Welcome to
Издательство Атмосфера

· Журналы
· Книги
· О проекте

·Электронные версии книг издательства “Атмосфера” поступили в продажу

Издательство “Атмосфера” идет в ногу со временем и открывает на своем сайте раздел электронных версий своих книг. Теперь вы можете приобрести не только традиционную бумажную книгу в картонном переплете, но и .pdf-файл, снабженный гиперссылками на каждую главу издания. В электронной версии проще отыскать рисунок и таблицу. Чтобы обратиться к ним, достаточно нажать на ссылку на каждый из них. Исчезла зависимость от тиража – вы можете приобрести даже те бестселлеры, тираж которых уже разошелся, такие как “Саркоидоз” или “Заболевания органов дыхания при беременности”. Упростилось получение вами книг – в течение двух рабочих дней после оплаты требуемые файлы придут на ваш e-mail. Ну и наконец, цена – электронные версии наших книг гораздо дешевле, чем бумажные издания. Заказывайте электронные версии книг издательства “Атмосфера” на сайте, а также по телефону: (495) 730-63-51 и по e-mail: atm-press2012@yandex.ru



ИНТЕРНЕТ-МАГАЗИН
Издательство Атмосфера


Телефон издательства

  
Лечебное дело
2023 / N 1

Диосмин и гесперидин: все ли эффекты нам известны?
А.С. Петриков

Список литературы/References

1. Gerges SH, Wahdan SA, Elsherbiny DA, El-Demerdash E. Pharmacology of diosmin, a citrus flavone glycoside: an updated review. European Journal of Drug Metabolism and Pharmacokinetics. 2022;47(1):1-18. DOI: 10.1007/s13318-021-00731-y
2. Huwait E, Mobashir M. Potential and therapeutic roles of diosmin in human diseases. Biomedicines. 2022;10(5):1076. DOI: 10.3390/biomedicines10051076
3. Тараховский Ю.С., Ким Ю.А., Абдрасилов Б.С., Музафаров Е.Н. Флавоноиды: биохимия, биофизика, медицина. Отв. ред. Маевский Е.И. Пущино: Sуnchrobook; 2013. 310 c. [Tarakhovskiy YuS, Kim YuA, Abdrasilov BS, Muzafarov EN. Flavonoids: biochemistry, biophysics, medicine. Mayevskiy EI, editor. Pushchino: Sunchrobook; 2013. 310 p. (In Russ.).]
4. Талибов О.Б. Диосмин в лечении венозной патологии: основы фармакокинетики и фармакодинамики. Хирургия. Журнал им. Н.И. Пирогова. 2019;3:135-140. [Talibov OB. Diosmin in treatment of venous syndrome: pharmacokinetics and pharmacodynamics. Pirogov Russian Journal of Surgery. 2019;3:135-140. (In Russ.).] DOI: 10.17116/hirurgia2019031135
5. Зверев Я.Ф. Флавоноиды глазами фармаколога. Особенности и проблемы фармакокинетики. Обзоры по клинической фармакологии и лекарственной терапии. 2017;15(2):4-11. [Zverev YaF. Flavonoids through the eyes of a pharmacologist. Features and problems of pharmacokinetics. Reviews of Clinical Pharmacology and Drug Therapy. 2017;15(2):4-11. (In Russ.).] DOI: 10.17816/RCF1524-11
6. Silvestro L, Tarcomnicu I, Dulea C, Attili NR, Ciuca V, Peru D, Rizea Savu S. Confirmation of diosmetin 3-O-glucuronide as major metabolite of diosmin in humans, using micro-liquid-chromatography-mass spectrometry and ion mobility mass spectrometry. Analytical and Bioanalytical Chemistry. 2013;405(25):8295-8310. DOI: 10.1007/s00216-013-7237-y
7. Garner RC, Garner JV, Gregory S, Calam A, Leong D. Comparison of the absorption of micronized (Daflon 500 mg) and nonmicronized 14C-diosmin tablets after oral administration to healthy volunteers by accelerator mass spectrometry and liquid scintillation counting. Journal of Pharmaceutical Sciences. 2002;91(1):32-40. DOI: 10.1002/jps.1168
