal World Congress of Gastroenterology. Canad. J. Gastroenterol., 2005, Vol. 19, Suppl. A, pp. 5A-36A.; Werner L., Sturm A., Roggenbuck D., Yahav L., Zion T., Meirowithz E., Ofer A., Guzner-Gur H., Tulchinsky H., Dotan I. Antibodies against glycoprotein 2 are novel markers of intestinal inflammation in patients with an ileal pouch. J. Crohn’s Colitis, 2013, Vol. 7, no. 11, pp. e522-e532.; https://www.mimmun.ru/mimmun/article/view/1998
-
10Academic Journal
المؤلفون: S. V. Petrichuk, L. V. Miroshkina, E. L. Semikina, A. P. Toptygina, A. S. Potapov, E. G. Tsimbalova, T. V. Radygina, C. В. Петричук, Л. В. Мирошкина, E. Л. Семикина, А. П. Топтыгина, А. С. Потапов, E. Г. Цимбалова, Т. В. Радыгина
المصدر: Medical Immunology (Russia); Том 20, № 5 (2018); 721-730 ; Медицинская иммунология; Том 20, № 5 (2018); 721-730 ; 2313-741X ; 1563-0625 ; 10.15789/1563-0625-2018-5
مصطلحات موضوعية: эффективность терапии, популяции CD4+ лимфоцитов, ВЗК, дети, инфликсимаб
وصف الملف: application/pdf
Relation: https://www.mimmun.ru/mimmun/article/view/1640/1064; Белоусова Е.А., Морозова Н.А., Великанов Е.В., Гаганов Л.Е. Инфликсимаб при язвенном колите: обзор исследований и собственные результаты // Фарматека, 2011. № 2. С. 72-77.; Маянская И.В., Маянский А.Н. Неиммунные и иммунные механизмы, участвующие в развитии хронических воспалительных заболеваний кишечника // Вопросы диагностики в педиатрии, 2013. Т. 5, № 4. С. 14-23.; Мирошкина Л.В., Петричук С.В., Цимбалова Е.Г., Потапов А.С., Венедиктова М.М., Топтыгина А.П., Курбатова О.В., Измайлова Т.Д., Семикина Е.Л., Варичкина М.А. Цитокиновый профиль у детей с воспалительными заболеваниями кишечника как предиктор эффективности терапии инфликсимабом // Вопросы диагностики в педиатрии, 2013. Т. 5, № 4. С. 33-38.; Сагынбаева В.Э., Лазебник Л.Б., Князев О.В., Ефремов Л.И. Антитела к инфликсимабу и к антигенам HLA I–II класса как свидетели иммунного ответа на биологическую терапию воспалительных заболеваний кишечника // Экспериментальная и клиническая гастроэнтерология, 2011. № 12. С. 7-14.; Топтыгина А.П., Семикина Е.Л., Петричук С.В., Закиров Р.Ш., Курбатова О.В., Копыльцова Е.А., Комах Ю.А. Изменение уровня субпопуляций Т-регуляторных клеток и Т-хелперов 17 в периферической крови здоровых людей в зависимости от возраста // Медицинская иммунология, 2017. Т. 19, № 4. С. 409-420.doi:10.15789/1563-0625-2017-4-409-420.; Хайдуков С.В., Байдун Л.В., Зурочка А.В., Тотолян А.А. Стандартизованная технология «Исследование субпопуляционного состав лимфоцитов периферической крови с применением проточных цитофлюориметров-анализаторов» // Российский иммунологический журнал, 2014. № 4 (17). С. 974-992.; Abraham C., Cho J.H. Inflammatory bowel disease. N. Engl. J. Med., 2009, Vol. 361, pp. 2066-2078.; Annunziato F., Cosmi L., Santarlasci V., Maggi L., Liotta F., Mazzinghi B., Parente E., Filì L., Ferri S., Frosali F., Giudici F., Romagnani P., Parronchi P., Tonelli F., Maggi E., Romagnani S. Phenotypic and functional features of human Th17 cells. J. Exp. Med., 2007, Vol. 204, no. 8, pp. 1849-1861.; Arnott I.D., Mcneill G., Satsangi J. An analysis of factors influencing short-term and sustained response to infliximab treatment for Crohn’s disease. Aliment. Pharmacol. Ther., 2003, Vol. 17, no. 12, pp. 1451-1457.; Bogaert S., Laukens D., Harald Peeters H., Melis L., Olievier K., Boon N., Verbruggen G., Vandesompele J., Elewaut D., De Vos M. Differential mucosal expression of Th17-related genes between the inflamed colon and ileum of patients with inflammatory bowel disease. BMC Immunology, 2010, Vol. 11, pp. 1-11.; Cseh A., Vasarhelyi B., Molnar K. ,Szalay B., Svec P., Treszl A., Dezsofi A., Lakatos P.L., Arato A., Tulassay T., Veres G. Immune phenotype in children with therapy naïve remitted and relapsed Crohn’s disease.World J. Gastroenterol., 2010, Vol. 16, no. 47, pp. 6001-6009.; Chamouard P., Monneaux F., Richert Z., Voegeli A.C., Lavaux T., Gaub M.P., Baumann R., Oudet P., Muller S. Diminution of сirculating CD4+CD25high T cells in naïve Crohn’s disease. Dig. Dis. Sci., 2009, Vol. 54, pp. 2084-2093.; Danese S. Immune and nonimmune components orchestrate the pathogenesis of inflammatory bowel disease. Am. J. Physiol. Gastrointest. Liver Physiol., 2011, Vol. 300, pp. 716-722.; Elgbratt K., Kurlberg G., Hahn-Zohric M., Hörnquist E.H. Rapid migration of thymic emigrants to the colonic mucosa in ulcerative colitis patients. Clin. Exp. Immunol., 2010, Vol. 162, no. 2, pp. 325-336.; El-Hodhod M.A., Aly R.H., Youssef S.R., Mohamedet S.I. Enhanced blood lymphocytes apoptosis in children with inflammatory bowel disease. ISRN Gastroenterology, Vol. 2013, pp. 415-417.; La Scaleia R., Morrone S., Stoppacciaro A., Scarpino S., Antonelli M., Bianchi E., di Nardo G., Oliva S., Viola F., Cucchiara S., Santoni A., Palmieri G., Uccini S. Peripheral and intestinal CD4+ T cells with a regulatory phenotype in pediatric patients with inflammatory bowel disease. J. Pediatr. Gastroenterol. Nutr., 2010, Vol. 51, no. 5, pp. 563-572.; Lierop P.P., Samsom J.N., Escher J.C., Nieuwenhuis E. Role of the innate immune system in the pathogenesis of inflammatory bowel disease. J. Pediatr. Gastroenterol. Nutr., 2009, Vol. 48, no. 2, pp. 142-151.; Maul J., Loddenkemper C., Mundt P., Berget E., Giese T., Stallmach A., Zeitz M., Duchmann R. Peripheral and intestinal regulatory CD4+ CD25high T cells in inflammatory bowel disease. Gastroenterology, 2005, Vol. 128, pp. 1868-1878.; Mishima Y., Ishihara S., Amano Y., Oshima N., Kadota Ch., Otani A. Alterations of peripheral blood CD5+ B cells in inflammatory bowel disease. Scand J. Gastroenterol, 2009, Vol. 44, no. 2, pp. 172-179.; Monteleone G., Fina D., Caruso R., Pallone F. New mediators of immunity and inflammation in inflammatory bowel disease. Curr. Opin. Gastroenterol., 2006, Vol. 22, no. 4, pp. 361-364.; Ogawa K., Matsumoto T., Esaki M., Torisu T., Iida M. Profiles of circulating cytokines in patients with Crohn’s disease under maintenance therapy with infliximab. J. Crohn’s Colitis, 2012, Vol. 6, no. 5, pp. 529-535.; Orlando A., Colombo E., Kohn A., Biancone L., Rizzello F., Viscido A., Sostegni R., Benazzato L., Castiglione F., Papi C., Meucci G., Riegler G., Mocciaro F., Cassinotti A., Cosintino R., Geremia A., Morselli C., Angelucci E., Lavagna A., Rispo A., Bossa F., Scimeca D., Cottone M., Italian Multicentric Study Group on Infliximab. Infliximab in the treatment of Crohn’s disease: predictors of response in an Italian multicentric open study. Dig. Liver. Dis., 2005, Vol. 37, no. 8, pp. 577-583.; Reikvam D.H., Perminow G., Lyckander L.G., Gran J.M., Brandtzaeg P., Vatn M., Carlsen H.S. Increase of regulatory T cells in ileal mucosa of untreated pediatric Crohn’s disease patients. Scand. J. Gastroenterol., 2011, Vol. 46, pp. 550-560.; Ricciardelli I., Lindley K.J., Londei M., Quaratino S. Anti tumour necrosis-alpha therapy increases the number of FOXP3 regulatory T cells in children affected by Crohn’s disease. Immunology, 2008, Vol. 125, no. 2, pp. 178-183.; Sanchez-Muñoz F., Dominguez-Lopez A., Yamamoto-Furusho J.K. Role of cytokines in inflammatory bowel disease. World J. Gastroenterol., 2008, Vol. 14, no. 27, pp. 4280-4288. 26. Souza H., Fiocchi C. Immunopathogenesis of IBD: current state of the art. Nat. Rev. Gastroenterol. Hepatol., 2016, Vol. 13, pp. 13-27.; Taylor K.D., Plevy S.E., Yang H., Landers C.J., Barry M.J., Rotter J.I. Targan S.R. ANCA pattern and LTA haplotype relationship to clinical responses to anti-TNF antibody treatment in Crohn’s disease. Gastroenterology, 2001,Vol. 120, no. 6, pp. 1347-1355.; Vermeire S., Louis E., Carbonez A., van Assche G., Noman M., Belaiche J., de Vos M., van Gossum A., Pescatore P., Fiasse R., Pelckmans P., Reynaert H., D’Haens G., Rutgeerts P. Demographic and clinical parameters influencing the short-term outcome of anti-tumor necrosis factor (Infliximab) treatment in Crohn’s disease. J. Gastroenterol., 2002, Vol. 97, no. 9, pp. 2357-2363.; Voiglio E., Salle B., Lemaitre D., Cloix P., Bancel B., Cozon G. Activation of T-lymphocytes in Crohn disease and in ulcerative hemorrhagic rectocolitis. Therapeutic implications. Pathol. Biol. (Paris), 1996, Vol. 44, no. 4, pp. 287-292.; Yadav P.K., Chen C., Liu Z. Potential role of NK cells in the pathogenesis of inflammatory bowel disease. J. Biomed. Biotechnol., 2011, Vol. 2011, 348530, 6 p. doi:10.1155/2011/348530.; Zhang Y-Z., Li Y-Y. Inflammatory bowel disease: Pathogenesis. World J. Gastroenterol., 2014, Vol. 20, no. 1, pp. 91-99.; https://www.mimmun.ru/mimmun/article/view/1640
-
11Academic Journal
المؤلفون: M. M. Lokhmatov, T. N. Budkina, V. I. Oldakovsky, E. Y. Dyakonova, A. S. Potapov, М. М. Лохматов, Т. Н. Будкина, В. И. Олдаковский, Е. Ю. Дьяконова, А. С. Потапов
المصدر: Pediatric pharmacology; Том 13, № 6 (2016); 587-591 ; Педиатрическая фармакология; Том 13, № 6 (2016); 587-591 ; 2500-3089 ; 1727-5776
مصطلحات موضوعية: болезнь Крона, video-capsule endoscopy, side optics, Crohn’s disease, видеокапсульная эндоскопия, боковая оптика
وصف الملف: application/pdf
