يعرض 1 - 10 نتائج من 1,151 نتيجة بحث عن '"ДИСЛИПИДЕМИЯ"', وقت الاستعلام: 0.90s تنقيح النتائج
  1. 1
    دورية أكاديمية
  2. 2
    دورية أكاديمية

    المساهمون: The article is sponsored by Novartis., Статья спонсируется компанией «Новартис».

    المصدر: Neurology, Neuropsychiatry, Psychosomatics; Vol 16, No 3 (2024); 136-143 ; Неврология, нейропсихиатрия, психосоматика; Vol 16, No 3 (2024); 136-143 ; 2310-1342 ; 2074-2711 ; 10.14412/2074-2711-2024-3

    وصف الملف: application/pdf

    العلاقة: https://nnp.ima-press.net/nnp/article/view/2284/1671Test; Федеральная служба государственной статистики. Здравоохранение в России. 2019. Статистический сборник. Москва: Росстат; 2019. 170 с. Доступно по ссылке: https://www.gks.ru/storage/mediabank/Zdravoohran-2019.pdfTest; Feigin VL, Owolabi MO. Pragmatic solutions to reduce the global burden of stroke: a World Stroke Organization-Lancet Neurology Commission. Lancet Neurol. 2023 Dec;22(12):1160-206. doi:10.1016/S1474-4422(23)00277-6; Бойцов СА, Проваторов СИ. Возможности диспансерного наблюдения в снижении смертности от ишемической болезни сердца. Терапевтический архив. 2023;95(1):5-10. doi:10.26442/00403660.2023.01.20203; Boehme AK, Esenwa C, Elkind MS. Stroke Risk Factors, Genetics, and Prevention. Circ Res. 2017 Feb 3;120(3):472-95. doi:10.1161/CIRCRESAHA.116.308398; Braunwald E.How to live to 100 before developing clinical coronary artery disease: a suggestion. Eur Heart J. 2022 Jan 31;43(4):249-50. doi:10.1093/eurheartj/ehab532; Kronenberg F, Mora S, Stroes ESG, et al. Lipoprotein(a) in atherosclerotic cardiovascular disease and aortic stenosis: a European Atherosclerosis Society consensus statement. Eur Heart J. 2022 Oct 14;43(39):3925-46. doi:10.1093/eurheartj/ehac361; Reyes-Soffer G, Ginsberg HN, Berglund L, et al. Lipoprotein(a): A Genetically Determined, Causal, and Prevalent Risk Factor for Atherosclerotic Cardiovascular Disease: A Scientific Statement From the American Heart Association. Arterioscler Thromb Vasc Biol. 2022 Jan;42(1):e48-e60. doi:10.1161/ATV.0000000000000147. Epub 2021 Oct 14.; Jawi MM, Frohlich J, Chan SY. Lipoprotein(a) the Insurgent: A New Insight into the Structure, Function, Metabolism, Pathogenicity, and Medications Affecting Lipoprotein(a) Molecule. J Lipids. 2020 Feb 1;2020:3491764. doi:10.1155/2020/3491764; Khovidhunkit W. Lipoprotein(a). Endotext. Oct. 2023. Available at: https://www.ncbi.nlm.nih.gov/books/NBK596274Test/ (accessed 11.03.2024).; Ahmed S, Shah P, Ahmed O. Biochemistry, Lipids. StatPearls. May 2023. Available at: https://www.ncbi.nlm.nih.gov/books/NBK525952Test/ (accessed 30.12.2024).; Iannuzzo G, Tripaldella M, Mallardo V, et al. Lipoprotein(a) Where Do We Stand? From the Physiopathology to Innovative Terapy. Biomedicines. 2021 Jul 19;9(7):838. doi:10.3390/biomedicines9070838; Utermann G, Menzel HJ, Kraft HG, et al. Lp(a) glycoprotein phenotypes. Inheritance and relation to Lp(a)-lipoprotein concentrations in plasma. J Clin Invest. 1987 Aug;80(2):458-65. doi:10.1172/JCI113093; Cegla J, Neely RDG, France M, et al. HEART UK consensus statement on Lipoprotein(a): A call to action. Atherosclerosis. 2019 Dec;291:62-70. doi:10.1016/j.atherosclerosis.2019.10.011. Epub 2019 Oct 14. Erratum in: Atherosclerosis. 2020 Feb 1;296:48.; Koschinsky ML, Kronenberg F. The long journey of lipoprotein(a) from cardiovascular curiosity to therapeutic target. Atherosclerosis. 2022 May;349:1-6. doi:10.1016/j.atherosclerosis.2022.04.017; Shah NP, Pajidipati NJ, McGarrah RW, et al. Lipoprotein (a): An Update on a Marker of Residual Risk and Associated Clinical Manifestations. Am J Cardiol. 2020 Jul 1;126:94-102. doi:10.1016/j.amj-card.2020.03.043. Epub 2020 Apr 7.; Enkhmaa B, Petersen KS, Kris-Etherton PM, Berglund L. Diet and Lp(a): Does Dietary Change Modify Residual Cardiovascular Risk Conferred by Lp(a)? Nutrients. 2020 Jul 7;12(7):2024. doi:10.3390/nu12072024; Feng Z, Xu J, Jin A, et al. Elevated Homocysteine Intensify the Effect of Lipoprotein(a) on Stroke Recurrence. J Am Heart Assoc. 2023 Jan 3;12(1):e026707. doi:10.1161/JAHA.122.026707. Epub 2022 Dec 24.; Yang C, Zhu CG, Sui YG, et al. Synergetic impact of lipoprotein(a) and fibrinogen on stroke in coronary artery disease patients. Eur J Clin Invest. 2024 Feb 16:e14179. doi:10.1111/eci.14179. Epub 2024 Feb 16.; Arnold N, Blaum C, Gossling A, et al. C-reactive protein modifies lipoprotein(a)-related risk for coronary heart disease: the BiomarCaRE project. Eur Heart J. 2024 Mar 27;45(12):1043-54. doi:10.1093/eur-heartj/ehad867; Liu H, Fu D, Luo Y, Peng D. Independent association of Lp(a) with platelet reactivity in subjects without statins or antiplatelet agents. Sci Rep. 2022 Oct 5;12(1):16609. doi:10.1038/s41598-022-21121-7; Афанасьева ОИ, Пылаева ЕА, Клесарева ЕА идр. Липопротеид(а), аутоантитела к нему и циркулирующие субпопуляции Т-лимфоцитов как независимые факторы риска атеросклероза коронарных артерий. Терапевтический архив. 2016;88(9):31-8. doi:10.17116/terarkh201688931-38; Клесарева ЕА, Афанасьева ОИ, Кононова ЕВ идр. Повышенный титр IgM аутоантител к липопротеиду(a) как антиатерогенный фактор у пациентов с тяжелой гиперхолестеринемией. Российский кардиологический журнал. 2018;(8):13-20. doi:10.15829/1560-4071-2018-8-13-20; Van Buuren F, Horstkotte D, Knabbe C, et al. Incidence of elevated lipoprotein (a) levels in a large cohort of patients with cardiovascular disease. Clin Res Cardiol Suppl. 2017 Mar;12(Suppl 1):55-9. doi:10.1007/s11789-017-0087-y; Емельянчик ВС, Мариловцева ОВ, Хомченков РВ идр. Липопротеин (а) в диагностике сердечно-сосудистого риска. Значения липопротеина (а) и аполипопротеина Вво взрослой популяции г. Красноярска. Российский кардиологический журнал. 2023;28(7):5499. doi:10.15829/15604071-2023-5499; Nissen SE, Wolski K, Cho L, et al. Lipoprotein(a) levels in a global population with established atherosclerotic cardiovascular disease. Open Heart. 2022 Oct;9(2):e002060. doi:10.1136/openhrt-2022-002060; Patel AP, Wang M, Pirruccello JP, et al. Lp(a) (Lipoprotein[a]) Concentrations and Incident Atherosclerotic Cardiovascular Disease: New Insights From a Large National Biobank. Arterioscler Thromb Vasc Biol. 2021 Jan;41(1):465-74. doi:10.1161/ATVBA-HA.120.315291; Panza GA, Blazek O, Tortora J, et al. Prevalence of lipoprotein(a) measurement in patients with or at risk of cardiovascular disease. J Clin Lipidol. 2023 Nov-Dec;17(6):748-55. doi:10.1016/j.jacl.2023.09.016; Matveyenko A, Pavlyha M, Reyes-Soffer G. Supporting evidence for lipoprotein(a) measurements in clinical practice. Best Pract Res Clin Endocrinol Metab. 2023 May;37(3):101746. doi:10.1016/j.beem.2023.101746; Aggarwal HK, Jain D, Lathar M, et al. Lipoprotein-A and carotid intima media thickness as cardiovascular risk factors in patients of chronic kidney disease. Ren Fail. 2010 Jul;32(6):647-52. doi:10.3109/0886022X.2010.486097; Lin J, Reilly MP, Terembula K, Wilson FP. Plasma lipoprotein(a) levels are associated with mild renal impairment in type 2 diabetics independent of albuminuria. PLoS One. 2014 Dec 9;9(12):e114397. doi:10.1371/journal.pone.0114397; Albers JJ, Koschinsky ML, Marcovina SM. Evidence mounts for a role of the kidney in lipoprotein(a) catabolism. Kidney Int. 2007 May;71(10):961-2. doi:10.1038/sj.ki.5002240; Reblin T, Donarski N, Fineder L, et al. Renal handling of human apolipoprotein(a) and its fragments in the rat. Am J Kidney Dis. 2001 Sep;38(3):619-30. doi:10.1053/ajkd.2001.26889; Frank S, Hrzenjak A, Blaschitz A, et al. Role of various tissues in apo(a) fragmentation and excretion of fragments by the kidney. Eur J Clin Invest. 2001 Jun;31(6):504-12. doi:10.1046/j.1365-2362.2001.00811.x; Kovesdy CP, Astor BC, Longenecker JC, Coresh J. Association of kidney function with serum lipoprotein(a) level: the third National Health and Nutrition Examination Survey (1991-1994). Am J Kidney Dis. 2002 Nov;40(5):899-908. doi:10.1053/ajkd.2002.36319; Vaziri ND. Disorders of lipid metabolism in nephrotic syndrome: mechanisms and consequences. Kidney Int. 2016 Jul;90(1):41-52. doi:10.1016/j.kint.2016.02.026. Epub 2016 Apr 26.; Kronenberg F. Causes and consequences of lipoprotein(a) abnormalities in kidney disease. Clin Exp Nephrol. 2014 Apr;18(2):234-7. doi:10.1007/s10157-013-0875-8. Epub 2013 Oct 16.; De Sain-Van Der Velden MG, Reijngoud DJ, Kaysen GA, et al. Evidence for increased synthesis of lipoprotein(a) in the nephrotic syndrome. J Am Soc Nephrol. 