8. Zhao M, Du L, Tao J, Qian D, Shang EX, Jiang S, Guo J, Liu P, Su SL, Duan JA. Determination of metabolites of diosmetin 7-O-glucoside by a newly isolated Escherichia coli from human gut using UPLC-Q-TOF/MS. Journal of Agricultural and Food Chemistry. 2014;62(47):11441-11448. DOI: 10.1021/jf502676j
9. Spanakis M, Kasmas S, Niopas I. Simultaneous determination of the flavonoid aglycones diosmetin and hesperetin in human plasma and urine by a validated GC/MS method: in vivo metabolic reduction of diosmetin to hesperetin. Biomedical Chromatography. 2009;23(2):124-131. DOI: 10.1002/bmc.1092
10. Zheng Y, Zhang R, Shi W, Li L, Liu H, Chen Z, Wu L. The metabolism and pharmacological activities of the natural health-benefiting compound diosmin. Food & Function. 2020;11(10):8472-8492. DOI: 10.1039/D0FO01598A
11. Li C, Schluesener H. Health-promoting effects of the citrus flavanone hesperidin. Critical Review in Food Science and Nutrition. 2017;57(3):613-631. DOI: 10.1080/10408398.2014.906382
12. Цуканов Ю.Т., Цуканов А.Ю., Баженов B.Н. Влияние оральных контрацептивов на диаметр магистральных вен нижних конечностей в ортостазе и его коррекция. Ангиология и сосудистая хирургия. 2008;14(1):77-78. [Tsukanov YuT, Tsukanov Ayu, Bazhenov BN. The effect of oral contraceptives on the orthostatic diameter of lower limbs major veins and its correction. Angiology and Vascular Surgery. 2008;14(1):77-78. (In Russ.).]
13. Воронков А. В., Гамзелева О.Ю. Обзор современных флеботропных препаратов на основе флавоноидов как перспективных эндотелиопротекторов при лечении хронических заболеваний вен. Амбулаторная хирургия. 2019;1-2:27-33. [Voronkov AV, Gamzeleva OYu. Review of modern phlebotropic drugs based on flavonoids as perspective endothelioprotectors in the treatment of chronic venous diseases. Ambulatory Surgery. 2019;1-2:27-33. (In Russ.).] DOI: 10.21518/1995-1477-2019-1-2-27-33
14. Martinez-Zapata MJ, Vernooij RWM, Simancas-Racines D, Uriona Tuma SM, Stein AT, Moreno Carriles RMM, Vargas E, Bonfill Cosp X. Phlebotonics for venous insufficiency. The Cochrane Database of Systematic Reviews. 2020;11(11):CD003229. DOI: 10.1002/14651858.CD003229.pub4
15. Ahmed S, Mundhe N, Borgohain M, Chowdhury L, Kwatra M, Bolshette N, Ahmed A, Lahkar M. Diosmin modulates the NF-kB signal transduction pathways and downregulation of various oxidative stress markers in alloxan-induced diabetic nephropathy. Inflammation. 2016;39(5):1783-1797. DOI: 10.1007/s10753-016-0413-4
16. Shalkami AS, Hassan M, Bakr AG. Anti-inflammatory, antioxidant and anti-apoptotic activity of diosmin in acetic acid-induced ulcerative colitis. Human & Experimental Toxicology. 2017;37(1):78-86. DOI: 10.1177/0960327117694075
17. Ali N, AlAsmari AF, Imam F, Ahmed MZ, Alqahtani F, Alharbi M, AlSwayyed M, AlAsmari F, Alasmari M, Alshammari A, Fantoukh OI, Alanazi MM. Protective effect of diosmin against doxorubicin-induced nephrotoxicity. Saudi Journal of Biological Sciences. 2021;28(8):4375-4383. DOI: 10.1016/j.sjbs.2021.04.030
18. Yao X, Gu X, Jin S, Shi K, Gao X, Wang Q, Zhao J, Zhang H, Lai X. Anticancer and anti-inflammatory effect of diosmin against Dalton ascitic lymphoma induced leukemia. Journal of Oleo Science. 2021;70(5):665-673. DOI: 10.5650/jos.ess21022