Relation: https://www.pedpharma.ru/jour/article/view/1484/872; Лохматов М.М. Внутрипросветноэндоскопические изменения желудочно-кишечного тракта при болезнях органов пищеварения у детей: Автореф. дис. … докт. мед. наук. — М.; 2008. 44 с. [Lokhmatov M.M. Vnutriprosvetnoendoskopicheskie izmeneniya zheludochno-kishechnogo trakta pri boleznyakh organov pishchevareniya u detei. [dissertation abstract] Moscow; 2008. 44 p. (In Russ).]; Pennazio M, Spada C, Eliakim R, et al. Small-bowel capsule endoscopy and device-assisted enteroscopy for diagnosis and treatment of small-bowel disorders: European Society of Gastrointestinal Endoscopy (ESGE) Clinical Guideline. Endoscopy. 2015;47:352–376. doi:10.1055/s-0034-1391855.; Лохматов М.М. Интестиноскопическая оценка состояния слизистой оболочки при хронических болезнях кишечника у детей: Автореф. дис…канд. мед. наук. — М.; 2004. 24 с. [Lokhmatov MM. Intestinoskopicheskaya otsenka sostoyaniya slizistoi obolochki pri khronicheskikh boleznyakh kishechnika u detei. [dissertation abstract] Moscow; 2004. 24 p. (In Russ).]; Лохматов М.М. Развитие энтероскопии на современном этапе // Педиатрическая фармакология. — 2014. — Т. 11. — № 4 — С. 88–92. [Lokhmatov MM. Current development of enteroscopy. Pediatric pharmacology. 2014;11(4):88–92. (In Russ).] doi:10.15690/pf.v11i4.1070.; Мальков В.А., Кузьмин-Крутецкий М.И., Сенчило С.И. Первый опыт в России применения видеокапсулы с панорамным обзором. / Всероссийская научно-практическая конференция «Актуальные вопросы эндоскопии»; Март 30 – Апрель 1, 2016; Санкт-Петербург. С. 317–318. [Mal’kov VA, Kuz’min-Krutetskii MI, Senchilo SI. Pervyi opyt v Rossii primeneniya videokapsuly s panoramnym obzorom. (Conference proceedigs) Vserossiiskaya nauchno-prakticheskaya konferentsiya «Aktual’nye voprosy endoskopii»; 2016 mar 30 – apr 1; St. Petersburg. p. 317–318. (In Russ).] Доступно по: http://rusendo.ru/ru/. Ссылка активна на 12.12.2016.; Pioche M, Vanbiervliet G, Jacob P, et al. Prospective randomized comparison between axialand lateral-viewing capsule endoscopy systems in patients with obscure digestive bleeding. Endoscopy. 2014;46(6):479–484. doi:10.1055/s-0033-1358832.; Friedrich K, Gehrke S, Stremmel W, Sieg A. First clinical trial of a newly developed capsule endoscope with panoramic side view for small bowel: a pilot study. J Gastroenterol Hepatol. 2013;28(9):1496–1501. doi:10.1111/jgh.12280.; https://www.pedpharma.ru/jour/article/view/1484
-
12Academic Journal
المؤلفون: A. O. Anushenko, A. S. Potapov, Ye. G. Tsimbalova, O. B. Gordeeva, А. О. Анушенко, А. С. Потапов, Е. Г. Цимбалова, О. Б. Гордеева
المصدر: Current Pediatrics; Том 15, № 2 (2016); 128-140 ; Вопросы современной педиатрии; Том 15, № 2 (2016); 128-140 ; 1682-5535 ; 1682-5527
مصطلحات موضوعية: лечение, anemia, inflammatory bowel diseases, iron deficiency anemia, anemia of chronic diseases, treatment, анемия, воспалительные заболевания кишечника, железодефицитная анемия, анемия хронических заболеваний
وصف الملف: application/pdf
Relation: https://vsp.spr-journal.ru/jour/article/view/1603/587; Fernandes A, Bacalhau S, Cabral J. Pediatric inflammatory bowel disease: is it still increasing? Acta Med Port. 2011; 24 Suppl 2:333–338.; Benchimol E, Guttmann A, Griffiths A, et al. Increasing incidence of paediatric inflammatory bowel disease in Ontario, Canada: evidence from health administrative data. Gut. 2009;58(11): 1490–1497. doi:10.1136/gut.2009.188383.; Hammer T, Nielsen KR, Burisch J, et al. DOP010. Incidence of inflam matory bowel diseases in the Faroe Islands from 1960–2014: A 54-year overview from a population-based cohort. J Crohns Colitis. 2015;9 (suppl 1):S24. doi:10.1093/ecco-jcc/jju027.038.; Nielsen KR, Hammer T, Burisch J, et al. Su1070 Incidence of inflammatory bowel diseases in the Faroe Islands from 1960–2014: A 54-year overview from a population-based cohort. Gastroenterology. 2015;148(4):S399. doi:10.1016/S0016- 5085(15)31342-1.; Ravikumara M, Sandhu B. Epidemiology of inflammatory bowel diseases in childhood. Indian J Pediatr. 2006;73(8):717–721. doi:10.1007/bf02898451.; Burisch J, Munkholm P. The epidemiology of inflammatory bowel disease. Scand J Gastroenterol. 2015;50(8):942–951. doi:10.3109/00365521.2015.1014407.; Jose F, Garnett E, Vittinghoff E, et al. Development of Extraintestinal Manifestations in Pediatric Patients with Inflammatory Bowel Disease. Inflamm Bowel Dis. 2009;15(1):63–68. doi:10.1002/ibd.20604.; Mamula P, Markowitz J, Baldassano R. Pediatric Inflammatory Bowel Disease. New York, Springer; 2008. Vol. 8. p. 88.; Диагностика и лечение железодефицитной анемии у детей / Под ред. А.Г. Румянцева, Н.А. Коровиной. — М.; 2004. 48 с. [Diagnostika i lechenie zhelezodefitsitnoi anemii u detei. Ed by Rumyantsev A.G., Korovina N.A. Moscow; 2004. 48 p. (In Russ).]; Тарасова И.С. Разработка и научное обоснование скрининга железодефицитных состояний у подростков: Автореф. дис. … докт. мед. наук. — М.; 2013. 67 с. [Tarasova IS. Razrabotka i nauchnoe obosnovanie skrininga zhelezodefitsitnykh sostoyanii u podrostkov. [dissertation abstract] Moscow; 2013. 67 p. (In Russ).]; World Health Organization. Anaemia [Internet]. Geneva, Switzerland: WHO; 2011 [cited 2016 Apr 19]. Available from: http://www.who.int/topics/anaemia/en/.; Gasche C. Anemia in IBD: the overlooked villain. Inflamm Bowel Dis. 2000;6(2):142–150. doi:10.1002/ibd.3780060214.; Cucino C, Sonnenberg A. Cause of death in patients with inflammatory bowel disease. Inflamm Bowel Dis. 2001;7(3): 250–255. doi:10.1097/00054725-200108000-00011.; Gomollon F, Gisbert J. Anemia and inflammatory bowel diseases. World J Gastroenterol. 2009;15(37):4659–4665. doi:10.3748/wjg.15.4659.; Долгов В.В., Луговская С.А., Почтарь М.Е., Федорова М.М. Лабораторная диагностика нарушений обмена железа. — СПб.: Витал Диагностикс; 2002. 52 с. [Dolgov VV, Lugovskaya SA, Pochtar’ ME, Fedorova MM. Laboratornaya diagnostika narushenii obmena zheleza. St. Petersburg: Vital Diagnostiks; 2002. 52 p. (In Russ).]; Kugathasan S, Judd R, Hoffmann R, et al. Epidemiologic and clinical characteristics of children with newly diagnosed inflammatory bowel disease in Wisconsin: a state wide populationbased study. J Pediatr. 2003;143(4):525–531. doi:10.1067/s0022-3476(03)00444-x.; Thomas C, Thomas L. Biochemical markers and hematologic indices in the diagnosis of functional iron deficiency. Clinical Chem. 2002;48(7):1066–1076.; Cartwright GE, Lauritsen MA, Jones PJ, et al. The anemia of infection. I. Hypoferremia, hypercupremia, and alterations in porphyrin metabolism in patients. J Clin Invest. 1946;25(1):65–80. doi:10.1172/JCI101690.; Weiss G, Gasche C. Pathogenesis and treatment of anemia in inflammatory bowel disease. Haematologica. 2010;95(2): 175–178. doi:10.3324/haematol.2009.017046.; Bergamaschi G, Di Sabatino A, Albertini R, et al. Prevalence and pathogenesis of anemia in inflammatory bowel disease. Influence of anti-tumor necrosis factor-alpha treatment. Haematologica. 2010;95(2):199–205. doi:10.3324/haematol.2009.009985.; Weiss G, Goodnough L. Anemia of chronic disease. N Engl J Med. 2005;352(10):1011–1023. doi:10.1056/nejmra041809.; Grimm MС. Inflammatory bowel disease: genes, germs and the gastrointestinal immune system. Today’s Life Science. 2001;13(1):24–30.; Wilson A, Reyes E, Ofman J. Prevalence and outcomes of anemia in inflammatory bowel disease: a systematic review of the literature. Am J Med. 2004;116(7Suppl.1):44–49. doi:10.1016/j.amjmed.2003.12.011.; Ruiz-Arguelles G. Altitude above sea level as a variable for definition of anemia. Blood. 2006;108(6):2131–2132. doi:10.1182/blood-2006-04-016352.; Voegtlin M, Vavricka S, Schoepfer A, et al. Prevalence of anaemia in inflammatory bowel disease in Switzerland: a cross sectional study in patients from private practices and university hospitals. J Crohns Colitis. 2010;4(6):642–648. doi:10.1016/ j.crohns.2010.07.008.; Goodhand J, Kamperidis N, Rao A, et al. Prevalence and management of anemia in children, adolescents, and adults with inflammatory bowel disease. Inflamm Bowel Dis. 2012;18(3): 513–519. doi:10.1002/ibd.21740.; naspghan.org [Internet]. Monitoring disease activity in pediatric IBD patients. A case-based monograph focusing on pediatric IBD [updated 2009 Dec 15; cited 2016 Mar 12]. Available from: http://www.naspghan.org/files/documents/pdfs/cme/podcasts/MonitoringDiseaseActivity_PediatricIBDPatients.pdf.; Beard J. One person’s view of iron deficiency, development, and cognitive function. Am J Clin Nutr. 1995;62(4):709–710.; Kretchmer N, Beard JL, Carlson S. The role of nutrition in the development of normal cognition. Am J Clin Nutr. 1996; 63(6):997–1001.; Lozoff B, Beard J, Connor J, et al. Long-lasting neural and behavioral effects of iron deficiency in infancy. Nutr Rev. 2006; 64(5):34–43. doi:10.1301/nr.2006.may.s34-s43.; Bridges K, Pearson H. Anemias and other red cell disorders. USA: McGraw-Hill; 2008. 97 p.; Skikne B, Punnonen K, Caldron P, et al. Improved differential diagnosis of anemia of chronic disease and iron deficiency anemia: A prospective multicenter evaluation of soluble transferrin receptor and the sTfR/log ferritin index. Am J Hematol. 2011;86(11): 923–927. doi:10.1002/ajh.22108.; Dignass A, Gasche C, Bettenworth D, et al. European consensus on the diagnosis and management of iron deficiency and anaemia in inflammatory bowel diseases. J Crohns Colitis. 2015;9(3):211–222. doi:10.1093/ecco-jcc/jju009.; Wallach J. Interpretation of diagnostic test. New York; 2008. 1200 р.; Brailsford S, Lunec J, Winyard P, et al. A possible role for ferritin during inflammation. Free Radic Res Commun. 