1998 Aug;9(8):1474-81. doi:10.1681/ASN.V981474; Doucet C, Mooser V, Gonbert S, et al. Lipoprotein(a) in the nephrotic syndrome: molecular analysis of lipoprotein(a) and apolipoprotein(a) fragments in plasma and urine. J Am Soc Nephrol. 2000 Mar;11(3):507-13. doi:10.1681/ASN.V113507; Wang S, Zha L, Chen J, et al. The relationship between lipoprotein(a) and risk of cardiovascular disease: a Mendelian randomization analysis. Eur J Med Res.2022 Oct 27;27(1):211. doi:10.1186/s40001-022-00825-6; Willeit P, Ridker PM, Nestel PJ, et al. Baseline and on-statin treatment lipoprotein(a) levels for prediction of cardiovascular events: individual patient-data meta-analysis of statin outcome trials. Lancet. 2018 Oct 13;392(10155):1311-20. doi:10.1016/S0140-6736(18)31652-0. Epub 2018 Oct 4.; Nordestgaard BG, Langsted A. Lipoprotein (a) as a cause of cardiovascular disease: insights from epidemiology, genetics, and biology. J Lipid Res. 2016 Nov;57(11):1953-75. doi:10.1194/jlr.R071233. Epub 2016 Sep 27.; Тмоян НА, Афанасьева ОИ, Зотиков АЕ и др. Повышенный уровень липопротеида(а) как предиктор сердечно-сосудистых осложнений после реваскуляризации артерий нижних конечностей. Российский кардиологический журнал. 2018;(8):7-12. doi:10.15829/1560-4071-2018-8-7-12; Study Details. Assessing the Impact of Lipoprotein (a) Lowering With Pelacarsen (TQJ230) on Major Cardiovascular Events in Patients With CVD. ClinicalTrials.gov. Available at: https://clinicaltrials.gov/study/NCT04023552Test (accessed 04.01.2024).; Rubio-Serrano J, Gullon Ojesto A, Suarez Fernandez C. Clinical characteristics associated with elevated levels of lipoprotein(a) in patients with vascular risk. Adv Lab Med. 2023 Dec 1;4(4):396-401. doi:10.1515/almed-2023-0150; Cicero AFG, Fogacci F, Giovannini M, et al. Estimating the Prevalence and Characteristics of Patients Potentially Eligible for Lipoprotein(a)-Lowering Therapies in a Real-World Setting. Biomedicines. 2023 Dec 12;11(12):3289. doi:10.3390/biomedicines11123289; Gyabaah S, Adu-Boakye Y, Sarfo-Kantanka O, et al. Frequency & factors associated with elevated lipoprotein-a among Ghanaian stroke survivors. J Neurol Sci. 2024 Jan 15;456:122839. doi:10.1016/j.jns.2023.122839; Ziogos E, Vavuranakis MA, Harb T, et al. Lipoprotein(a) concentrations in acute myocardial infarction patients are not indicative of levels at six month follow-up. Eur Heart J Open. 2023 Apr 5;3(2):oead035. doi:10.1093/ehjopen/oead035; Myasoedova E, Crowson CS, Kremers HM, et al. Lipid paradox in rheumatoid arthritis: the impact of serum lipid measures and systemic inflammation on the risk of cardiovascular disease. Ann Rheum Dis. 2011 Mar;70(3):482-7. doi:10.1136/ard.2010.135871; Tsimikas S, Gordts PLSM, Nora C, et al. Statin therapy increases lipoprotein(a) levels. Eur Heart J.2020 Jun 21;41(24):2275-84. doi:10.1093/eurheartj/ehz310; Ward NC, Watts GF, Bishop W, et al. Australian Atherosclerosis Society Position Statement on Lipoprotein(a): Clinical and Implementation Recommendations. Heart Lung Circ. 2023 Mar;32(3):287-96. doi:10.1016/j.hlc.2022.11.015 . Epub 2023 Jan 25.; Zhang YQ, Duan YL, Sun JY. Study on the 5-year trend and influencing factors of lipoprotein (a) concentration in communitybased population. Zhonghua Xin Xue Guan Bing Za Zhi. 2023 Dec 24;51(12):1234-9. Chinese. doi:10.3760/cma.j.cn112148-20230817-00090; Simony SB, Mortensen MB, Langsted A, et al. Sex differences of lipoprotein(a) levels and associated risk of morbidity and mortality by age: The Copenhagen General Population Study. Atherosclerosis. 2022 Aug;355:76-82. doi:10.1016/j.atherosclerosis.2022.06.1023; Ежов МВ, Кухарчук ВВ, Сергиенко ИВ идр. Нарушения липидного обмена. Клинические рекомендации 2023. Российский карДиологический журнал. 2023;28(5):5471. doi:10.15829/156040712023-5471; Reyes-Soffer G, Yeang C, Michos ED, et al. High lipoprotein(a): Actionable strategies for risk assessment and mitigation. Am J Prev Cardiol. 2024 Apr 3;18:100651. doi:10.1016/j.ajpc.2024.100651; Smolders B, Lemmens R, Thijs V. Lipoprotein (a) and stroke: a meta-analysis of observational studies. Stroke. 2007 Jun;38(6):1959-66. doi:10.1161/STROKEA-HA.106.480657; Nave AH, Lange KS, Leonards CO, et al. Lipoprotein (a) as a risk factor for ischemic stroke: a meta-analysis. Atherosclerosis. 2015 Oct;242(2):496-503. doi:10.1016/j.atherosclerosis.2015.08.021; Kumar P, Swarnkar P, Misra S, Nath M. Lipoprotein (a) level as a risk factor for stroke and its subtype: A systematic review and metaanalysis. Sci Rep. 2021 Aug 2;11(1):15660. doi:10.1038/s41598-021-95141-0; Huang Y, Zhang R, Han L, et al. Lipoprotein(a) and stroke: a two-sample Mendelian randomization study. Front Aging Neurosci. 2023 May 12;15:1178079. doi:10.3389/fnagi.2023.1178079; Helgadottir A, Gretarsdottir S, Thorleifsson G, et al. Apolipoprotein(a) genetic sequence variants associated with systemic atherosclerosis and coronary atherosclerotic burden but not with venous thromboembolism. J Am Coll Cardiol. 2012 Aug 21;60(8):722-9. doi:10.1016/j.jacc.2012.01.078; Arnold M, Schweizer J, Nakas CT, et al. Lipoprotein(a) is associated with large artery atherosclerosis stroke aetiology and stroke recurrence among patients below the age of 60 years: results from the BIOSIGNAL study. Eur Heart J.2021 Jun 7;42(22):2186-96. doi:10.1093/eurheartj/ehab081; Pan Y, Li H, Wang Y, et al. Causal Effect of Lp(a) [Lipoprotein(a)] Level on Ischemic Stroke and Alzheimer Disease: A Mendelian Randomization Study. Stroke. 2019 Dec;50(12):3532-9. doi:10.1161/STROKEA-HA.119.026872; Lange KS, Nave AH, Liman TG, et al. Lipoprotein(a) Levels and Recurrent Vascular Events After First Ischemic Stroke. Stroke. 2017 Jan;48(1):36-42. doi:10.1161/STROKEA-HA.116.014436. Epub 2016 Nov 17.; Hong XW, Wu DM, Lu J, et al. Lipoprotein (a) as a Predictor of Early Stroke Recurrence in Acute Ischemic Stroke. Mol Neurobiol. 2018 Jan;55(1):718-26. doi:10.1007/s12035-016-0346-9. Epub 2016 Dec 21.; Zhang W, Zhang XA. Prognostic value of serum lipoprotein(a) levels in patients with acute ischemic stroke. Neuroreport. 2014 Mar 5;25(4):262-6. doi:10.1097/WNR.0000000000000094; Larsson SC, Gill D, Mason AM, et al. Lipoprotein(a) in Alzheimer, Atherosclerotic, Cerebrovascular, Thrombotic, and Valvular Disease: Mendelian Randomization Investigation. Circulation. 2020 Jun 2;141(22):1826-8. doi:10.1161/CIRCULA-TIONAHA.120.045826. Epub 2020 Jun 1.; Kunutsor SK, Khan H, Nyyss?nen K, Laukkanen JA. Is lipoprotein (a) protective of dementia? Eur J Epidemiol. 2016 Nov;31(11):1149-52. doi:10.1007/s10654-016-0184-0. Epub 2016 Jul 13.; Bhatia HS, Becker RC, Leibundgut G, et al. Lipoprotein(a), platelet function and cardiovascular disease. Nat Rev Cardiol. 2024 May;21(5):299-311. doi:10.1038/s41569-023-00947-2. Epub 2023 Nov 8.; Cui K, Wu S, Yin D, et al. Prolonged dual antiplatelet therapy in invasively treated acute coronary syndrome patients with different lipoprotein(a) concentrations. Cardiol J. 2024;31(1):32-44. doi:10.5603/cj.93062. Epub 2023 Oct 19.; Patel SM, Bonaca MP, Morrow DA, et al. Lipoprotein(a) and Benefit of Antiplatelet Therapy: Insights from the PEGASUS-TIMI 54 Trial. JACC Adv. 2023 Nov;2(9):100675. doi:10.1016/j.jacadv.2023.100675. Epub 2023 Oct 28.; Sukkari MH, Al-Bast B, Al Tamimi R, et al. Is there a benefit of aspirin therapy for primary prevention to reduce the risk of atherosclerotic cardiovascular disease in patients with elevated Lipoprotein (a)-A review of the evidence. Am J Prev Cardiol. 2023 Sep 1;15:100579. doi:10.1016/j.ajpc.2023.100579; Katsiki N, Vrablik M, Banach M, Gouni-Berthold I. Inclisiran, Low-Density Lipoprotein Cholesterol and Lipoprotein (a). Pharmaceuticals (Basel). 2023 Apr 12;16(4):577. doi:10.3390/ph16040577; Stoekenbroek RM. Inclisiran-mediated reductions in Lp(a) in the ORION-1 trial. Available at: https://academic.oup.com/eur-heartj/article/40/Supplement_1/ehz746.0015/5595089Test (accessed 11.03.2024).; Parish S, Hopewell JC, Hill MR, et al. Impact of Apolipoprotein(a) Isoform Size on Lipoprotein(a) Lowering in the HPS2-THRIVE Study. Circ Genom Precis Med. 2018 Feb;11(2):e001696. doi:10.1161/CIRC-GEN.117.001696; Koutsogianni AD, Liamis G, Liberopoulos E, et al. Effects of Lipid-Modifying and Other Drugs on Lipoprotein(a) Levels-Potent Clinical Implications. Pharmaceuticals (Basel). 2023 May 16;16(5):750. doi:10.3390/ph16050750; https://nnp.ima-press.net/nnp/article/view/2284Test