19. Imam F, Al-Harbi NO, Al-Harbi MM, Ansari MA, Zoheir KM, Iqbal M, Anwer MK, Al Hoshani AR, Attia SM, Ahmad SF. Diosmin downregulates the expression of T cell receptors, pro-inflammatory cytokines and NF-κB activation against LPS-induced acute lung injury in mice. Pharmacological Research. 2015;102:1-11. DOI: 10.1016/j.phrs.2015.09.001
20. Berkoz M. Diosmin suppresses the proinflammatory mediators in lipopolysaccharide-induced RAW264.7 macrophages via NF-κB and MAPKs signal pathways. General Physiology and Biophysics. 2019;38(4):315-324.
21. Feldo M, Woźniak M, Wójciak-Kosior M, Sowa I, Kot-Waśik A, Aszyk J, Bogucki J, Zubilewicz T, Bogucka-Kocka A. Influence of diosmin treatment on the level of oxidative stress markers in patients with chronic venous insufficiency. Oxidative Medicine and Cellular Longevity. 2018;2018:2561705. DOI: 10.1155/2018/2561705
22. Abdel-Salam OM, Youness ER, Mohammed NA, Abd-Elmoniem M, Omara E, Sleem AA. Neuroprotective and hepatoprotective effects of micronized purified flavonoid fraction (Daflon®) in lipopolysaccharide-treated rats. Drug Discoveries & Therapeutics. 2012;6(6):306-314. PMID: 23337818
23. Mustafa S, Akbar M, Khan MA, Sunita K, Parveen S, Pawar JS, Massey S, Agarwal NR, Husain SA. Plant metabolite diosmin as the therapeutic agent in human diseases. Current Research in Pharmacology and Drug Discovery. 2022;3:100122. DOI: 10.1016/j.crphar.2022.100122
24. Srinivasan S, Pari L. Ameliorative effect of diosmin, a citrus flavonoid against streptozotocin-nicotinamide generated oxidative stress induced diabetic rats. Chemico-Biological Interactions. 2012;195(1):43-51. DOI: 10.1016/j.cbi.2011.10.003
25. Chikara S, Nagaprashantha LD, Singhal J, Horne D, Awasthi S, Singhal SS. Oxidative stress and dietary phytochemicals: role in cancer chemoprevention and treatment. Cancer Letters. 2018;413:122-134. DOI: 10.1016/j.canlet.2017.11.002
26. Pfeffer PE, Lu H, Mann E.H, Chen YH, Ho TR, Cousins DJ, Corrigan C, Kelly FJ, Mudway IS, Hawrylowicz CM. Effects of vitamin D on inflammatory and oxidative stress responses of human bronchial epithelial cells exposed to particulate matter. PLoS One. 2018;13(8):e0200040. DOI: 10.1371/journal.pone.0200040
27. Zraika S, Hull RL, Udayasankar J, Aston-Mourney K, Subramanian S.L, Kisilevsky R, Szarek WA, Kahn SE. Oxidative stress is induced by islet amyloid formation and time-dependently mediates amyloid-induced beta cell apoptosis. Diabetologia. 2009;52(4):626-635. DOI: 10.1007/s00125-008-1255-x
28. Khosravi M, Poursaleh A, Ghasempour G, Farhad S, Najafi M. The effects of oxidative stress on the development of atherosclerosis. Biological Chemistry. 2019;400(6):711-732. DOI: 10.1515/hsz-2018-0397
29. Sepidarkish M, Farsi F, Akbari-Fakhrabadi M, Namazi N, Almasi-Hashiani A, Maleki Hagiagha A, Heshmati J. The effect of vitamin D supplementation on oxidative stress parameters: a systematic review and meta-analysis of clinical trials. Pharmacological Research. 2019;139:141-152. DOI: 10.1016/j.phrs.2018.11.011
30. Hallak M, Vazana L, Shpilberg O, Levy I, Mazar J, Nathan I. A molecular mechanism for mimosine-induced apoptosis involving oxidative stress and mitochondrial activation. Apoptosis. 2008;13(1):147-155. DOI: 10.1007/s10495-007-0156-7
31. Poznyak A, Grechko A.V, Poggio P, Myasoedova VA, Alfieri V, Orekhov AN. The diabetes mellitus-atherosclerosis connection: the role of lipid and glucose metabolism and chronic inflammation. International Journal of Molecular Sciences. 2020;21(5):1835. DOI: 10.3390/ijms21051835