1985;1(2): 101–109. doi:10.3109/10715768509056542.; Gasche C, Berstad A, Befrits R, et al. Guidelines on the diagnosis and management of iron deficiency and anemia in inflammatory bowel diseases. Inflamm Bowel Dis. 2007;13(12): 1545–1553. doi:10.1002/ibd.20285.; Cronin C, Shanahan F. Anemia in patients with chronic inflammatory bowel disease. Am J Gastroenterol. 2001;96(8): 2296–2298. doi:10.1111/j.1572-0241.2001.04032.x.; Headstrom P, Rulyak S, Lee S. Prevalence of and risk factors for vitamin B(12) deficiency in patients with Crohn’s disease. Inflamm Bowel Dis. 2008;14(2):217–223. doi:10.1002/ibd.20282.; Alkhouri R, Hashmi H, Baker R, et al. Vitamin and mineral status in patients with inflammatory bowel disease. J Pediatr Gastroenterol Nutr. 2013;56(1):89–92. doi:10.1097/MPG.0b013e 31826a105d.; Goldberg N. Iron deficiency anemia in patients with inflammatory bowel disease. Clin Exp Gastroenterol. 2013; 6: 61-70. doi:10.2147/CEG.S43493.; Suominen P, Punnonen K, Rajamaki A, Irjala K. Serum transferrin receptor and transferrin receptor-ferritin index identify healthy subjects with subclinical iron deficits. Blood. 1998; 92(8):2934-2939.; Ботвиньева В.В., Гордеева О.Б., Потапов А.С., и др. Оценка показателей воспалительного ответа и состояния периферического гемопоэза при неспецифическом язвенном колите у детей // Педиатрическая фармакология. — 2013. — Т. 10. — №5. — С. 52–55. [Botvin'eva VV, Gordeeva OB, Potapov AS, et al. Assessment of inflammatory response indicators and peripheral hemopoiesis at non-specific ulcerative colitis in children. Pediatricheskaya farmakologiya. 2013;10(5):52–55. (In Russ).] doi:10.15690/pf.v10i5.823.; Баркова Э.Н. Патофизиологическое обоснование хронодиагностики и хронотерапии дефицита железа / Сб. научных статей: Патогенез и фармакокоррекция экстремальных и терминальных состояний. — М.; 1995. С. 11–14. [Barkova EN. Patofiziologicheskoe obosnovanie khronodiagnostiki i khronoterapii defitsita zheleza. In: Patogenez i farmakokorrektsiya ekstremal’nykh i terminal’nykh sostoyanii. Moscow; 1995. p. 11–14. (In Russ).]; Stein J, Dignass A. Management of iron deficiency anemia in inflammatory bowel disease — a practical approach. Ann Gastroenterol. 2013;26(2):104–113.; Baynes R. Assessment of iron status. Clin Biochem. 1996; 29(3):209–215. doi:10.1016/0009-9120(96)00010-k.; Thorstensen K, Romslo I. The transferrin receptor: Its diagnostic value and its potential as therapeutic target. Scand J Clin Lab Invest. 1993;53(suppl215):113–120. doi:10.3109/00365519309090703.; Baillie F, Morrison A, Fergus I. Soluble transferrin receptor: A discriminating assay for iron deficiency. Clin Lab Haematol. 2003;25(6):353–357. doi:10.1046/j.0141-9854.2003.00548.x.; Flowers CH, Skikne BS, Covell AM, Cook JD. The clinical measurement of serum transferrin receptor. J Lab Clin Med. 1989;114(4):368–377.; Dudkowiak R, Neubauer K, Poniewierka E. Hepcidin and its role in inflammatory bowel disease. Adv Clin Exp Med. 2013; 22(4):585–591.; Kaitha S, Bashir M, Ali T. Iron deficiency anemia in inflammatory bowel disease. World J Gastrointest Pathophysiol. 2015;6(3): 62–72. doi:10.4291/wjgp.v6.i3.62.; Sanad M, Gharib A. Urinary hepcidin level as an early predictor of iron deficiency in children: A case control study. Ital J Pediatr. 2011;37:37. doi:10.1186/1824-7288-37-37.; Konz T, Montes-Bayon M, Vaulont S. Hepcidin quantification: methods and utility in diagnosis. Metallomics. 2014;6(9): 1583–1590. doi:10.1039/c4mt00063c.; Arnold J, Sangwaiya A, Manglam V, et al. Presence of hepcidin-25 in biological fluids: Bile, ascitic and pleural fluids. World J Gastroenterol. 2010;16(17):2129–2133. doi:10.3748/wjg.v16.i17.2129.; Arnold J, Sangwaiya A, Bhatkal B, et al. Hepcidin and inflammatory bowel disease: dual role in host defence and iron homoeostasis. Eur J Gastroenterol Hepatol. 2009;21(4):425–429. doi:10.1097/MEG.0b013e32830e2885.; Pakoz Z, Cekic C, Arabul M, et al. Evaluation of the Correlation between Hepcidin Serum Levels and Disease Activity in Inflammatory Bowel Disease. Gastroenterol Res Pract. 2015;810942. doi:10.1155/2015/810942.; Nagy J, Lakner L, Poor VS, et al. Serum prohepcidin levels in chronic inflammatory bowel diseases. J Crohns Colitis. 2010; 4(6):649–653. doi:10.1016/j.crohns.2010.07.010.; Павлов А.Д. Биологическая и клиническая роль эритропоэтина и его рецептора в негемопоэтических тканях // Вопросы гематологии, онкологии и иммунопатологии в педиатрии. — 2004. — Т.3. — №1. — С. 12–20. [Pavlov AD. Biological and clinical role of erythropoietin and its receptor in non-hematopoietic tissues. Voprosy gematologii, onkologii i immunopatologii v pediatrii. 2004;3(1):12–20. (In Russ).]; Павлов А.Д., Морщакова Е.Ф. Синдром неадекватной продукции эритропоэтина на анемию // Гематология и трансфузиология. — 1999. — Т. 44. — №3. — С. 30–32. [Pavlov AD, Morshchakova EF. Sindrom neadekvatnoi produktsii eritropoetina na anemiyu. Gematol Transfuziol. 1999;44(3):30–32. (In Russ).]; Katsanos KH, Tatsioni A, Natsi D, et al. Recombinant human erythropoietin in patients with inflammatory bowel disease and refractory anemia: a 15-year single center experience. J Crohns Colitis. 2012;6(1):56–61. doi:10.1016/j.crohns.2011.07.004.; Basseri R, Nemeth E, Vassilaki M, et al. Hepcidin is a key mediator of anemia of inflammation in Crohn’s disease. J Crohns Colitis. 2013;7(8):286–291. doi:10.1016/j.crohns.2012.10.013.; Wine E, Mack D, Hyams J, et al. Interleukin-6 is associated with steroid resistance and reflects disease activity in severe pediatric ulcerative colitis. J Crohns Colitis. 2013;7(11):916–922. doi:10.1016/j.crohns.2012.12.012.; Ito H, Takazoe M, Fukuda Y, et al. A pilot randomized trial of a human anti-interleukin-6 receptor monoclonal antibody in active Crohn’s disease. Gastroenterology. 2004;126(4):989–996. doi:10.1053/j.gastro.2004.01.012.; Bayraktar UD, Bayraktar S. Treatment of iron deficiency anemia associated with gastrointestinal tract diseases. World J Gastroenterol. 2010;16(22):2720–2725. doi:10.3748/wjg.v16. i22.2720.; Moreno Chulilla JA, Romero Colas MS, Gutierrez Martin M. Classification of anemia for gastroenterologists. World J Gastroenterol. 2009;15(37):4627–4637. doi:10.3748/wjg.15.4627.; Fishbane S, Shapiro W, Dutka P, et al. A randomized trial of iron deficiency testing strategies in hemodialysis patients. Kidney Int. 2001;60(6):2406–2411. doi:10.1046/j.1523-1755. 2001.00077.x.; d’Onofrio G, Chirillo R, Zini G, et al. Simultaneous measurement of reticulocyte and red blood cell indexes in healthy subjects and patients with microcytic and macrocytic anemia. Blood. 1995;85(3):818–823.; Piva E, Brugnara C, Spolaore F, Plebani M. Clinical utility of reticulocyte parameters. Clin Lab Med. 2015;35(1):133–163. doi:10.1016/j.cll.2014.10.004.; Rodak B. Hematology: clinical principles and applications. 3 ed. Philadelphia: Saunders; 2007. 220 р.; Black M. Micronutrient deficiencies and cognitive functioning. J Nutr. 2003;133(11 Suppl 2):3927–3931.; de Andraca Oyarzun I, Salas Aliaga I, de la Parra Cieciwa A, Gonzalez Lopez B. Mother–child interaction and child behavior in preschool children with a history of iron-deficiency anemia in infancy. Arch Latinoam Nutr. 1993;43(3):191–198.; Olson C, Acosta L, Hochberg N, et al. Anemia of inflammation is related to cognitive impairment among children in Leyte, the Philippines. PLoS Neglect Trop D. 2009;3(10):533. doi:10.1371/ journal.pntd.0000533.; Gonzalez H, Malpeli A, Etchegoyen G. Acquisition of visuomotor abilities and intellectual quotient in children aged 4–10 years: relationship with micronutrient nutritional status. Biol Trace Elem Res. 2007;120(1-3):92–101. doi:10.1007/s12011-007-8023-5.; Jelkmann W. Proinflammatory cytokines lowering erythropoietin production. J Interferon Cytokine Res. 1998;18(8):555–559. doi:10.1089/jir.1998.18.555.; Nemeth E, Tuttle M, Powelson J, et al. Hepcidin regulates cellular iron efflux by binding to ferroportin and inducing its internalization. Science. 2004;306(5704):2090–2093. doi:10.1126/science.1104742.; Gisbert J, Gomollon F. Common misconceptions in the diagnosis and management of anemia in inflammatory bowel disease. Am J Gastroenterol. 2008;103(5):1299–1307. doi:10.1111/j.1572-0241.2008.01846.x.; Cable R, Carlson B, Chambers L, et al. Practice guidelines for blood transfusion: a compilation from recent peer reviewed literature [Internet]. New York; 2002 [cited 2016 Jan 27]. Available from: http://chapters.redcross.org/br/indianaoh/hospitals/transfusionguidelines.htm.re.; Davies P, Robertson S, Hedge S, et al. Calculating the required transfusion volume in children. Transfusion. 2007;47(2):212–216. doi:10.1111/j.1537-2995.2007.01091.x.; Elzik M, Dirschl D, Dahners L. Correlation of transfusion volume to change in hematocrit. Am J Hematol. 2006;81(2):145–146. doi:10.1002/ajh.20517.; Joint Working Party of the Transfusion and Clinical Haematology Task Forces of the British Committee for Standards in Haematology. Guidelines for the clinical use of red cell transfusions. Br J Haematol. 2001;113(1):24–31. doi:10.1046/j.1365-2257.1998.00151.x.; Crosby E, Ferguson D, Hume H, et al. Guidelines for red blood cell and plasma transfusion for adults and children. Can Med Assoc J. 1997;156 Suppl 11:1–24.; Национальный стандарт РФ. Кровь донорская и ее компоненты. Руководство по применению компонентов донорской крови. — М.; 2010. [Natsional’nyi standart RF. Krov’ donorskaya i ee komponenty. Rukovodstvo po primeneniyu komponentov donorskoi krovi. Moscow; 2010. (In Russ).]; Румянцев А.