    الإتاحة: https://doi.org/10.14412/2074-2711-2024-3-136-14310.14412/2074-2711-2024-310.1016/S1474-4422Test(23)00277-610.26442/00403660.2023.01.2020310.1161/CIRCRESAHA.116.30839810.1093/eurheartj/ehab53210.1093/eurheartj/ehac36110.1161/ATV.000000000000014710.1155/2020/349176410.1172/JCI11309310.1016/j.atherosclerosis.2019.10.01110.1016/j.atherosclerosis.2022.04.01710.1016/j.amj-card.2020.03.04310.1161/JAHA.122.02670710.1111/eci.1417910.1093/eur-heartj/ehad86710.1038/s41598-022-21121-710.17116/terarkh201688931-3810.15829/1560-4071-2018-8-13-2010.1007/s11789-017-0087-y10.15829/15604071-2023-549910.1136/openhrt-2022-00206010.1161/ATVBA-HA.120.31529110.1016/j.jacl.2023.09.01610.1016/j.beem.2023.10174610.3109/0886022X.2010.48609710.1371/journal.pone.011439710.1038/sj.ki.500224010.1053/ajkd.2001.2688910.1046/j.1365-2362.2001.00811.x10.1053/ajkd.2002.3631910.1016/j.kint.2016.02.02610.1007/s10157-013-0875-810.1681/ASN.V98147410.1681/ASN.V11350710.1186/s40001-022-00825-610.1016/S0140-6736(18)31652-010.1194/jlr.R07123310.15829/1560-4071-2018-8-7-1210.1515/almed-2023-015010.3390/biomedicines1112328910.1093/ehjopen/oead03510.1136/ard.2010.13587110.1093/eurheartj/ehz31010.1016/j.hlc.2022.11.01510.3760/cma.j.cn112148-20230817-0009010.1016/j.atherosclerosis.2022.06.102310.15829/156040712023-547110.1016/j.ajpc.2024.10065110.1161/STROKEA-HA.106.48065710.1016/j.atherosclerosis.2015.08.02110.1038/s41598-021-95141-010.3389/fnagi.2023.117807910.1016/j.jacc.2012.01.07810.1093/eurheartj/ehab08110.1161/STROKEA-HA.119.02687210.1161/STROKEA-HA.116.01443610.1007/s12035-016-0346-910.1097/WNR.000000000000009410.1161/CIRCULA-TIONAHA.120.04582610.1038/s41569-023-00947-210.5603/cj.9306210.1016/j.jacadv.2023.10067510.1016/j.ajpc.2023.10057910.3390/ph1604057710.1161/CIRC-GEN.117.00169610.3390/ph16050750
    https://nnp.ima-press.net/nnp/article/view/2284Test

  3. 3
    دورية أكاديمية

    المصدر: SCIENTIFIC JOURNAL OF APPLIED AND MEDICAL SCIENCES; Vol. 3 No. 4 (2024): AMALIY VA TIBBIYOT FANLARI ILMIY JURNALI; 90-94 ; НАУЧНЫЙ ЖУРНАЛ ПРИКЛАДНЫХ И МЕДИЦИНСКИХ НАУК; Том 3 № 4 (2024): AMALIY VA TIBBIYOT FANLARI ILMIY JURNALI; 90-94 ; 2181-3469

    وصف الملف: application/pdf

  4. 4
    دورية أكاديمية

    المساهمون: Настоящее исследование выполнено в рамках фундаментальной темы № 1021051402790-6 «Изучение иммунопатологии, диагностики и терапии на ранних стадиях системных ревматических заболеваний».

    المصدر: Rheumatology Science and Practice; Vol 62, No 1 (2024); 81-89 ; Научно-практическая ревматология; Vol 62, No 1 (2024); 81-89 ; 1995-4492 ; 1995-4484