32. Zhang Z, Liu Q, Yang J, Yao H, Fan R, Cao C, Liu C, Zhang S, Lei X, Xu S. The proteomic profiling of multiple tissue damage in chickens for a selenium deficiency biomarker discovery. Food & Function. 2020;11(2):1312-1321. DOI: 10.1039/c9fo02861g
33. Abdel-Reheim MA, Messiha BAS, Abo-Saif AA. Hepatoprotective effect of diosmin on iron-induced liver damage. International Journal of Pharmacology. 2017;13(6):529-540. DOI: 10.3923/ijp.2017.529.540
34. Perumal S, Langeshwaran K, Selvaraj J, Ponnulakshmi R, Shyamaladevi B, Balasubramanian MP. Effect of diosmin on apoptotic signaling molecules in N-nitrosodiethylamine-induced hepatocellular carcinoma in experimental rats. Molecular and Cellular Biology. 2018;449(1-2):27-37. DOI: 10.1007/s11010-018-3339-3
35. Pendeville H, Carpino N, Marine J.C, Takahashi Y, Muller M, Martial JA, Cleveland JL. The ornithine decarboxylase gene is essential for cell survival during early murine development. Molecular and Cellular Biology. 2001;21(19):6549-6558. DOI: 10.1128/MCB.21.19.6549-6558.2001
36. Tanaka T, Makita H, Kawabata K, Mori H, Kakumoto M, Satoh K, Hara A, Sumida T, Tanaka T, Ogawa H. Chemoprevention of azoxymethane-induced rat colon carcinogenesis by the naturally occurring flavonoids, diosmin and hesperidin. Carcinogenesis. 1997;18(5):957-965. DOI: 10.1093/carcin/18.5.957
37. Martínez C, Vicente V, Yáñez J, Alcaraz M, Castells MT, Canteras M, Benavente-García O, Castillo J. The effect of the flavonoid diosmin, grape seed extract and red wine on the pulmonary metastatic B16F10 melanoma. Histology and Histopathology. 2005;20(4):1121-1129. DOI: 10.14670/HH-20.1121
38. Álvarez N, Vicente V, Martínez C. Synergistic effect of diosmin and interferon-α on metastatic pulmonary melanoma. Cancer Biotherapy & Radiopharmaceuticals. 2009;24(3):347-352. DOI: 10.1089/cbr.2008.0565
39. Martínez Conesa C, Vicente Ortega V, Yáñez Gascón MJ, Alcaraz Baños M, Canteras Jordana M, Benavente-García O, Castillo J. Treatment of metastatic melanoma B16F10 by the flavonoids tangeretin, rutin, and diosmin. Journal of Agricultural and Food Chemistry. 2005;53(17):6791-6797. DOI: 10.1021/jf058050g
40. Dung TD, Chang HC, Chen CY, Peng WH, Tsai CH, Tsai FJ, Kuo WW, Chen LM, Huang CY. Diosmin induces cell apoptosis through protein phosphatase 2A activation in HA22T human hepatocellular carcinoma cells and blocks tumour growth in xenografted nude mice. Food Chemistry. 2012;132(4):2065-2073. DOI: 10.1016/j.foodchem.2011.11.149
41. Pari L, Srinivasan S. Antihyperglycemic effect of diosmin on hepatic key enzymes of carbohydrate metabolism in streptozotocin-nicotinamide-induced diabetic rats. Biomedicine & Pharmacotherapy. 2010;64(7):477-481. DOI: 10.1016/j.biopha.2010.02.001
42. Farmer JA. Diabetic dyslipidemia and atherosclerosis: evidence from clinical trials. Current Diabetes Reports. 2008;8:71-77. DOI: 10.1007/s11892-008-0013-2
43. Shepherd J. Does statin monotherapy address the multiple lipid abnormalities in type 2 diabetes? Atherosclerosis. Supplements. 2005;6(3):15-19. DOI: 10.1016/j.atherosclerosissup.2005.06.004
44. Jain D, Bansal MK, Dalvi R, Upganlawar A, Somani R. Protective effect of diosmin against diabetic neuropathy in experimental rats. Journal of Integrative Medicine. 2014 Jan;12(1):35-41. DOI: 10.1016/S2095-4964(14)60001-7