Г., Аграненко В.А. Гемотрансфузионная терапия в педиатрии и неонатологии. — М.: Макс Пресс; 2002. 643 с. [Rumyantsev AG, Agranenko VA. Gemotransfuzionnaya terapiya v pediatrii i neonatologii. Moscow: Maks Press; 2002. 643 p. (In Russ).]; Oldenburg B, Koningsberger J, Van Berge Henegouwen G, et al. Iron and inflammatory bowel disease. Aliment Pharmacol Ther. 2001;15(4):429–438. doi:10.1046/j.1365- 2036.2001.00930.x.; Gasche C, Lomer MC, Cavill I, Weiss G. Iron, anaemia, and inflammatory bowel diseases. Gut. 2004;53(8):1190–1197. doi:10.1136/gut.2003.035758.; Hayat A. Safety issues with intravenous iron products in the management of anemia in chronic kidney disease. Clin Med Res. 2008;6(3–4):93–102. doi:10.3121/cmr.2008.811.; de Silva A, Mylonaki M, Rampton D. Oral iron therapy in inflammatory bowel disease: usage, tolerance, and efficacy. Inflamm Bowel Dis. 2003;9(5):316–320. doi:10.1097/00054725-200309000-00005.; Авдеев В.Г., Моисеев С.В. Анемия при болезни Крона // Клиническая фармакология и терапия. — 2013. — Т. 22. — №1. — С. 1–6. [Avdeev VG, Moiseev SV. Anemiya pri bolezni Krona. Klinicheskaya farmakologiya i terapiya. 2013;22(1):1–6. (In Russ).]; Toblli J, Cao G, Olivieri L, et al. Comparison of the renal, cardiovascular and hepatic toxicity data of original intravenous iron compounds. Nephrol Dial Transplant. 2010;25(11):3631–3140. doi:10.1093/ndt/gfq260.; Милованов Ю.С., Милованова Л.Ю., Добросмыслов И.А. Железа (III) гидроксид сахарозный комплекс (Венофер) или его копии в лечении анемии у больных хронической болезнью почек: на что ориентироваться при выборе препарата? // Клиническая нефрология. — 2012.— №1. — С. 49–59. [Milovanov YuS, Milovanova LYu, Dobrosmyslov IA. Iron sucrose in treatment of anemia in chronic kidney disease — original and generic drugs: what are the guidelines in drug choose? Klinicheskaya nefrologiya. 2012;(1):49–59. (In Russ).]; Beshara S, Sorensen J, Lubberink M. Pharmacokinetics and red cell utilization of 52Fe/59Fe-labelled iron polymaltose in anaemic patients using positron emission tomography. Br J Haematol. 2003;120(5):853–859. doi:10.1046/j.1365- 2141.2003.03590.x.; Moore R, Gaskell H, Rose H, Allan J. Meta-analysis of efficacy and safety of intravenous ferric carboxymaltose (Ferinject) from clinical trial reports and published trial data. BMC Blood Disorders. 2011;11:4. doi:10.1186/1471-2326-11-4.; Kulnigg S, Stoinov S, Simanenkov V. A novel intravenous iron formulation for treatment of anemia in inflammatory bowel disease: the ferric carboxymaltose (FERINJECT®) randomized controlled trial. Am J Gastroenterol. 2007;103(5):1182–1192. doi:10.1111/j.1572-0241.2007.01744.x.; Hachemi S, Mutalib M, Chadokufa S, et al. Assessment of safety and efficacy of ferric carboxymaltose (Ferinject®) in the management of iron deficiency with anaemia (IDA) or without anaemia in children and adolescents with inflammatory bowel disease (IBD). Abstracts of the 10th Congress of ECCO — European Crohn’s and Colitis Organisation. New York; 2015. p. 266.; Laass MW, Straub S, Chainey S, et al. Effectiveness and safety of ferric carboxymaltose treatment in children and adolescents with inflammatory bowel disease and other gastrointestinal diseases. BMC Gastroenterol. 2014;14:184. doi:10.1186/1471- 230x-14-184.; Cella D, Dobrez D, Glaspy J. Control of cancer-related anemia with erythropoietic agents: a review of evidence for improved quality of life and clinical outcomes. Ann Oncol. 2003;14(4):511–519. doi:10.1093/annonc/mdg167.; Littlewood T, Bajetta E, Nortier J, et al. Effects of epoetin alfa on hematologic parameters and quality of life in cancer patients receiving nonplatinum chemotherapy: results of a randomized, double blind, placebo controlled trial. J Clin Oncol. 2001; 19(11):2865–2874.; Kaltwasser J, Kessler U, Gottschalk R, et al. Effect of recombinant human erythropoietin and intravenous iron on anemia and disease activity in rheumatoid arthritis. J Rheumatol. 2001; 28(11):2430–2436.; Yasuda Y, Fujita Y, Matsuo T, et al. Erythropoietin regulates tumour growth of human malignancies. Carcinogenesis. 2003; 24(9):1021–1029. doi:10.1093/carcin/bgg126.; Arcasoy M, Amin K, Karayal A, et al. Functional significance of erythropoietin receptor expression in breast cancer. Lab Invest. 2002;82(7):911–918. doi:10.1097/01.lab.00000 20415.72863.40.; Румянцев А.Г., Морщакова Е.Ф., Павлов А.Д. Эритропоэтин: диагностика, профилактика и лечение анемий. — М.; 2003. 448 с. [Rumyantsev AG, Morshchakova EF, Pavlov AD. Eritropoetin: diagnostika, profilaktika i lechenie anemii. Moscow; 2003. 448 p. (In Russ).]; Theurl I, Aigner E, Theurl M, et al. Regulation of iron homeostasis in anemia of chronic disease and iron deficiency anemia: diagnostic and therapeutic implications. Blood. 2009;113(21): 5277–5286. doi:10.1182/blood-2008-12-195651.; https://vsp.spr-journal.ru/jour/article/view/1603
-
13Academic Journal
المؤلفون: Е. А. Roslavtseva, Т. E. Borovik, Е. G. Tsimbalova, А. О. Anushenko, А. S. Potapov, М. М. Lokhmatov, I. L. Chashchina, М. D. Bakradze, Е. А. Рославцева, Т. Э. Боровик, Е. Г. Цимбалова, А. О. Анушенко, А. С. Потапов, М. М. Лохматов, И. Л. Чащина, М. Д. Бакрадзе
المصدر: Current Pediatrics; Том 14, № 3 (2015); 416-421 ; Вопросы современной педиатрии; Том 14, № 3 (2015); 416-421 ; 1682-5535 ; 1682-5527
مصطلحات موضوعية: синдромная (фенотипическая) диарея, intestinal failure, parenteral, enteral nutrition, tricho-hepato-enteric syndrome, syndromic (phenotypic) diarrhea, кишечная недостаточность, парентеральное, энтеральное питание, трихогепатоэнтеральный синдром
وصف الملف: application/pdf
Relation: https://vsp.spr-journal.ru/jour/article/view/709/528; Berni Canani R., Terrin G., Cardillo G., Tomaiuolo R., Costaldo G. Congenital diarrheal disorders: improved understanding of gene defects is leading to advances in intestinal physiology and clinical management. J. Pediatr. Gastroenterol. Nutr. 2010; 50: 360–366.; Stankler L., Lloyd D., Pollitt R. J., Gray E. S., Thom H., Russell G. Unexplained diarrhoea and failure to thrive in 2 siblings with unusual faces and abnormal scalp hair shafts: a new syndrome. Arch. Dis. Child. 1982; 57: 212–216.; Verloes A., Lombet J., Lambert Y., Hubert A. F., Deprez M., Fridman V., Gosseye S., Rigo J., Sokal E. Tricho-hepato-enteric syndrome: further delineation of a distinct syndrome with neonatal hemochromatosis phenotype, intractable diarrhea, and hair anomalies. Am. J. Med. Genet. 1997; 68: 391–395.; Girault D., Goulet O., Le Deist F., Brousse N., Colomb V., Cesarini J. P., de Potter S., Canioni D., Griscelli C., Fischer A. Intractable infant diarrhea associated with phenotypic abnormalities and immunodeficiency. J. Pediatr. 1994; 125: 36–42.; Fabre A., Andre N., Breton A., Broue P., Badens C., Roquelaure B. Intractable diarrhea with phenotypic anomalies and tricho-hepatoenteric syndrome: two names for the same disorder. Am. J. Med. Genet. A. 2007; 143: 584–588.; Al Qoaer K., Al Mehaidib A., Shabib S., Banemai M. Chronic diarrhea and skin hyperpigmentation: a new association. Saudi J. Gastroenterol. 2008; 14: 187–191.; Hartley J. L., Zachos N. C., Dawood B., Donowitz M., Forman J., Pollitt R.J., Morgan N.V., Tee L., Gissen P., Kahr W.H., Knisely A.S., Watson S., Chitayat D., Booth I. W., Protheroe S., Murphy S., de Vries E., Kelly D. A., Maher E. R. Mutations in TTC37 cause trichohepatoenteric syndrome (phenotypic diarrhea of infancy). Gastroenterology. 2010; 138: 2388–2398.; Fabre A., Martinez-Vinson C., Roquelaure B., Missirian C., Andre N., Breton A., Lachaux A., Odul E., Colomb V., Lemale J., Cezard J. P., Goulet O., Sarles J., Levy N., Badens C. Novel mutations in TTC37 associated with tricho-hepato-enteric syndrome. Hum. Mutat. 2011; 32: 277–281.; Fabre A., Charroux B., Martinez-Vinson C., Roquelaure B., Odul E., Sayar E., Smith H., Colomb V., Andre N., Hugot J. P., Goulet O., Lacoste C., Sarles J., Royet J., Levy N., Badens C. SKIV2L mutations cause syndromic diarrhea, or trichohepatoenteric syndrome. Am. J. Hum. Genet. 2012; 90: 689–692.; Fabre A., Martinez-Vinson C., Goulet O., Badens C. Syndromic diarrhea/Tricho-hepatoenteric syndrome. Orphanet J. Rare Dis. 2013; 8: 5.; Fabre A., Breton A., Coste M. E., Colomb V., Dubern B., Lachaux A., Lemale J., Mancini J., Marinier E., Martinez-Vinson C., Peretti N., Perry A., Roquelaure B., Venaille A., Sarles J., Goulet O., Badens C. Syndromic (phenotypic) diarrhoea of infancy/trichohepato-enteric syndrome. Arch. Dis Child. 2014; 99: 35–38.; Kotecha U. H., Movva S., Puri R. D., Verma I. C. Trichohepatoenteric syndrome: founder mutation in asian indians. Mol. Syndromol. 2012; 3 (2): 89–93. Doi:10.1159/000339896.; Monies D. M., Rahbeeni Z., Abouelhoda M., Naim E. A., AlYounes B., Al-Mahadib A. Expanding phenotypic and allelic heterogeneity of Tricho-Hepato-Enteric Syndrome (THES). J. Pediatr. Gastroenterol. Nutr. 2015; 60 (3): 352–356. Doi:10.1097/ MPG.0000000000000627.; Goulet O., Vinson C., Roquelaure B., Brousse N., Bodemer C., CezardJ.P. Syndromic (phenotypic) diarrhea in early infancy. Orphanet J. Rare Dis. 2008; 3: 6.; Egritas O., Dalgic B., Onder M. Tricho-hepatoenteral syndrome presenting with mild colitis. Eur. J. Pediatr. 2008; 12, 168 (8): 993–995. 16. Dweikat I., Sultan M., Maraqa N., Hindi T., Abu-Rmeileh S., Abu-Libdeh B. Clinical report. Tricho-hepato-enteric syndrome: a case of hemochromatosis with intractable diarrhea, dismorphic features, and hair abnormality. Am. J. Med. Genet. 2007; Part A, 143 (6): 581–583.; https://vsp.spr-journal.ru/jour/article/view/709