    وصف الملف: application/pdf

    العلاقة: https://rsp.mediar-press.net/rsp/article/view/3516/2351Test; England BR, Thiele GM, Anderson DR, Mikuls TR. Increased cardiovascular risk in rheumatoid arthritis: Mechanisms and implications. BMJ. 2018;23(361):1036. doi:10.1136/bmj.k1036; Balsa A, Lojo-Oliveira L, Alperi-López M, García-Manrique M, Ordóñez-Cañizares C, Pérez L, et al. Prevalence of comorbidities in rheumatoid arthritis and evaluation of their monitoring in clinical practice: The Spanish cohort of the COMORA study. Reumatol Clin (Engl Ed). 2019;15(2):102-108. doi:10.1016/j.reuma.2017.06.002; Насонов ЕЛ (ред.). Российские клинические рекомендации. Ревматология. М.:ГЭОТАР-Медиа;2020.; Baghdadi LR, Woodman RJ, Shanahan EM, Mangoni AA. The impact of traditional cardiovascular risk factors on cardiovascular outcomes in patients with rheumatoid arthritis: A systematic review and meta-analysis. PLoS One. 2015;10(2):0117952. doi:10.1371/journal.pone.0117952; Удачкина ЕВ, Новикова ДС, Попкова ТВ, Насонов ЕЛ. Роль интерлейкина 6 в развитии атеросклероза при ревматоидном артрите. Современная ревматология. 2013;7(3):25-32. doi:10.14412/1996-7012-2013-7; Eltoft A, Arntzen KA, Wilsgaard T, Mathiesen EB, Johnsen SH. Interleukin-6 is an independent predictor of progressive atherosclerosis in the carotid artery: The Tromsø Study. Atherosclerosis. 2018;271:1-8.; Lubrano V, Gabriele M, Puntoni MR, Longo V, Pucci L. Relationship among IL-6, LDL cholesterol and lipid peroxidation. Cell Mol Biol Lett. 2015;20(2):310-322. doi:10.1515/cmble-2015-0020; Barra LJ, Pope JE, Hitchon C, Boire G, Schieir O, Lin D, et al.; CATCH group. The effect of rheumatoid arthritis-associated autoantibodies on the incidence of cardiovascular events in a large inception cohort of early inflammatory arthritis. Rheumatology (Oxford). 2017;56(5):768-776. doi:10.1093/rheumatology/kew474; Насонов ЕЛ, Попкова ТВ. Атеросклероз: перспективы противовоспалительной терапии. Терапевтический архив. 2018;90(5):4-12. doi:10.26442/terarkh20189054-12; McInnes IB, Thompson L, Giles JT, Bathon JM, Salmon JE, Beaulieu AD, et al. Effect of interleukin-6 receptor blockade on surrogates of vascular risk in rheumatoid arthritis: MEASURE, a randomised, placebo-controlled study. Ann Rheum Dis. 2015;74(4):694-702. doi:10.1136/annrheumdis-2013-204345; Castagné B, Viprey M, Martin K, Schott A-M, Cucherat M, Soubrier M. Cardiovascular safety of tocilizumab: A systematic review and network meta-analysis. PLoS One. 2019;14(8):0220178. doi:10.1371/journal.pone.0220178; Jones G, Wallace T, McIntosh MJ, Brockwell L, Gómez-Reino JJ, Sebba A. Five-year efficacy and safety of tocilizumab monotherapy in patients with rheumatoid arthritis who were methotrexateand biologic-naive or free of methotrexate for 6 months: The AMBITION study. J Rheumatol. 2017;44(2):142-146. doi:10.3899/jrheum.160287; Насонов ЕЛ, Лила АМ. Ингибиция интерлейкина 6 при иммуновоспалительных ревматических заболеваниях: достижения, перспективы и надежды. Научно-практическая ревматология. 2017;55(6):590-599. doi:10.14412/1995-4484-2017-590-599; Комитет экспертов РКО. Национальные рекомендации по кардиоваскулярной профилактике. Кардиоваскулярная терапия и профилактика. 2011;10(6 Прил. 2.):46.; Герасимова ЕВ, Попкова ТВ, Мартынова АВ, Кириллова ИГ, Маркелова ЕИ, Семашко АС, и др. Динамика традиционных модифицируемых факторов риска, суммарного сердечно-сосудистого риска и структурных изменений сонных артерий у больных ревматоидным артритом на фоне длительной терапии ингибитором рецепторов интерлейкина-6. Научно-практическая ревматология. 2021;59(1):84-92. doi:10.47360/1995-4484-2021-84-92; Попкова ТВ, Новикова ДС, Панасюк ЕЮ, Авдеева АС, Удачкина ЕВ, Александрова ЕН, и др. Влияние тоцилизумаба на систему транспорта холестерина крови и ранние проявления атеросклероза у больных ревматоидным артритом. Терапевтический архив. 2012;84(5):9-18.; Gabay C, McInnes IB, Kavanaugh A, Tuckwell K, Klearman M, Pulley J, et al. Comparison of lipid and lipid-associated cardiovascular risk marker changes after treatment with tocilizumab or adalimumab in patients with rheumatoid arthritis. Ann Rheum Dis. 2016;75(10):1806-1812. doi:10.1136/annrheumdis-2015-207872; Toussirot E. The Interrelations between biological and targeted synthetic agents used in inflammatory joint diseases, and obesity or body composition. Metabolites. 2020;10(3):107. doi:10.3390/metabo10030107; Choi I, Sagawa A, Lee E, Lee EB, Song YW. Tocilizumab increases body weight and serum adipokine levels in patients with rheumatoid arthritis independently of their treatment response: A retrospective cohort study. J Korean Med Sci. 2020;35(22):e55. doi:10.3346/jkms.2020.35.e155; Tournadre A, Pereira B, Dutheil F, Giraud C, Courteix D, Sapin V, et al. Changes in body composition and metabolic profile during interleukin 6 inhibition in rheumatoid arthritis. J Cachexia Sarcopenia Muscle. 2017;8(4):639-646. doi:10.1002/jcsm.12189; Wada E, Tanihata J, Iwamura A, Takeda S, Hayashi YK, Matsuda R. Treatment with the anti-IL-6 receptor antibody attenuates muscular dystrophy via promoting skeletal muscle regeneration in dystrophin-/utrophin-deficient mice. Skelet Muscle. 2017;7(1):23. doi:10.1186/s13395-017-0140-z; Ando K, Takahashi F, Kato M, Kaneko N, Doi T, Ohe Y, et al. Tocilizumab, a proposed therapy for the cachexia of interleukin 6-expressing lung cancer. PLoS One. 2014;9(7):e102436. doi:10.1371/journal.pone.0102436; Genovese MC, McKay JD, Nasonov EL, Mysler EF, da Silva NA, Alecock E, et al. Interleukin-6 receptor inhibition with tocilizumab reduces disease activity in rheumatoid arthritis with inadequate response to disease-modifying antirheumatic drugs: The tocilizumab in combination with traditional disease-modifying antirheumatic drug therapy study. Arthritis Rheum. 2008;58(10):2968-2980. doi:10.1002/art.23940; Soubrier M, Pei J, Durand F, Gullestad L, John A. Concomitant use of statins in tocilizumab-treated patients with rheumatoid arthritis: A post hoc analysis. Rheumatol Ther. 2017;4(1):133-149. doi:10.1007/s40744-016-0049-8; Hoffman E, Rahat MA, Feld J, Elias M, Rosner I, Kaly L, et al. Effects of tocilizumab, an anti-interleukin-6 receptor antibody, on serum lipid and adipokine levels in patients with rheumatoid arthritis. Int J Mol Sci. 2019;20(18):4633. doi:10.3390/ijms20184633; Schiff MH, Kremer JM, Jahreis A, Vernon E, Isaacs JD, van Vollenhoven RF. Integrated safety in tocilizumab clinical trials. Arthritis Res Ther. 2011;13:141. doi:10.1186/ar3455; IL6R Genetics Consortium Emerging Risk Factors Collaboration; Sarwar N, Butterworth AS, Freitag DF, Gregson J, Willeit P, Gorman DN, et al. Interleukin-6 receptor pathways in coronary heart disease: a collaborative meta-analysis of 82 studies. Lancet. 2012; 379(9822):1205-1213. doi:10.1016/S0140-6736(11)61931-4; Navarro G, Taroumian S, Barroso N, Duan L, Furs D. Tocilizumab in rheumatoid arthritis: A meta-analysis of efficacy and selected clinical conundrums. Semin Arthritis Rheum. 2014;43(4):458-469. doi:10.1016/j.semarthrit.2013.08.001; Kitas GD, Nightingale P, Armitage J, Sattar N, Belch JJF, Symmons DP. A multicenter, randomized, placebo-controlled trial of atorvastatin for the primary prevention of cardiovascular events in patients with rheumatoid arthritis. Arthritis Rheumatol. 2019;71(9):1437-1449. doi:10.1002/art.40892; Abeles AM, Pillinge MH. Statins as antiinflammatory and immunomodulatory agents: A future in rheumatologic therapy? Arthritis Rheumatol. 2006;54(2):393-407. doi:10.1002/art.21521; Halacoglu J, Shea LA. Cardiovascular risk assessment and therapeutic implications in rheumatoid arthritis. J Cardiovasc Transl Res. 2020;13:878-90. doi:10.1007/s12265-020-09964-9; Tanaka T, Narazaki M, Ogata A, Kishimoto T. A new era for the treatment of inflammatory autoimmune diseases by interleukin-6 blockade strategy. Semin Immunol. 2014;26(1):88-96. doi:10.1016/j.smim.2014.01.009; Cacciapaglia F, Anelli MG, Rinaldi A, Fornaro M, Lopalco G, Scioscia C, et al. Lipids and atherogenic indices fluctuation in rheumatoid arthritis patients on long-term tocilizumab treatment. Mediators Inflamm. 2018;2018:2453265. doi:10.1155/2018/2453265; Kim SC, Solomon DH, Rogers JR, Gale S, Klearman M, Sarsour K, et al. Cardiovascular safety of tocilizumab versus tumor necrosis factor inhibitors in patients with rheumatoid arthritis: A multi-database cohort study. Arthritis Rheumatol. 2017;69(6):1154-1164. doi:10.1002/art.40084; Ferraz-Amaro I, Hernández-Hernández MV, Tejera-Segura B, Delgado-Frías E, Macía-Díaz M, Machado JD, et al. Effect of IL-6 receptor blockade on proprotein convertase subtilisin/kexin type9 and cholesterol efflux capacity in rheumatoid arthritis patients. Horm Metab Res. 2019;51(3):200-209. doi:10.1055/a-0833-4627; Strang AC, Bisoendial RJ, Kootte RS, Schulte DM, DallingaThie GM, Levels JH, et al. Pro-atherogenic lipid changes and decreased hepatic LDL receptor expression by tocilizumab in rheumatoid arthritis. Atherosclerosis. 2013;229(1):174-181. doi:10.1016/j.atherosclerosis.2013.04.031; Ruiz-Limón P, Ortega R, Arias de la Rosa I, Abalos-Aguilera MDC, Perez-Sanchez C, Jimenez-Gomez Y, et al. Tocilizumab improves the proatherothrombotic profile of rheumatoid arthritis patients modulating endothelial dysfunction, NETosis, and inflammation. Transl Res. 2017;183:87-103. doi:10.1016/j.trsl.2016.12.003; Bacchiega BC, Bacchiega AB, Usnayo MJG, Bedirian R, Singh G, Pinheiro G. Interleukin 6 inhibition and coronary artery disease in a high-risk population: A prospective community-based clinical study. J Am Heart Assoc. 2017;6(3):e005038. doi:10.1161/JAHA.116.005038; Ikonomidis I, Pavlidis G, Katsimbri P, Andreadou I, Triantafyllidi H, Tsoumani M, et al. Differential effects of inhibition of interleukin 1 and 6 on myocardial, coronary and vascular function. Clin Res Cardiol. 2019;108(10):1093-1101. doi:10.1007/s00392-019-01443-9; Kume K, Amano K, Yamada S, Hatta K, Ohta H, Kuwaba N. Tocilizumab monotherapy reduces arterial stiffness as effectively as An open-label randomized controlled trial. J Rheumatol. 2011;38(10):2169-2171. doi:10.3899/jrheum.110340; Chen DY, Chen YM, Hsieh TY, Hsieh CW, Lin CC, Lan JL. Significant effects of biologic therapy on lipid profiles and insulin resistance in patients with rheumatoid arthritis. Arthritis Res Ther. 2015;17(1):52. doi:10.1186/s13075-015-0559-8; Rao VU, Pavlov A, Klearman M, Musselman D, Giles JT, Bathon JM, et al. An evaluation of risk factors for major adverse cardiovascular events during tocilizumab therapy. Arthritis Rheumatol. 2015;67(2):372-380. doi:10.1002/art.38920; Okazaki S, Sakaguchi M, Miwa K, Furukado S, Yamagami H, Yagita Y, et al. Association of interleukin-6 with the progression of carotid atherosclerosis: A 9-year follow-up study. Stroke. 2014;45(10):2924-2929. doi:10.1161/STROKEAHA.114.005991; Puz P, Lasek-Bal A. Repeated measurements of serum concentrations of TNF-alpha, interleukin-6 and interleukin-10 in the evaluation of internal carotid artery stenosis progression. Atherosclerosis. 2017;263:97-103. doi:10.1016/j.atherosclerosis.2017.06.008; Pauli N, Puchałowicz K, Kuligowska A, Krzystolik A, Dziedziejko V, Safranow K, et al. Associations between IL-6 and echoparameters in patients with early onset coronary artery disease. Diagnostics (Basel). 2019;9(4):189. doi:10.3390/diagnostics9040189; Zhang J, Xie F, Yun H, Chen L, Muntner P, Levitan EB, et al. Comparative effects of biologics on cardiovascular risk among older patients with rheumatoid arthritis. Ann Rheum Dis. 2016;75(10):1813-1818. doi:10.1136/annrheumdis-2015-207870; Grange S, Schmitt C, Ganeshalingam K, Choy EH. Tocilizumab did not significantly increase serum cholesterol levels in healthy subjects. Rheumatology (Oxford). 2014;53:95-96. doi:10.1093/rheumatology/keu101.023; Specker C, Aringer M, Burmester GR, Killy B, Hofmann MW, Kellner H, et al. The safety and effectiveness of tocilizumab in elderly patients with rheumatoid arthritis and in patients with comorbidities associated with age. Clin Exp Rheumatol. 2022;40(9):1657-1665. doi:10.55563/clinexprheumatol/f7ff6q; Specker C, Alberding A, Aringer M, Burmester GR, Flacke JP, Hofmann MW, et al. ICHIBAN, a non-interventional study evaluating tocilizumab long-term effectiveness and safety in patients with active rheumatoid arthritis. Clin Exp Rheumatol. 2021;39(2):319-328. doi:10.55563/clinexprheumatol/3qdgi1; Jones G, Panova E. New insights and long-term safety of tocilizumab in rheumatoid arthritis. Ther Adv Musculoskelet Dis. 2018;10(10):195-199. doi:10.1177/1759720X18798462; Gale S, Trinh H, Tuckwell K, Collinson N, Stone JH, Sarsour K, et al. Adverse events in giant cell arteritis and rheumatoid arthritis patient populations: Analyses of tocilizumab clinical trials and claims data. Rheumatol Ther. 2019;6(1):77-88. doi:10.1007/s40744-019-0139-5; https://rsp.mediar-press.net/rsp/article/view/3516Test