-
14Academic Journal
المؤلفون: T. A. Skvortsova, G. V. Volynets, A. S. Poyapov, E. L. Semikina, Т. А. Скворцова, Г. В. Волынец, А. С. Потапов, Е. Л. Семикина
المصدر: Current Pediatrics; Том 12, № 3 (2013); 70-73 ; Вопросы современной педиатрии; Том 12, № 3 (2013); 70-73 ; 1682-5535 ; 1682-5527
مصطلحات موضوعية: противовирусная терапия, morbidity of children population, interferon therapy, antiviral therapy, pegylated interferon α, ribavirin, recombinant interferon α, recombinant interleukin 2, заболеваемость детского населения, интерферонотерапия
وصف الملف: application/pdf
Relation: https://vsp.spr-journal.ru/jour/article/view/333/236; Delgado-Borrego A. Treating hepatitis C: are children the same as adults? Gastroenterology. 2011; 140: 389–392.; Baranov A. A., Namazova-Baranova L. S., Volynets G. V., Potapov A. S., Il'in A. G., Konova S. R., Skvortsova T. A., Pakhomovskaya N. L., Chetkina T. S. Mediko-statisticheskii obzor «Zabole vae most' khronicheskimi virusnymi gepatitami detskogo naseleniya v Rossiiskoi Federatsii za period 2003–2011 gg.» [Medico-statistical review “Rate of Morbidity of Chronic Viral Hepatitis in Children in Russian Federation during the period of 2003–2011”]. Moscow, 2012. pp. 1–15.; Volynets G. V., Skvortsova T. A. Mediko-sotsial'naya ekspertiza i reabilitatsiya — Medico-social evaluation and rehabilitation. 2011; 3–4: 30–48.; Bortolotti F., Vajro P., Cadrobbi P., Lepore L., Zancan L., Barbera C., Crivellaro C. et al. Cryptogenic chronic liver disease and hepatitis C virus infection in children. J. Hepatol. 1992; 15: 73–76.; Kage M., Fujisawa T., Shiraki K., Tanaka T., Kimura A., Shi-ma matsu K., Nakashima E. et al. Pathology of chronic hepatitis C in children. Child Liver Study Group of Japan. Hepatology. 1997; 26: 771–775.; Badizadegan K., Jonas M. M., Ott M. J., Nelson S. P., PerezAtayde A. R. Histopathology of the liver in children with chronic hepatitis C viral infection. Hepatology. 1998; 28: 1416–1423.; Guido M., Rugge M., Jara P., Hierro L., Giacchino R., Larra-uri J., Zancan L. et al. Chronic hepatitis C in children: the pathological and clinical spectrum. Gastroenterology. 1998; 115: 1525–1529.; Birnbaum A. H., Shneider B. L., Moy L. Hepatitis C in children. N. Engl. J. Med. 2000; 342: 290–291.; Gonzblez-Peralta R.P., Langham M. R., Jr., Andres J. M. et al. Hepatocellular carcinoma in 2 young adolescents with chronic hepatitis C. J. Pediatr. Gastroenterol. Nutr. 2009; 48: 630–635.; Mohan P., Colvin C. A., Glymph C. et al. Clinical spectrum and histopathologic features of chronic hepatitis C infection in children. J. Pediatr. 2007; 150: 168–174.; Nydegger A., Srivastava A., Wake M. et al. Health-related quality of life in children with hepatitis C acquired in the first year of life. J. Gastroenterol. Hepatol. 2008; 23: 226–230.; Rodrigue J. R., Balistreri W., Haber B. et al. Impact of hepatitis C virus infection on children and their caregivers: quality of life, cognitive, and emotional outcomes. J. Pediatr. Gastroenterol. Nutr. 2009; 48: 341–347.; Serranti D., Buonsenso D., Ceccarelli M. et al. Pediatric hepatitis C infection: to treat or not to treat… what's the best for the child? Eur. Rev. Med. Pharmacol. Sci. 2011; 15: 1057–1067.; Danish F. A., Koul S. S., Subhani F. R. et al. Managing HCV infection in pediatric age group: suggested recommendations. Saudi J. Gastroenterol. 2010; 16: 230–235.; Yeung L. T., Roberts E. A. Current issues in the management of paediatric viral hepatitis. Liver Int. 2010; 30: 5–18.; Arshad M., El-Kamary S. S., Jhaveri R. Hepatitis C virus infection during pregnancy and the newborn period — are they opportunities for treatment? J. Viral Hepat. 2011; 18: 229–236.; Powell M., Bailey J., Maggio L. A. Clinical inquiries. How should you manage children born to hepatitis C-positive women? J. Fam. Pract. 2010; 59: 289–290.; Tajiri H., Inui A., Kiyohara Y. et al. Peginterferon alpha-2b and ribavirin for the treatment of chronic hepatitis C in Japanese pediatric and young adult patients: a survey of the Japan Society of Pediatric Hepatology. Eur. J. Gastroenterol. Hepatol. 2009; 21: 1256–1260.; American Academy of Pediatrics. Committee on Infectious Diseases. Hepatitis C virus infection. Pediatrics. 1998; 101 (3, Pt. 1): 481–485.; Zhang H. F., Yang X. J., Shu S. S. et al. An open-label pilot study evaluating the efficacy and safety of peginterferon alfa-2a combined with ribavirin in children with chronic hepatitis C. Zhonghua Shi. Yan. He. Lin. Chuang Bing. Du. Xue. Za. Zhi. 2005; 19: 185–187.; Schwarz K. B., Mohan P., Markewicz C. et al. Safety, efficacy and pharmacokinetics of peginterferon alpha2a (40 kd) in children with chronic hepatitis C. J. Pediatr. Gastroenterol. Nutr. 2006; 43: 499–505.; Volynets G. V., Potapov A. S., Pakhomovskaya N. L., Skvortsova T. A. Povyshenie effektivnosti i bezopasnosti interferonoterapii khronicheskogo gepatita S u detei s pomoshch'yu Ronkoleikina — rekombinantnogo interleikina 2. Mediko-statisticheskii obzor «Zabolevaemost' khronicheskimi virusnymi gepatitami detskogo naseleniya v Rossiiskoi federatsii za period 2003–2011 gg.» [Efficiency and Safe Improvement of IF Therapy of Chronic Hepatitis C in Children with the Help of Roncoleikin — recombinant interleukin 2. Medicostatistical review “Rate of Morbidity of Chronic Viral Hepatitis in Children in Russian Federation during the period of 2003–2011”]. Moscow, 2012. pp. 33–37.; The European Association for the Study of the Liver (EASL). Monitoring response to treatment and duration of HCV treatment depending on the severity of virology response with the HCV genotype. Vienna, Austria. 2010.; Pellicano R. Autoimmune manifestation during interferon therapy in patients with chronic hepatitis C: the hepatologist’s view. Minerva Gastroenterol. Dietol. 2005; 51: 55–61.; Peck-Radosavljevic M. Rapid suppression of hematopoiesis by standard or pegylated interferon-alpha. Gastroenterology. 2002; 123: 141–151.; https://vsp.spr-journal.ru/jour/article/view/333
-
15Academic Journal
المؤلفون: A. N. Surkov, A. S. Potapov, A. L. Lozovator, E. L. Tumanova, А. Н. Сурков, А. С. Потапов, А. П. Лозоватор, Е. Л. Туманова
المصدر: Current Pediatrics; Том 12, № 6 (2013); 24-28 ; Вопросы современной педиатрии; Том 12, № 6 (2013); 24-28 ; 1682-5535 ; 1682-5527
مصطلحات موضوعية: морфология, glycogenosis, liver, morphology, гликогеновая болезнь, печень
وصف الملف: application/pdf
Relation: https://vsp.spr-journal.ru/jour/article/view/252/179; Baranov A. A., Namazova-Baranova L. S., Surkov A. N., Potapov A. S., Bakanov M. I., Polyakova S. I., Gundobina O. S., Lozovator A. L. Glikogenovaya bolezn u detey: uchebnoe posobie [Glycogen disease in children: A Tutorial]. Moscow, Pediatr Publ., 2012. 128 p.; Krasnopolskaya K. D. Nasledstvennye bolezni obmena veshchestv. Spravochnoye posobiye dlya vrachey [Hereditary metabolic diseases. A Reference Guide for Physicians]. Moscow, Tsentr sotsialnoy adaptatsii i reabilitatsii detey “Fokhat” [Fohat — Centre for social adaptation and rehabilitation of children]. 2005. 364 p.; Atlas redkikh bolezney [Atlas of Rare Diseases]. Edited by A. A. Baranov, L. S. Namazova-Baranova. Moscow, Pediatr Publ. 2013. 304 p.; Rozenfeld E. L., Popova I. A. Glikogenovaya bolezn [Glycogen disease]. Moscow, Meditsina Publ., 1979. 288 p.; Ozen H. Glycogen storage diseases: new perspectives. World J. Gastroenterol. 2007; 13 (18): 2541–2553.; Surkov A. N. Glikogenovaya bolezn u detey: sovremennye predstavleniya (Chast I) [Glycogen disease in children: Modern concepts (Part I)]. Voprosy sovremennoy pediatrii — Current Pediatrics. 2012; 11 (2): 30–42.; Dvoryakovskaya G. M., Uvarova E. V., Dvoryakovskiy I. V., Chistova L. V., Chibisov I. V., Egorova M. V. Rol ultrazvukovoy diagnostiki pri obsledovanii detey s pechenochnoy formoy glikogenozov [Role of ultrasound diagnosis when examining children with hepatic form of glycogenosis]. Ultrazvukovaya i funktsionalnaya diagnostika — Ultrasound and functional diagnostics. 2002; 4: 53–59.; Popovich Yu. G., Chibisov I. V., Potapova-Vinogradova I. N., Chistova L. V. Kliniko-biokhimicheskie i morfologicheskie osobennosti pechenochnoy formy glikogenozov u detey [Clinical, biochemical and morphological features of hepatic glycogenosis in children]. Pediatriya — Pediatrics. 1988; 1: 35–39.; Pathology of the Liver: 4th edition. R. N. M. MacSween, A. D. Burt, B. C. Portman et al. (eds.). London: Churchill Livingstone. 2001. 982 p.; Surkov A. N., Potapov A.S., Tumanova E. L. Rezultaty morfologicheskogo issledovaniya pecheni u detey s glikogenozami [Results of the morphological study of liver in children with glycogen storage]. Gastroenterologiya Sankt-Peterburga — Gastroenterology of St. Petersburg. 2009; 2–3: 76–77.; Surkov A., Potapov A., Tumanova E., Averkina N. Morphological changes of the liver in children with hepatic type of glycogen storage disease. 4th Europaediatrics, 03–06 July 2009. Moscow. Р. 632.; Knodell R. G., Ishak K. G., Black W. C. et al. Formulation and application of a numerical scoring system for assessing histological activity in asymptomatic chronic active hepatitis. Hepatology. 1981; 1 (5): 431–435.; Desmet V. J., Gerber M., Hoofnagle J. H. et al. Classification of chronic hepatitis: diagnosis, grading and staging. Hepatology. 1994; 19 (6): 1513–1520.; Jevon G. P., Finegold M. J. Reliability of histological criteria in glycogen storage disease of the liver. Pediatr. Pathol. 1994; 14 (4): 709–721.; Gogus S., Kocak N., Ciliv G., Karabulut E., Akcoren Z., Kale G., Caglar M. Histologic features of the liver in type Ia glycogen storage disease: comparative study between different age groups and consecutive biopsies. Pediatr. Dev. Pathol. 2002; 5 (3): 299–304.; Kishnani P. S., Austin S. L., Arn P., Bali D. S., Boney A., Case L. E., Chung W. K., Desai D. M., El-Gharbawy A., Haller R., Smit G. P., Smith A. D., Hobson-Webb L. D., Wechsler S. B., Weinstein D. A., Watson M. S. Glycogen storage disease type III diagnosis and management guidelines. Genet. Med. 2010; 12 (7): 446–463.; https://vsp.spr-journal.ru/jour/article/view/252
-
16Academic JournalEVALUATION OF LIVER FAILURE STAGE IN CHILDREN ; ОПРЕДЕЛЕНИЕ СТЕПЕНИ НАРУШЕНИЯ ФУНКЦИИ ПЕЧЕНИ У ДЕТЕЙ
المؤلفون: G. V. Volynets, A. S. Potapov, S. I. Polyakova, A. E. Aleksandrov, V. M. Senyakovich, A. N. Surkov, T. S. Chetkina, N. N. Evlukhina, T. A. Skvortsova, Г. В. Волынец, А. С. Потапов, С. И. Полякова, А. Е. Александров, В. М. Сенякович, А. Н. Сурков, Т. С. Четкина, Н. Н. Евлюхина, Т. А. Скворцова
المصدر: Current Pediatrics; Том 12, № 4 (2013); 47-51 ; Вопросы современной педиатрии; Том 12, № 4 (2013); 47-51 ; 1682-5535 ; 1682-5527
مصطلحات موضوعية: нарушения, liver functions, failure, функции печени
وصف الملف: application/pdf
Relation: https://vsp.spr-journal.ru/jour/article/view/304/221; Puzin S. N., Lavrova D. I., Kuzmishin L. E. etc. Reglamenty ekspertno-reabilitatsionnoi diagnostiki dlya glavnogo byuro medikosotsial'noi ekspertizy (inf. pis'mo) [Regulations of Expert and Rehabilitation Diagnosis for Central Bureau of Social Security Medical Assessment Board. Informational Letter]. Moscow, 2006. pp. 12–19.; Agata I.D.D', Balistreri W. F. Evaluation of liver diseases in the pediatric patient. Pediatr. Rev. 1999; 20 (11): 376–389.; McDiarmid S. V., Anand R., Lindblad A. S. Development of a pediatric end-stage liver disease score to predict poor outcome in children awaiting liver transplantation. Transplantation. 2002; 74 (2): 173–181.; Murray K. F., Carithers R. L. AASLD Practice Guidelines: Evaluation of the Patient for Liver Transplantation. Hepatology. 2005; 41 (6): 1–26.; Mezhdunarodnaya klassifikatsiya funktsionirovaniya, ogranichenii zhiznedeyatel'nosti i zdorov'ya [International Classification of Functioning, Disability and Health]. Translated from English by G. D. Shostka, V.Yu. Ryasnyanskii, A. V. Kvashin etc. WHO, Geneva, 2001. 342 p.; Komov V. P., Shvedova V. N. Biokhimiya: Uchebn. dlya vuzov (Vysshee obrazovanie: Sovremennyi uchebnik) [Biochemistry: Textbook for Institutions of Higher Education (Higher Education: Contemporary Textbook)]. Moscow, Drofa, 2004. 640 p.; Shevchenko O. P. Laboratoriya — Laboratory. 1996; 1: 3–6.; Bairoch A. The ENZYME database in 2000. Nucleic Acids Res. 2000; 28: 304–305.; Aleshkin V. A., Novikova L. I., Lyutov A. G., Aleshkina T.N. Klinicheskaya meditsina — Clinical Medicine. 1988; 8: 39–48.; Volodin N. N., Dolgov V. V., Degtyarev D. N., Rakov S. S., Lipagina A. A., Krivonozhko A. V. Rossiyskiy Vestnik Perinatologii i Pediatrii — Russian Bulletin of Perinatology and Pediatrics. 2000; 1: 10–13.; Fomin V. V., Kozlovskaya L.V. Consilium medicum — Consiliummedicum. 2003; 5 (5): 247–250.; Koj A. Metabolic studies of acute-phase proteins. Pathophysiology of plasma protein metabolism. N.-Y., London. 1984. P. 221–248.; Wolberg A. S. Thrombin generation and fibrin clot structure. Blood Rev. 2007; 21 (3): 131–142.; Zaborske J.M., Wu Xiaochen, Wek Ronald C., Pan Tao. Selective control of amino acid metabolism by the GCN2 eIF2 kinase pathway in Saccharomyces cerevisiae. BMC Biochemistry. 2010; 11: 29.; Grady J. G., Lake J. R., Howdle P. D. Glycogen storage disease, comprehensive clinical hepatology. Gastrointest. Liver Dis. 2000; 715: 122–128.; Berman R. E., Kligman R. M., Jackson H. B. Pediatriya po Nel'sonu: v 5 t. Per. s angl [Nelson Textbook of Pediatrics: in 5 Volumes. Translated from English. 4th Volume]. Edited by A. A. Baranov. Moscow, Rid Elsiver, 2009. 1112 p.; Bluger A. F. Osnovy gepatologii [Basics of Pathology]. Riga, Zvaigzne, 1975. 469 p.; J. I. Wolfsdorf, J. F. Crigler, Jr., Rukovodstvo po endokrinologii. Glikogenozy [Endocrinology Guideline. Glycogen Storage Diseases]. 1990. pp. 741–755.; Colman J., Rem K.-T. Naglyadnaya Biokhimiya [Visual Biochemistry]. Moscow, Mir, 2000. 469 p.; Cohn R., Roth K. Rannyaya diagnostika boleznei obmena veshchestv [Metabolic Diseases: A Guide to Early Recognition]. Moscow, Meditsina, 1986. 637 p.; Metzler D. Biokhimiya. Per. s angl. T. 2 [Biochemistry. Translated from English. 2nd Volume]. Moscow, 1980. 609 p.; Nikolaev A.Ya. Biologicheskaya khimiya [Biological Chemistry]. Moscow, MIA, 2004. 568 p.; Severin E. S. Biokhimiya [Biochemistry]. Moscow, GEOTARMedia, 2003. 779 p.; Sherlok Sh., Duli J. Zabolevaniya pecheni i zhelchnykh putei [The diseases of the liver and biliary tract]. Moscow, GEOTAR-Media, 1999. 864 p.; Baranov A. A. etc. Diagnostika i lechenie khronicheskikh virusnykh gepatitov B, C i D u detei. Pos. dlya vrachei [Diagnosis and Treatment of Chronic Viral Hepatitis B, C and D in Children. Guideline for Doctors.]. Moscow, 2003. 84 p.; https://vsp.spr-journal.ru/jour/article/view/304
-
17Academic Journal
المؤلفون: A. N. Surkov, V. V. Chernikov, A. A. Baranov, L. S. Namazova-Baranova, A. S. Potapov, I. V. Vinyarskaya, А. Н. Сурков, В. В. Черников, А. А. Баранов, Л. С. Намазова-Баранова, А. С. Потапов, И. В. Винярская
المصدر: Pediatric pharmacology; Том 10, № 4 (2013); 90-94 ; Педиатрическая фармакология; Том 10, № 4 (2013); 90-94 ; 2500-3089 ; 1727-5776
مصطلحات موضوعية: дети, life quality, PedsQL, rehabilitation, children, качество жизни, реабилитация
وصف الملف: application/pdf
Relation: https://www.pedpharma.ru/jour/article/view/199/285; Розенфельд Е. Л., Попова И. А. Гликогеновая болезнь. М.: Медицина. 1979. С. 288.; Фармакотерапия детских болезней: Руководство для врачей. Под ред. А. Д. Царегородцева. М.: ООО «Медицинское информационное агентство». 2010. С. 880.; Зайчик А. Ш., Чурилов Л. П. Основы общей патологии. Часть 2. Основы патохимии. Учебник-пособие для студентов медицинских ВУЗов. СПб.: ЭЛБИ. 2000. С. 688.; Краснопольская К. Д. Наследственные болезни обмена веществ. Справочное пособие для врачей. М.: РОО «Центр социальной адаптации и реабилитации детей «Фохат». 2005. С. 364.; Баранов А. А., Намазова-Баранова Л. С., Сурков А. Н., Потапов А. С., Баканов М. И., Полякова С. И., Гундобина О. С., Лозоватор А. Л. Гликогеновая болезнь у детей. М.: Союз педиатров России. 2012. 128 с.; Ozen H. Glycogen storage diseases: new perspectives. World J. Gastroenterol. 2007; 13 (18): 2541–2553.; Попович Ю. Г., Чибисов И. В., Потапова-Виноградова И. Н. и др. Клинико-биохимические и морфологические особенности печеночной формы гликогенозов у детей. Педиатрия. 1988; 1: 35–39.; Клиническая диетология детского возраста: Руководство для врачей. Под ред. Т. Э. Боровик, К. С. Ладодо. М.: ООО «Медицинское информационное агентство». 2008. C. 608.; Goldbeck L., Melches J. Quality of life in families of children with congenital heart disease. Qual Life Res. 2005; 14 (8): 1915–1924.; Bisegger C., Cloetta B., von Rueden U. Health-related quality of life: gender differences in childhood and adolescence. Soz Praventivmed. 2005; 50 (5): 281–291.; Thwaites R. M. A., Price M. S. Уменьшение бремени бронхиальной астмы: улучшение качества жизни пациентов. Пульмонология. 1998; 3: 19–23.; Баранов А. А., Альбицкий В. Ю., Винярская И. В. Изучение качества жизни в педиатрии. М.: Союз педиатров России. 2010. C. 272. (Серия «Социальная педиатрия»; вып. 10).; Михайлова Т. Л., Румянцев В. Г., Кольченко И. И. и др. Качество жизни больных с синдромом раздраженного кишечника. Российский журнал гастроэнтерологии, гепатологии, колопроктологии. 2002; 12 (6): 70–75.; Новик А. А., Ионова Т. И. Руководство по исследованию качества жизни в медицине. СПб.: Нева, М.: ОЛМА-ПРЕСС. 2002. С. 321.; Новик А. А., Денисов Н. Л., Ионова Т. И. и др. Качество жизни больных язвенной болезнью желудка и двенадцатиперстной кишки. Материалы конференции «Исследование качества жизни в медицине». СПб., 2000. С. 97–98.; Гурылева М. Э., Карданова Л. Д. Проблема качества жизни в медицине. Нальчик: Эль-Фа. 2007. С. 96.; Подсеваткин В. Г., Токарева Н. Г. Клинико-психопатологическая и психологическая оценка качества жизни больных эпилепсией: методическое пособие. Саранск. 2000. С. 24.; Сурков А. Н., Черников В. В. Актуальные проблемы реабилитации детей с гликогеновой болезнью. Медико-социальная экспертиза и реабилитация детей с ограниченными возможностями. 2011; 1–2: 48–53.; Черников В. В., Сурков А. Н., Потапов А. С. Оценка качества жизни детей с гликогеновой болезнью. Сборник материалов XVI Съезда педиатров России «Актуальные проблемы педиатрии». Москва. 2009. С. 436–437.; Черюканов А. В., Витрищак А. А. Динамика качества жизни у детей и подростков с ограниченными возможностями. Вестн. СПб. Гос. мед. акад. им. И. И. Мечникова. 2007; 3: 24–28.; Fadrowski J., Cole S. R., Hwang W. et al. Changes in physical and psychosocial functioning among adolescents with chronic kidney disease. Pediatr Nephrol. 2006; 21 (3): 394–399.; Marklund B., Ahlstedt S., Nordstrom G. Health-related quality of life in food hypersensitive schoolchildren and their families: parents' perceptions. Health Qual Life Outcomes. 2006; 10 (4): 48.; Тимофеева А. Г., Винярская И. В. Исследование качества жизни у детей с хроническими заболеваниями. Вопросы современной педиатрии. 2008; 7 (6): 28–29.; Колбасина Е. В., Воробьева В. А., Азова Е. А., Рассохин В.Ф. Качество жизни детей и подростков, больных сахарным диабетом типа 1. Вопросы современной педиатрии. 2009; 8 (5): 14–18.; De Civita M., Regier D., Alamgir A. H. et al. Evaluating healthrelated quality-of-life studies in paediatric populations: some conceptual, methodological and developmental considerations and recent applications. Pharmacoeconomics. 2005; 23 (7): 659–685.; https://www.pedpharma.ru/jour/article/view/199