  5. 5
    دورية أكاديمية

    المصدر: Acta Biomedica Scientifica; Том 9, № 1 (2024); 233-240 ; 2587-9596 ; 2541-9420

    وصف الملف: application/pdf

    العلاقة: https://www.actabiomedica.ru/jour/article/view/4617/2750Test; Collaborators GBDO, Afshin A, Forouzanfar MH, Reitsma MB, Sur P, Estep K, et al. Health effects of overweight and obesity in 195 countries over 25 years. N Engl J Med. 2017; 377: 13-27. doi:10.1056/NEJMoa1614362; Saklayen MG. The global epidemic of the metabolic syndrome. Curr Hypertens Rep. 2018; 20(2): 12. doi:10.1007/s11906-018-0812-z; Ляпина Л.А., Григорьева М.Е., Оберган Т.Ю., Мясоедов Н.Ф., Андреева Л.А. Коррекция проявлений экспериментального метаболического синдрома у крыс с помощью некоторых аргининсодержащих пептидов. Вестник Московского университета. Серия 16. Биология. 2017; 72(2): 92-98.; Lavallee KL, Zhang XC, Schneider S, Margraf J. Obesity and mental health: A longitudinal, cross-cultural examination in Germany and China. Front Psychol. 2021; 12: 712567.; Monserrat-Mesquida М, Quetglas-Llabres М, Capo X, Bouzas C, Mateos D, Pons A, et al. Metabolic syndrome is associated with oxidative stress and proinflammatory state. Antioxidants (Basel). 2020; 9(3): 236. doi:10.3390/antiox9030236; Panchal SK, Poudyal H, Iyer A, Nazer R, Alam A, Diwan V, et al. High-сarbohydrate high-fat diet-induced metabolic syndrome and cardiovascular remodeling in rats. J Cardiovasc Pharmacol. 2011; 57(1): 51-64. doi:10.1097/FJC.0b013e3181feb90a; Mulkey SB, du Plessis AJ. Autonomic nervous system development and its impact on neuropsychiatric outcome. Pediatr Res. 2019; 85(2): 120-126. doi:10.1038/s41390-018-0155-0; Sadock BJ, Sadock VA. Kaplan & Sadock’s comprehensive textbook of psychiatry. Philadelphia: Lippincott Williams & Wilkins Publishers; 2000.; Panossian A, Seo EJ, Efferth Т. Effects of anti-inflammatory and adaptogenic herbal extracts on gene expression of eicosanoids signaling pathways in isolated brain cells. Phytomedicine. 2019; (1): 1528-1581. doi:10.1016/j.phymed.2019.152881; Panossian AG, Efferth Т, Shikov AN, Pozharitskaya ON, Kuchta K, Mukherjee PK, et al. Evolution of the adaptogenic concept from traditional use to medical systems: Pharmacology of stress- and aging-related diseases. Med Res Rev. 2020; 20(4): 1-74. doi:10.1002/med.21743; Tian C, Hao L, Yi W, Ding S, Xu F. Polyphenols. Oxidative stress and metabolic syndrome. Gxid Med Cell Longev. 2020; 2020: 7398453. doi:10.1155/2020/7398453; Zhang Y, Scarpace PJ. The role of leptin in leptin resistance and obesity. Physiol Behav. 2006; 88: 249-256. doi:10.1016/j.physbeh.2006.05.038; Scarpace PJ, Matheny M, Kirichenko N, Gao YX, Tumer N, Zhang Y. Leptin overexpression in VTA trans-activates the hypothalamus whereas prolonged leptin action in either region crossdesensitizes. Neuropharmacology. 2013; 65: 90-100. doi:10.1016/j.neuropharm.2012.09.005; Перцов С.С., Алексеева И.В., Абрамова А.Ю., Никенина Е.В., Козлов А.Ю., Коплик Е.В., и др. Динамика метаболических показателей у крыс на разных стадиях постстрессорного периода в условиях антигенного воздействия при введении липополисахарида. Российский физиологический журнал им. И.М. Сеченова. 2021; 107(3): 321-331. doi:10.31857/S086981392103008; Pezze MA, Feldon J. Mesolimbic dopaminergic pathways in fear conditioning. Progr Neurobiol. 2004; 74: 301-320. doi:10.1016/j.pneurobio.2004.09.004; Marazziti D, Rutigliano G, Baroni S, Landi P, Dell’Osso L. Metabolic syndrome and major depression. CNS Spectr. 2014; 19(4): 293-304. doi:10.1017/S1092852913000667; Hasan MK, Mondal MSA, Kabir Y. Phytochemistry, pharmacological activity and potential health benefits of Glycyrrhiza glabra. Heliyon. 2021; 7(6): 07240. doi:10.1016/j.heliyon.2021. e07240; Zheng Y, Lee J, Lee EH, In G, Kim J, Lee MH, et al. A combination of Korean red ginseng extract and Glycyrrhiza glabra L. extract enhances their individual anti-obesity properties in 3T3- L1 adipocytes and C57BL/6J obese mice. J Med Food. 2020; 23(3): 215-223. doi:10.1002/med.21743; Li Z, Chen C, Zhu X, Li Y, Yu R, Xu W. Glycyrrhizin suppresses RANKL-Induced osteoclastogenesis and oxidative stress through inhibiting NF-kB and МАРК and activating AMPK/Nrf2. Calcif Tissue Int. 2018; 103(3): 324-327. doi:10.1007/s00223-018-0425-1; Lee OH, Kwon YI, Apostolidis E, Shetty K, Kim YC. Rhodiolainduced inhibition of adipogenesis involves antioxidant enzyme response associated with pentose phosphate pathway. Phytother Res. 2011; 25(1): 106-115. doi:10.1002/ptr.3236; https://www.actabiomedica.ru/jour/article/view/4617Test