-
18Academic Journal
المؤلفون: V. V. Botvinyeva, O. B. Gordeevа, A. S. Potapov, L. S. Namazova-Baranovа, I. V. Zubkova, A. O. Anushenko, I. Z. Dzhgarkava, A. K. Gevorkyan, M. A. Soloshenko, В. В. Ботвиньева, О. Б. Гордеева, А. С. Потапов, Л. С. Намазова-Баранова, И. В. Зубкова, А. О. Анушенко, И. З. Джгаркава, М. А. Солошенко, А. К. Геворкян
المصدر: Pediatric pharmacology; Том 10, № 5 (2013); 52-55 ; Педиатрическая фармакология; Том 10, № 5 (2013); 52-55 ; 2500-3089 ; 1727-5776
مصطلحات موضوعية: маркеры воспаления, ulcerative colitis, peripheral blood parameters, inflammatory markers, язвенный колит, показатели периферической крови
وصف الملف: application/pdf
Relation: https://www.pedpharma.ru/jour/article/view/175/243; https://www.pedpharma.ru/jour/article/view/175/244; Александрова Ю. Н. О системе цитокинов. Педиатрия. 2007; 86 (3): 124–128.; Кетлинский С. А., Симбирцев А. С. Цитокины. Санкт-Петербург: Фолиант. 2008. 550 с.; Ботвиньева В. В., Гордеева О. Б., Намазова-Баранова Л. С. Перспективы диагностики и лечения различных видов анемии у детей. Педиатрическая фармакология. 2012; 9 (5): 35–40.; Dan Turner, Simon P. L. Travis, Anne M. Griffiths, Frank M. Ruemmele, Arie Levine, Eric I. Benchimol, Marla Dubinsky, George Alex, Robert N. Baldassano, Jacob C. Langer, Robert Shamberger. The American Journal of Gastroenterology. 2011; 1038 (10): 574–588.; Briggs C. New red cell parameters on the Sysmex XE-2100 as potential markers of functional iron deficiency. Infus Ther Transf Med. 2001; 28: 256–258.; Ganz T. Hepcidin, a key regulator of iron metabolism and mediator of anemia of inflammation. Blood. 2003; 102: 783–788.; Титов В. Н. С-реактивный белок — тест нарушения «чистоты» межклеточной среды организма при накоплении «биологического мусора» большой молекулярной массы. Клиническая лабораторная диагностика. 2008; 2: 3–14.; Гордеева О. Б., Ботвиньева В. В., Намазова-Баранова Л. С. Эритроцитарные и ретикулоцитарные индексы у пациентов с воспалительными заболеваниями различного генеза. Педиатрическая фармакология. 2012; 9 (6): 110–112.; Намазова-Баранова Л. С. Научные исследования и инфраструктура платформы «Педиатрия». Педиатрическая фармакология. 2012; 9 (4): 15–24.; https://www.pedpharma.ru/jour/article/view/175
-
19Academic Journal
المؤلفون: N. L. Pakhomovskaya, A. S. Potapov, A. O. Anushenko, Н. Л. Пахомовская, А. С. Потапов, А. О. Анушенко
المصدر: Meditsinskiy sovet = Medical Council; № 1 (2013); 28-35 ; Медицинский Совет; № 1 (2013); 28-35 ; 2658-5790 ; 2079-701X ; 10.21518/2079-701X-2013-1
مصطلحات موضوعية: chronic gastritis, диспепсия, дети, Римские критерии, гастроэзофагеальный рефлюкс, прокинетики, хронический гастрит, functional disorders of gastro-intestinal tract, dyspepsia, children, Roman criteria, micturition, gastro-esophageal reflux, prokinetics
وصف الملف: application/pdf
Relation: https://www.med-sovet.pro/jour/article/view/855/855; https://www.med-sovet.pro/jour/article/view/855
-
20Academic Journal
المؤلفون: A. S. Potapov, E. I. Alieva, T. V. Gabruzskaya, A. V. Gorelov, I. N. Zakharova, E. А. Kornienko, V. N. Panfilova, D. V. Pechkurov, M. A. Ratnikova, M. O. Revnova, E. P. Sitnikova, N. I. Ursova, E. N. Fedulova, E. G. Tsimbalova, P. V. Shumilov, O. V. Shcherbakova, N. E. Shchigaleva, А. С. Потапов, Э. И. Алиева, Т. В. Габрузская, А. В. Горелов, И. Н. Захарова, Е. А. Корниенко, В. Н. Панфилова, Д. В. Печкуров, М. А. Ратникова, М. О. Ревнова, Е. П. Ситникова, Н. И. Урсова, Э. Н. Федулова, Е. Г. Цимбалова, П. В. Шумилов, О. В. Щербакова, Н. Е. Щиголева
المصدر: Current Pediatrics; Том 12, № 3 (2013); 18-30 ; Вопросы современной педиатрии; Том 12, № 3 (2013); 18-30 ; 1682-5535 ; 1682-5527
مصطلحات موضوعية: педиатрический индекс активности язвенного колита, clinical recommendations, diagnostics, treatment, pediatric ulcerative colitis activity index, дети, клинические рекомендации, диагностика и лечение
وصف الملف: application/pdf
Relation: https://vsp.spr-journal.ru/jour/article/view/326/229; Benchimol E. I., Fortinsky K. J., Gozdyra P. et al. Epidemiology of pediatric inflammatory bowel disease: a systematic review of international trends. Inflamm. Bowel. Dis. 2011; 17: 423–439.; Henderson P., Hansen R., Cameron F. L. et al. Rising incidence of pediatric inflammatory bowel disease in Scotland. Inflamm. Bowel. Dis. 2012; 18: 999–1005.; Heyman M. B., Kirschner B. S., Gold B. D. et al. Children with earlyonset inflammatory bowel disease (IBD): analysis of a pediatric IBD consortium registry. J. Pediatr. 2005; 146: 35–40.; Van Limbergen J., Russell R. K., Drummond H. E. et al. Definition of phenotypic characteristics of childhood-onset inflammatory bowel disease. Gastroenterology. 2008; 135: 1114–1122.; Gower-Rousseau C., Dauchet L., Vernier-Massouille G. et al. The natural history of pediatric ulcerative colitis: a population-based cohort study. Am. J. Gastroenterol. 2009; 104: 2080–2088.; Travis S. P. L., Stange E. F., Leґmann M. et al. European evidencebased consensus on the management of ulcerative colitis: current management. J. Crohn Colitis. 2008; 2: 24–62.; Turner D., Travis S. P., Griffiths A. M. et al. Consensus for managing acute severe ulcerative colitis in children: a systematic review and joint statement from ECCO, ESPGHAN, and the Porto IBDWorking Group of ESPGHAN. Am. J. Gastroenterol. 2011; 106: 574–588.; Glickman J. N., Bousvaros A., Farraye F. A. et al. Pediatric patients with untreated ulcerative colitis may present initially with unusual morphologic findings. Am. J. Surg. Pathol. 2004; 28: 190–197.; Levine A., de Bie C. L., Turner D. et al. Atypical disease phenotypes in paediatric ulcerative colitis: 5-year analyses of the EUROKIDS registry. Inflamm. Bowel. Dis. 2012 [Epub ahead of print].; Robert M. E., Tang L., Hao L. M. et al. Patterns of inflammation in mucosal biopsies of ulcerative colitis: perceived differences in pediatric populations are limited to children younger than 10 years. Am. J. Surg. Pathol. 2004; 28: 183–189.; Haskell H., Andrews C. W., Jr., Reddy S. I. et al. Pathologic features and clinical significance of «backwash» ileitis in ulcerative colitis. Am. J. Surg. Pathol. 2005; 29: 1472–1481.; de Bie C. I., Buderus S., Sandhu B. K. et al. Diagnostic workup of paediatric patients with inflammatory bowel disease in Europe: results of a 5-year audit of the EUROKIDS registry. J. Pediatr. Gastroenterol. Nutr. 2012; 54: 374–380.; Koletzko S., Niggemann B., Arato A. et al. Diagnostic approach and management of cow’s milk protein allergy in infants and children: a practical guideline of the GI-committee of ESPGHAN. J Pediatr Gastroenterol Nutr. 2012; 55: 221–229.; Glocker E. O., Frede N., Perro M. et al. Infant colitis — it’s in the genes. Lancet. 2010; 376: 1272. 15. Glocker E. O., Kotlarz D., Boztug K. et al. Inflammatory bowel disease and mutations affecting the interleukin-10 receptor. N. Engl. J. Med. 2009; 361: 2033–2045.; Begue B., Verdier J., Rieux-Laucat F. et al. Defective IL10 signaling defining a subgroup of patients with inflammatory bowel disease. Am. J. Gastroenterol. 2011; 106: 1544–1555.; Beattie R. M., Nicholls S. W., Domizio P. et al. Endoscopic assessment of the colonic response to corticosteroids in children with ulcerative colitis. J. Pediatr. Gastroenterol. Nutr. 1996; 22: 373–379.; Mack D. R., Langton C., Markowitz J. et al. Laboratory values for children with newly diagnosed inflammatory bowel disease. Pediatrics. 2007; 119: 1113–1119.; Weinstein T. A., Levine M., Pettei M. J. et al. Age and family history at presentation of pediatric inflammatory bowel disease. J. Pediatr. Gastroenterol. Nutr. 2003; 37: 609–613.; Henderson P., Casey A., Lawrence S. J. et al. The diagnostic accuracy of fecal calprotectin during the investigation of suspected pediatric inflammatory bowel disease. Am. J. Gastroenterol. 2012; 107: 941–949.; Venediktova M. M., Tsimbalova E. G., Potapov A. S., Mayanskii N. A. Voprosi diagnostiki v pediatrii — Diagnostics in pediatrics. 