  6. 6
    دورية أكاديمية
  7. 7
    دورية أكاديمية
  8. 8
    دورية أكاديمية

    وصف الملف: application/pdf

    العلاقة: Уральский медицинский журнал. 2023. Т. 22, № 3.; Чжен Т.Р. Клинические проявления функционально автономной секреции кортизола у больных с образованиями надпочечников / Т. Р. Чжен, Т. П. Киселева. – Текст: электронный // Уральский медицинский журнал. - 2023. – T. 22, № 3. – С. 13-21.; http://elib.usma.ru/handle/usma/14604Test

  9. 9
    دورية أكاديمية
  10. 10
    دورية أكاديمية

    المساهمون: The study was carried out with the financial support of Kazan State Medical University, grant from the International research board for young scientists (recipient — K.R. Salakhova, 2022), Работа выполнена при финансовой поддержке Казанского государственного медицинского университета, грант Международного научного совета для молодых ученых (получатель — Салахова К.Р., 2022)

    المصدر: Current Pediatrics; Том 22, № 3 (2023); 231-240 ; Вопросы современной педиатрии; Том 22, № 3 (2023); 231-240 ; 1682-5535 ; 1682-5527

    وصف الملف: application/pdf

    العلاقة: https://vsp.spr-journal.ru/jour/article/view/3223/1297Test; Cardiovascular diseases. In: World Health Organization: Official website. Available online: https://www.who.int/health-topics/cardiovascular-diseases#tab=tab_1Test. Accessed on April 12, 2023.; Маль Г.С., Смахтина А.М. Вторичная гиперлипидемия: определение, фенотипы и индуцирующие факторы // Международный журнал сердца и сосудистых заболеваний. — 2021. — Т. 9. — № 32. — С. 43–51. — doi: https://doi.org/10.24412/2311-16232021-32-43-51Test; Thongtang N, Sukmawan R, Llanes EJB, Lee ZV. Dyslipidemia management for primary prevention of cardiovascular events: Best in-clinic practices. Preventive Medicine Reports. 2022;27:a101819. doi: https://doi.org/10.1016/j.pmedr.2022.101819Test; Концевая А.В., Баланова Ю.А., Имаева А.Э. и др. Экономический ущерб от гиперхолестеринемии на популяционном уровне в Российской Федерации // Рациональная фармакотерапия в кардиологии. — 2018. — Т. 14. — № 3. — С. 393–401. — doi: https://doi.org/10.20996/1819-6446-2018-14-3-393401Test; Садыкова Д.И., Лутфуллин И.Я. Первичная артериальная гипертензия и гипертрофия миокарда в детском и подростковом возрасте // Педиатрия. Журнал им. Г.Н. Сперанского. — 2009. — Т. 88. — № 5. — С. 16–21.; Vaduganathan M, Mensah GA, Turco JV, et al. The global burden of cardiovascular diseases and risk: a compass for future health. J Am Coll Cardiol. 2022;80(25):2361–2371. doi: https://doi.org/10.1016/j.jacc.2022.11.005Test; Gujral J, Gupta J. Pediatric Dyslipidemia. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2022. Available online: https://www.ncbi.nlm.nih.gov/books/NBK585106Test. Accessed on July 28, 2022.; McGill HC Jr, McMahan CA, Malcom GT, et al. Effects of serum lipoproteins and smoking on atherosclerosis in young men and women. The PDAY Research Group. Pathobiological Determinants of Atherosclerosis in Youth. Arterioscler Thromb Vasc Biol. 1997; 17(1):95–106. doi: https://doi.org/10.1161/01.atv.17.1.95Test; Berenson GS, Srinivasan SR, Bao W, et al. Association between multiple cardiovascular risk factors and atherosclerosis in children and young adults. The Bogalusa Heart Study. N Engl J Med. 1998;338(23):1650–1656. doi: https://doi.org/10.1056/NEJM199806043382302Test; Ежов М.В., Бажан С.С., Ершова А. И. и др. Клинические рекомендации по семейной гиперхолестеринемии // Атеросклероз. — 2019. — Т. 15. — № 1. — С. 58–98.; Di Taranto MD, Fortunato G. Genetic heterogeneity of Familial hypercholesterolemia: repercussions for molecular diagnosis. Int J Mol Sci. 2023;24(4):3224. doi: https://doi.org/10.3390/ijms24043224Test; Beheshti SO, Madsen CM, Varbo A, Nordestgaard BG. Worldwide prevalence of Familial hypercholesterolemia: meta-analyses of 11 million subjects. J Am Coll Cardiol. 2020;75(20):2553–2566. doi: https://doi.org/10.1016/j.jacc.2020.03.057Test; Toft-Nielsen F, Emanuelsson F, Benn M. Familial hypercholesterolemia prevalence among ethnicities-systematic review and meta-analysis. Front Genet. 2022;13:840797. doi: https://doi.org/10.3389/fgene.2022.840797Test; Mainieri F, Tagi VM, Chiarelli F. Recent advances on Familial Hypercholesterolemia in children and adolescents. Biomedicines. 2022;10(5):1043. doi: https://doi.org/10.3390/biomedicines10051043Test; Wiegman A, Gidding SS, Watts GF, et al. Familial hyperc holesterolaemia in children and adolescents: gaining decades of life by optimizing detection and treatment. Eur Heart J. 2015;36(36): 2425–2437. doi: https://doi.org/10.1093/eurheartj/ehv157Test; Чубыкина У.В., Ежов М.В., Рожкова Т.А. и др. Пятилетний период наблюдения за пациентами с гомо- и гетерозиготной семейной гиперхолестеринемией в регистре РЕНЕССАНС // Атеросклероз и дислипидемии. — 2023. — № 1. — С. 5–18. — doi: https://doi.org/10.34687/2219-8202.JAD.2023.01.0001Test; Леонтьева И.В. Семейная гомозиготная гиперхолестеринемия у детей: стратегия ранней диагностики и лечения // Российский вестник перинатологии и педиатрии. — 2021. — Т. 66. — № 4. — С. 118–128. — doi: https://doi.org/10.21508/10274065-2021-66-4-118-128Test; Леонтьева И.В. Современная стратегия диагностики и лечения семейной гетерозиготной гиперхолестеринемии у детей // Российский вестник перинатологии и педиатрии. — 2020. — Т. 65. — № 4. — С. 27–40. — doi: https://doi.org/10.21508/1027-40652020-65-4-27-40Test; Исаева А.С., Исакова Е.А. Генетические причины нарушений липидного обмена // Norwegian Journal of Development of the International Science. — 2018. — № 17-2. — С. 34–39.; Berberich AJ, Hegele RA. A modern approach to dyslipidemia. Endocr Rev. 2022;43(4):611–653. doi: https://doi.org/10.1210/endrev/bnab037Test; Futema M, Taylor-Beadling A, Williams M, Humphries SE. Genetic testing for familial hypercholesterolemia-past, present, and future. J Lipid Res. 2021;62:100139. doi: https://doi.org/10.1016/j.jlr.2021.100139Test; Chora JR, Iacocca MA, Tichý L, et al. The clinical genome resource (clingen) familial hypercholesterolemia variant curation expert panel consensus guidelines for LDLR variant classification. Genet Med. 2022;24(2):293–306. doi: https://doi.org/10.1016/j.gim.2021.09.012Test; Futema M, Ramaswami U, Tichy L, et al. Comparison of the mutation spectrum and association with pre and post treatment lipid measures of children with heterozygous familial hypercholesterolaemia (FH) from eight European countries. Atherosclerosis. 2021;319:108–117. doi: https://doi.org/10.1016/j.atherosclerosis.2021.01.008Test; Abou Khalil Y, Marmontel O, Ferrières J, et al. APOE molecular spectrum in a French cohort with primary dyslipidemia. Int J Mol Sci. 2022;23(10):5792. doi: https://doi.org/10.3390/ijms23105792Test; Cao YX, Sun D, Liu HH, et al. Improvement of definite diagnosis of familial hypercholesterolemia using an expanding genetic analysis. JACC Asia. 2021;1(1):82–89. doi: https://doi.org/10.1016/j.jacasi.2021.04.001Test; Loaiza N, Hartgers ML, Reeskamp LF, et al. Taking one step back in familial hypercholesterolemia: STAP1 does not alter plasma LDL (Low-Density Lipoprotein) cholesterol in mice and humans. Arterioscler Thromb Vasc Biol. 2020;40(4):973–985. doi: https://doi.org/10.1161/ATVBAHA.119.313470Test; Tada H, Kawashiri MA, Nomura A, et al. Oligogenic familial hypercholesterolemia, LDL cholesterol, and coronary artery disease. J Clin Lipidol. 