2011; 3 (6): 23–26.; Turner D., Leach S. T., Mack D. et al. Faecal calprotectin, lactoferrin, M2-pyruvate kinase and S100A12 in severe ulcerative colitis: a prospective multicentre comparison of predicting outcomes and monitoring response. Gut. 2010; 59: 1207–1212.; Sylvester F. A., Turner D., Draghi A., 2nd et al. Fecal osteoprotegerin may guide the introduction of second-line therapy in hospitalized children with ulcerative colitis. Inflamm. Bowel. Dis. 2011; 17: 1726–1730.; Harris M. S., Lichtenstein G. R. Review article: delivery and efficacy of topical 5-aminosalicylic acid (mesalazine) therapy in the treatment of ulcerative colitis. Aliment. Pharmacol. Ther. 2011; 33: 996–1009.; Lichtenstein G. R., Kamm M. A. Review article: 5-aminosalicylate formulations for the treatment of ulcerative colitis — methods of comparing release rates and delivery of 5-aminosalicylate to the colonic mucosa. Aliment. Pharmacol. Ther. 2008; 28: 663–673.; Sutherland L., Macdonald J. K. Oral 5-aminosalicylic acid for induction of remission in ulcerative colitis. Cochr. Database Syst. Rev. 2006: CD000543.; Sutherland L., Macdonald J. K. Oral 5-aminosalicylic acid for maintenance of remission in ulcerative colitis. Cochr. Database Syst. Rev. 2006: CD000544.; Odera G., Giuliani B., Santini B. et al. Topical treatment with 5-SA and hydrocortisone. Riv. Ital. Pediatr. 1986; 12: 674–678.; Ford A. C., Khan K. J., Sandborn W. J. et al. Efficacy of topical 5-aminosalicylates in preventing relapse of quiescent ulcerative colitis: a meta-analysis. Clin. Gastroenterol. Hepatol. 2012; 10: 513–519.; Kolho K. L., Raivio T., Lindahl H. et al. Fecal calprotectin remains high during glucocorticoid therapy in children with inflammatory bowel disease. Scand. J. Gastroenterol. 2006; 41: 720–725.; Powell-Tuck J., Bown R. L., Lennard-Jones J. E. A comparison of oral prednisolone given as single or multiple daily doses for active proctocolitis. Scand. J. Gastroenterol. 1978; 13: 833–837.; Uchida K., Araki T., Toiyama Y. et al. Preoperative steroid-related complications in Japanese pediatric patients with ulcerative colitis. Dis. Colon Rectum. 2006; 49: 74–79.; Gionchetti P., Rizzello F., Ferrieri A. et al. Rifaximin in patients with moderate or severe ulcerative colitis refractory to steroidtreatment: a double-blind, placebo-controlled trial. Dig. Dis. Sci. 1999; 44: 1220–1221.; Ohkusa T., Kato K., Terao S. et al. Newly developed antibiotic combination therapy for ulcerative colitis: a double-blind placebo-controlled multicenter trial. Am. J. Gastroenterol. 2010; 105: 1820–1829.; Timmer A., McDonald J. W., Macdonald J. K. Azathioprine and 6-mercaptopurine for maintenance of remission in ulcerative colitis. Cochr. Database Syst. Rev. 2007: CD000478.; Gisbert J. P., Linares P. M., McNicholl A. G. et al. Meta-analysis: the efficacy of azathioprine and mercaptopurine in ulcerative colitis. Aliment. Pharmacol. Ther. 2009; 30: 126–137.; Khan K. J., Dubinsky M. C., Ford A. C. et al. Efficacy of immunosuppressive therapy for inflammatory bowel disease: a systematic review and meta-analysis. Am. J. Gastroenterol. 2011; 106: 630–642.; Barabino A., Torrente F., Ventura A. et al. Azathioprine in paediatric inflammatory bowel disease: an Italian multicentre survey. Aliment. Pharmacol. Ther. 2002; 16: 1125–1130.; Kader H. A., Mascarenhas M. R., Piccoli D. A. et al. Experiences with 6-mercaptopurine and azathioprine therapy in pediatric patients with severe ulcerative colitis. J. Pediatr. Gastroenterol. Nutr. 1999; 28: 54–58.; Verhave M., Winter H. S., Grand R. J. Azathioprine in the treatment of children with inflammatory bowel disease. J. Pediatr. 1990; 117: 809–814.; Pozler O., Chladek J., Maly J. et al. Steady-state of azathioprine during initiation treatment of pediatric inflammatory bowel disease. J. Crohns Colitis. 2010; 4: 623–628.; Sandborn W. J. A review of immune modifier therapy for inflam matory bowel disease: azathioprine, 6-mercaptopurine, cyclosporine, and methotrexate. Am. J. Gastroenterol. 1996; 91: 423–433.; De Greef E., Vandenplas Y., Veereman-Wauters G. Opportunistic infections in paediatric inflammatory bowel disease patients. Arch. Dis. Child. 2011; 91: 5–7.; Russell R. K., Wilson M. L., Loganathan S. et al. A British Society of Paediatric Gastroenterology, Hepatology and Nutrition survey of the effectiveness and safety of adalimumab in children with inflammatory bowel disease. Aliment. Pharmacol. Ther. 2011; 33: 946–953.; Turner D., Griffiths A. M. Acute severe ulcerative colitis in children: a systematic review. Inflamm. Bowel. Dis. 2011; 17: 40–449.; Lawson M. M., Thomas A. G., Akobeng A. K. Tumour necrosis factor alpha blocking agents for induction of remission in ulcerative colitis. Cochr. Database Syst. Rev. 2006: CD005112.; Rutgeerts P., Sandborn W. J., Feagan B. G. et al. Infliximab for induction and maintenance therapy for ulcerative colitis. N. Engl. J. Med. 2005; 353: 2462–2476.; Hyams J., Damaraju L., Blank M. et al. Induction and maintenance therapy with infliximab for children with moderate to severe ulcerative colitis. Clin. Gastroenterol. Hepatol. 2012; 10: 391–399.; Baranov A. A., Potapov A. S., Tsimbalova E. G. Vestnik Rossiiskoi akademii meditsinskikh nauk — Annals of the Russian Academy of Medical Sciences. 2011; 6: 36–41.; Hyams J. S., Lerer T., Griffiths A. et al. Outcome following infliximab therapy in children with ulcerative colitis. Am. J. Gastroenterol. 2010; 105: 1430–1436.; Panaccione R., Ghosh S., Middleton S. et al. Infliximab, azathioprine or infliximabюazathioprine for treatment of moderate to severe ulcerative colitis. The UC SUCCESS trial. J. Crohns Colitis. 2011; 5: 13.; Lichtenstein G. R., Diamond R. H., Wagner C. L. et al. Clinical trial: benefits and risks of immunomodulators and maintenance infliximab for IBD-subgroup analyses across four randomized trials. Aliment. Pharmacol. Ther. 2009; 30: 210–226.; Chaparro M., Guerra I., Munoz-Linares P. et al. Systematic review: antibodies and anti-TNF-alpha levels in inflammatory bowel disease. Aliment. Pharmacol. Ther. 2012; 35: 971–986.; Bernstein C. N., Fried M., Krabshuis J. H. et al. World Gastroenterology Organization Practice Guidelines for the diagnosis and management of IBD in 2010. Inflamm. Bowel. Dis. 2010; 16: 112–124.; Gonzalez-Huix F., Fernandez-Banares F., Esteve-Comas M. et al. Enteral versus parenteral nutrition as adjunct therapy in acute ulcerative colitis. Am. J. Gastroenterol. 1993; 88: 227–232.; McIntyre P. B., Powell-Tuck J., Wood S. R. et al. Controlled trial of bowel rest in the treatment of severe acute colitis. Gut. 1986; 27: 481–485.; Dickinson R. J., Ashton M. G., Axon A. T. et al. Controlled trial of intravenous hyperalimentation and total bowel rest as an adjunct to the routine therapy of acute colitis. Gastroenterology. 1980; 79: 1199–1204.; Barabino A., Tegaldo L., Castellano E. et al. Severe attack of ulcerative colitis in children: retrospective clinical survey. Dig. Liver Dis. 2002; 34: 44–49.; Bechtold S., Alberer M., Arenz T. et al. Reduced muscle mass and bone size in pediatric patients with inflammatory bowel disease. Inflamm. Bowel Dis. 2010; 16: 216–225.; Werkstetter K. J., Pozza S. B., Filipiak-Pittroff B. et al. Long-term development of bone geometry and muscle in pediatric inflammatory bowel disease. Am. J. Gastroenterol. 2011; 106: 988–998.; Gokhale R., Favus M. J., Karrison T. et al. Bone mineral density assessment in children with inflammatory bowel disease. Gastroenterology. 1998; 114: 902–911.; Sylvester F. A., Wyzga N., Hyams J. S. et al. Natural history of bone metabolism and bone mineral density in children with inflammatory bowel disease. Inflamm. Bowel. Dis. 2007; 13: 42–50.; Walther F., Fusch C., Radke M. et al. Osteoporosis in pediatric patients suffering from chronic inflammatory bowel disease with and without steroid treatment. J. Pediatr. Gastroenterol. Nutr. 2006; 43: 42–51.; Pappa H., Thayu M., Sylvester F. et al. Skeletal health of children and adolescents with inflammatory bowel disease. J. Pediatr. Gastroenterol. Nutr. 2011; 53: 11–25.; Greenley R. N., Stephens M., Doughty A. et al. Barriers to adherence among adolescents with inflammatory bowel disease. Inflamm. Bowel. Dis. 2010; 16: 36–41.; Hommel K. A., Denson L. A., Baldassano R. N. Oral medication adherence and disease severity in pediatric inflammatory bowel disease. Eur. J. Gastroenterol. Hepatol. 2011; 23: 250–254.; Hommel K. A., Baldassano R. N. Barriers to treatment adherence in pediatric inflammatory bowel disease. J. Pediatr. Psychol. 2010; 35: 1005–1010.; Reed-Knight B., Lewis J. D., Blount R. L. Association of disease, adolescent, and family factors with medication adherence in pediatric inflammatory bowel disease. J. Pediatr. Psychol. 2011; 36: 308–317.; Cassinotti A., Actis G. C., Duca P. et al. Maintenance treatment with azathioprine in ulcerative colitis: outcome and predictive factors after drug withdrawal. Am. J. Gastroenterol. 2009; 104: 2760–2767.; Actis G. C., Fadda M., Pellicano R. et al. The 17-year singlecenter experience with the use of azathioprine to maintain remission in ulcerative colitis. Biomed. Pharmacother. 2009; 63: 362–365.; Turner D., Otley A. R., Mack D. et al. Development and evaluation of a Pediatric Ulcerative Colitis Activity Index (PUCAI): a prospective multicenter study. Gastroenterology. 2007; 133: 423–432.; https://vsp.spr-journal.ru/jour/article/view/326