2018;(6):1436–1444. doi: https://doi.org/10.1016/j.jacl.2018.08.006Test; Nishikawa R, Furuhashi M, Hori M, et al. A resuscitated case of acute myocardial infarction with both familial hypercholesterolemia phenotype caused by possibly oligogenic variants of the PCSK9 and ABCG5 genes and type I CD36 deficiency. J Atheroscler Thromb. 2022; 29(4):551–557. doi: https://doi.org/10.5551/jat.58909Test; Paquette M, Fantino M, Bernard S, Baass A. Paternal inheritance predicts earlier cardiovascular event onset in patients with familial hypercholesterolemia. Atherosclerosis. 2021;329:9–13. doi: https://doi.org/10.1016/j.atherosclerosis.2021.06.006Test; Mourre F, Giorgi R, Gallo A, et al. Maternal inheritance of familial hypercholesterolemia gene mutation predisposes to coronary atherosclerosis as assessed by calcium score in adulthood. Arterioscler Thromb Vasc Biol. 2023;43(2):94–103. doi: https://doi.org/10.1161/ATVBAHA.122.318119Test; Abdul-Razak S, Rahmat R, Mohd Kasim A, et al. Diagnostic performance of various familial hypercholesterolaemia diagnostic criteria compared to Dutch lipid clinic criteria in an Asian population. BMC Cardiovasc Disord. 2017;17(1):264. doi: https://doi.org/10.1186/s12872-017-0694-zTest; Al-Rasadi K, Al-Waili K, Al-Sabti HA, et al. Criteria for Diagnosis of Familial Hypercholesterolemia: A Comprehensive Analysis of the Different Guidelines, Appraising their Suitability in the Omani Arab Population. Oman Med J. 2014;29(2):85–91. doi: https://doi.org/10.5001/omj.2014.22Test; Ежов М.В., Сергиенко И.В., Рожкова Т.А. и др. Российские рекомендации по диагностике и лечению семейной гиперхолестеринемии // Атеросклероз и дислипидемии. — 2016. — № 4. — С. 21–29.; Садыкова Д.И., Галимова Л.Ф., Леонтьева И.В., Сластникова Е.С. Оценка толщины комплекса интима-медиа у детей с семейной гиперхолестеринемией // Российский вестник перинатологии и педиатрии. — 2018. — Т. 63. — № 5. — С. 152–154. — doi: https://doi.org/10.21508/1027-4065-2018-63-5-152-154Test; Kusters DM, Wiegman A, Kastelein JJ, Hutten BA. Carotid intimamedia thickness in children with familial hypercholesterolemia. Circ Res. 2014;114(2):307–310. doi: https://doi.org/10.1161/CIRCRESAHA.114.301430Test; Pan J, Liu J, Wang H, et al. Association of carotid atherosclerosis with lipid components in asymptomatic low-income Chinese: a population-based cross-sectional study. Front Neurol. 2020;11: 276. doi: https://doi.org/10.3389/fneur.2020.00276Test; Gidding SS, Wiegman A, Groselj U, et al. Paediatric familial hypercholesterolaemia screening in Europe: public policy background and recommendations. Eur J Prev Cardiol. 2022;29(18): 2301–2311. doi: https://doi.org/10.1093/eurjpc/zwac200Test; Schefelker JM, Peterson AL. Screening and management of dyslipidemia in children and adolescents. J Clin Med. 2022; 11(21):6479. doi: https://doi.org/10.3390/jcm11216479Test; Šuštar U, Mlinarič M, Kovač J, et al. Universal screening is an effective strategy for detecting patients with Familial Hypercholesterolemia. Atherosclerosis. 2022;355:7. doi: https://doi.org/10.1016/j.atherosclerosis.2022.06.021Test; Alonso R, Perez de Isla L, Muñiz-Grijalvo O, Mata P. Barriers to early diagnosis and treatment of familial hypercholesterolemia: current perspectives on improving patient care. Vasc Health Risk Manag. 2020;16:11–25. doi: https://doi.org/10.2147/VHRM.S192401Test; Галимова Л.Ф., Садыкова Д.И., Сластникова Е.С., Усова Н.Э. Диагностика семейной гиперхолестеринемии у детей: каскадный скрининг от теории к практике // Кардиоваскулярная терапия и профилактика. — 2020. — Т. 19. — № 3. — С. 191–196. — doi: https://doi.org/10.15829/1728-8800-2020-2348Test; Groselj U, Kovac J, Sustar U, et al. Universal screening for familial hypercholesterolemia in children: The Slovenian model and literature review. Atherosclerosis. 2018;277:383–391. doi: https://doi.org/10.1016/j.atherosclerosis.2018.06.858Test; Medeiros AM, Bourbon M. Genetic testing in familial hypercholesterolemia: is it for everyone? Curr Atheroscler Rep. 2023;25(4): 127–132. doi: https://doi.org/10.1007/s11883-023-01091-5Test; Sanin V, Schmieder R, Ates S, et al. Population-based screening in children for early diagnosis and treatment of familial hypercholesterolemia: design of the VRONI study. Eur J Public Health. 2022; 32(3):422–428. doi: https://doi.org/10.1093/eurpub/ckac007Test; Lazaro P, Perez de Isla L, Watts GF, et al. Cost-effectiveness of a cascade screening program for the early detection of familial hypercholesterolemia. J Clin Lipidol. 2017;11(1):260–271. doi: https://doi.org/10.1016/j.jacl.2017.01.002Test; Louter L, Defesche J, Roeters van Lennep J. Cascade screening for familial hypercholesterolemia: Practical consequences. Atheroscler Suppl. 2017;30:77–85. doi: https://doi.org/10.1016/j.atherosclerosissup.2017.05.019Test; Vanhoye X, Bardel C, Rimbert A, et al. A new 165-SNP lowdensity lipoprotein cholesterol polygenic risk score based on next generation sequencing outperforms previously published scores in routine diagnostics of familial hypercholesterolemia. Transl Res. 2022;22:1931. doi: https://doi.org/10.1016/j.trsl.2022.12.002Test; Lima IR, Tada MT, Oliveira TGM, et al. Polygenic risk score for hypercholesterolemia in a Brazilian familial hypercholesterolemia cohort. Atheroscler Plus. 2022;49:47–55. doi: https://doi.org/10.1016/j.athplu.2022.06.002Test; Tandirerung FJ. The clinical importance of differentiating monogenic familial hypercholesterolemia from polygenic hypercholesterolemia. Curr Cardiol Rep. 2022;24(11):1669–1677. doi: https://doi.org/10.1007/s11886-022-01783-5Test; Martin AC, Gidding SS, Wiegman A, et al. Knowns and unknowns in the care of pediatric familial hypercholesterolemia. J Lipid Res. 2017;58(9):1765–1776. doi: https://doi.org/10.1194/jlr.S074039Test; Lampropoulou M, Chaini M, Rigopoulos R, et al. Association between serum lipid levels in Greek children with dyslipidemia and Mediterranean diet adherence, dietary habits, lifestyle and family socioeconomic factors. Nutrients. 2020;12(6):1–15. doi: https://doi.org/10.3390/nu12061600Test; Hui L, Kwok ME, Nelson AE, et al. Breastfeeding in infancy and lipid profile in adolescence. Pediatrics. 2019;143(5):1–8. doi: https://doi.org/10.1542/peds.2018-3075Test; Williams LA, Wilson DP. Nutritional management of pediatric dyslipidemia. In: Endotext [Internet]. South Dartmouth (MA): MDText. com, Inc.; 2000. Available online: https://www.ncbi.nlm.nih.gov/books/NBK395582Test. Accessed on November 01, 2022.; De Jesus JM. Expert panel on integrated guidelines for cardiovascular health and risk reduction in children and adolescents: summary report. Pediatrics. 2011;128(Suppl 5):S213–S256. doi: https://doi.org/10.1542/peds.2009-2107CTest; Fiorentino R, Chiarelli F. Treatment of dyslipidaemia in children. Biomedicines. 2021;9(9):1078. doi: https://doi.org/10.3390/biomedicines9091078Test; Yu-Poth S, Zhao G, Etherton T, et al. Effects of the National Cholesterol Education Program’s Step I and Step II dietary intervention programs on cardiovascular disease risk factors: a meta-analysis. Am J Clin Nutr. 1999;69(4):632–646. doi: https://doi.org/10.1093/ajcn/69.4.632Test; Simell O, Niinikoski H, Rönnemaa T, et al. Cohort Profile: the STRIP Study (Special Turku Coronary Risk Factor Intervention Project), an Infancy-onset Dietary and Life-style Intervention Trial. Int J Epidemiol. 2009;38(3):650–655. doi: https://doi.org/10.1093/ije/dyn072Test; Manousaki D, Barnett TA, Mathieu ME, et al. Tune out and turn in: the influence of television viewing and sleep on lipid profiles in children. Int J Obes (Lond). 2020;44(5):1173–1184. doi: https://doi.org/10.1038/s41366-020-0527-5Test; Langslet G, Johansen AK, Bogsrud MP, et al. Thirty percent of children and young adults with familial hypercholesterolemia treated with statins have adherence issues. Am J Prev Cardiol. 2021;6: 100180. doi: https://doi.org/10.1016/j.ajpc.2021.100180Test; Vuorio A, Kuoppala J, Kovanen PT, et al. Statins for children with familial hypercholesterolemia. Cochrane Database Syst Rev. 2019;11:CD006401. doi: https://doi.org/10.1002/14651858.CD006401.pub5Test; Tada H, Takamura M, Kawashiri M. Familial Hypercholesterolemia: a narrative review on diagnosis and management strategies for children and adolescents. Vasc Health Risk Manag. 2021;17: 59–67. doi: https://doi.org/10.2147/VHRM.S266249Test; Anagnostis P, Vaitsi K, Kleitsioti P, et al. Efficacy and safety of statin use in children and adolescents with familial hypercholesterolaemia: a systematic review and meta-analysis of randomized-controlled trials. Endocrine. 2020;69(2):249–261. doi: https://doi.org/10.1007/s12020-020-02302-8Test; Braamskamp M, Langslet G, McCrindle BW, et al. Effect of rosuvastatin on carotid intima-media thickness in children with heterozygous familial hypercholesterolemia: The CHARON study (Hypercholesterolemia in Children and Adolescents Taking Rosuvastatin Open Label). Circulation. 2017;136(4):359–366. doi: https://doi.org/10.1161/CIRCULATIONAHA.116.025158Test; Mytilinaiou M, Kyrou I, Khan M, et al. Familial Hypercholesterolemia: new horizons for diagnosis and effective management. Front Pharmacol. 2018;9:1–29. doi: https://doi.org/10.3389/fphar.2018.00707Test; Alonso R, Muñiz-Grijalvo O, Díaz-Díaz JL, et al. Efficacy of PCSK9 inhibitors in the treatment of heterozygous familial hypercholesterolemia: A clinical practice experience. J Clin Lipidol. 2021;15(4):584–592. doi: https://doi.org/10.1016/j.jacl.2021.04.011Test; Santos RD, Ruzza A, Hovingh GK, et al. Evolocumab in Pediatric Heterozygous Familial Hypercholesterolemia. N Engl J Med. 2020;383(14):1317–1327. doi: https://doi.org/10.1056/NEJMoa2019910Test; Ito MK, Sanros RD. PCSK9 Inhibition with monoclonal antibodies: modern management of hypercholesterolemia. J Clin Pharmacol. 2017;57(1):7–32. doi: https://doi.org/10.1002/jcph.766Test; Lee CJ, Yoon M, Kang HJ, et al. 2022 Consensus statement on the management of familial hypercholesterolemia in Korea. Korean J Intern Med. 2022;37(5):931–944. doi: https://doi.org/10.3904/kjim.2022.121Test; Erasmo L, Cefalu AB, Noto D, et al. Efficacy of lomitapide in the treatment of Familial Homozygous hypercholesterolemia: Results of a Real-World Clinical Experience in Italy. Adv Ther. 2017;34(5):1200–1210. doi: https://doi.org/10.1007/s12325017-0531-xTest; Ben-Omran T, Masana L, Kolovou G, et al. Real-World outcomes with lomitapide use in pediatric patients with homozygous familial hypercholesterolaemia. Adv Ther. 2019;36(7):1786–1811. doi: https://doi.org/10.1007/s12325-019-00985-8Test; Visser ME, Akdim F, Tribble DL, et al. Effect of apolipoprotein-B synthesis inhibition on liver triglyceride content in patients with familial hypercholesterolemia. J Lipid Res. 2010;51(5):1057–1062. doi: https://doi.org/10.1194/jlr.M002915Test; Raal FJ, Rosenson RS, Reeskamp LF, et al. Evinacumab for homozygous familial hypercholesterolemia. N Engl J Med. 2020;383(8):711–720. doi: https://doi.org/10.1056/NEJMoa2004215Test; Hermel M, Lieberman M, Slipczuk L, et al. Monoclonal antibodies, gene silencing and gene editing (CRISPR) therapies for the treatment of hyperlipidemia-the future is here. Pharmaceutics. 2023;15(2):459. doi: https://doi.org/10.3390/pharmaceutics15020459Test; Roch VZ, Santos R. Past, Present, and future of Familial Hypercholesterolemia management. Methodist Debakey Cardiovasc J. 2021;17(4):28–35. doi: https://doi.org/10.14797/mdcvj.887Test; Tomlinson B, Patil NG, Fok M, et al. Role of PCSK9 Inhibitors in patients with Familial Hypercholesterolemia. Endocrinol Metab (Seoul). 2021;36(2):279–295. doi: https://doi.org/10.3803/EnM.2021.964Test; Raal FJ, Kallend D, Ray KK, et al. Inclisiran for the treatment of heterozygous familial hypercholesterolemia. N Engl J Med. 2020:16;382(16):1520–1530. doi: https://doi.org/10.1056/NEJMoa1913805Test; Reijman MD, Schweizer A, Peterson ALH, et al. Rationale and design of two trials assessing the efficacy, safety, and tolerability of inclisiran in adolescents with homozygous and heterozygous familial hypercholesterolaemia. Eur J Prev Cardiol. 2022;29(9):1361–1368. doi: https://doi.org/10.1093/eurjpc/zwac025Test; Ishigaki Y, Kawagishi N, Hasegawa Y, et al. Liver transplantation for homozygous familial hypercholesterolemia. J Atheroscler Thromb. 2019;26(2):121–127. doi: https://doi.org/10.5551/jat.RV17029Test; Björkegren JLM, Lusis AJ. Atherosclerosis: Recent developments. Cell. 2022;185(10):1630–1645. doi: https://doi.org/10.1016/j.cell.2022.04.004Test; Berta E, Zsiros N, Bodor M, et al. Clinical aspects of genetic and non-genetic cardiovascular risk factors in familial hypercholesterolemia. Genes. 2022;13(7):1–19. doi: https://doi.org/10.3390/genes13071158Test; Braamskamp MJ, Kusters DM, Avis HJ, et al. Patients with familial hypercholesterolemia who initiated statin treatment in childhood are at lower risk for CHD then their affected parents. Circulation. 2013;128(Suppl 22):A17837.; Kesavan G. Innovations in CRISPR-Based Therapies. Mol Biotechnol. 2023;65(2):138–145. doi: https://doi.org/10.1007/s12033-021-00411-xTest; Asmamaw M, Zawdie B. Mechanism and applications of CRISPR/ Cas-9-Mediated Genome Editing. Biologics. 2021;15:353–361. doi: https://doi.org/10.2147/BTT.S326422Test; Alnouri F, Santos RD. New trends and therapies for familial hypercholesterolemia. J Clin Med. 2022;11(22):6638. doi: https://doi.org/10.3390/jcm11226638Test; Zhao H, Li Y, He L, et al. In Vivo AAV-CRISPR/Cas9-mediated gene editing ameliorates atherosclerosis in familial hypercholesterolemia. Circulation. 2020;141(1):67–79. doi: https://doi.org/10.1161/CIRCULATIONAHA.119.042476Test; Močnik M, Marčun Varda N. Lipid biomarkers and atherosclerosisold and new in cardiovascular risk in childhood. Int J Mol Sci. 2023; 24(3):2237. doi: https://doi.org/10.3390/ijms24032237Test; Fricaudet M, Di-Filippo, Mouli P, et al. Performance assessment of salivary screening in familial hypercholesterolemia in children. Atherosclerosis. 2021;331:E183. doi: https://doi.org/10.1016/j.atherosclerosis.2021.06.558Test; https://vsp.spr-journal.ru/jour/article/view/3223Test