45. Eraslan G, Sarıca ZS, Bayram LÇ, Tekeli MY, Kanbur M, Karabacak M. The effects of diosmin on aflatoxin-induced liver and kidney damage. Environmental Science and Pollution Research International. 2017;24(36):27931-27941. DOI: 10.1007/s11356-011-0708-9
46. Bozdağ M, Eraslan G. The effect of diosmin against lead exposure in rats. Naunyn-Schmiedeberg’s Archives of Pharmacology. 2020;393(4):639-649. DOI: 10.1007/s00210-019-01758-4
47. Adouani I, Qureshi AS, Hang TJ. Preparation, evaluation and pharmacokinetics of diosmin herbosome in beagle dogs. Pakistan Journal of Pharmaceutical Sciences. 2019;33(1):33-40. PMID: 32122828
48. Fattori V, Rasquel-Oliveira FS, Artero NA, Ferraz CR, Borghi SM, Casagrande R, Verri WA Jr. Diosmin treats lipopolysaccharide-induced inflammatory pain and peritonitis by blocking NF-κB activation in mice. Journal of Natural Products. 2020;83(4):1018-1026. DOI: 10.1021/acs.jnatprod.9b00887
49. Zielinska DF, Gnad F, Schropp K, Wiśniewski JR, Mann M. Mapping N-glycosylation sites across seven evolutionarily distant species reveals a divergent substrate proteome despite a common core machinery. Molecular Cell. 2012;46(4):542-548. DOI: 10.1016/j.molcel.2012.04.031
50. Nabel EG. Cardiovascular disease. The New England Journal of Medicine. 2003;349(1):60-72. DOI: 10.1056/NEJMra035098
51. Luepker RV. Cardiovascular disease: rise, fall, and future prospects. Annual Review of Public Health. 2011;32:1-3. DOI: 10.1146/annurev-publhealth-112810-151726
52. Martín Giménez VM, Kassuha DE, Manucha W. Nanomedicine applied to cardiovascular diseases: latest developments. Therapeutic Advances in Cardiovascular Disease. 2017;11(4):133-142. DOI: 10.1177/1753944717692293
53. Iafisco M, Alogna A, Miragoli M, Catalucci D. Cardiovascular nanomedicine: the route ahead. Nanomedicine. (London, England). 2019;14(18):2391-2394. DOI: 10.2217/nnm-2019-0228
54. Paredes MD, Romecín P, Atucha NM, O’Valle F, Castillo J, Ortiz MC, García-Estañ J. Beneficial effects of different flavonoids on vascular and renal function in L-NAME hypertensive rats. Nutrients. 2018;10(4):484. DOI: 10.3390/nu10040484
55. Silambarasan T, Raja B. Diosmin, a bioflavonoid reverses alterations in blood pressure, nitric oxide, lipid peroxides and antioxidant status in DOCA-salt induced hypertensive rats. European Journal of Pharmacology. 2012;679(1-3):81-89. DOI: 10.1016/j.ejphar.2011.12.040
56. Queenthy SS, John B. Diosmin exhibits anti-hyperlipidemic effects in isoproterenol induced myocardial infarcted rats. European Journal of Pharmacology. 2013;718(1-2):213-218. DOI: 10.1016/j.ejphar.2013.08.031
57. Queenthy SS, Stanely Mainzen Prince P, John B. Diosmin prevents isoproterenol-induced heart mitochondrial oxidative stress in rats. Cardiovascular Toxicology. 2018;18(2):120-130. DOI: 10.1007/s12012-017-9422-2
58. Sabarimuthu SQ, Ponnian SMP, John B. Diosmin prevents left ventricular hypertrophy, adenosine triphosphatases dysfunction and electrolyte imbalance in experimentally induced myocardial infarcted rats. European Journal of Pharmacology. 2017;814:124-129. DOI: 10.1016/j.ejphar.2017.07.049
59. El-Fawal R, El Fayoumi HM, Mahmoud MF. Mahmoud. Effects of diosmin and crocin on metabolic syndrome-associated cardio-vascular complications in rats. Naunyn-Schmiedeberg’s Archives of Pharmacology. 2019;392(12):1523-1536. DOI: 10.1007/s00210-019-01700-8
60. Rizza S, Muniyappa R, Iantorno M, Kim JA, Chen H, Pullikotil P, Senese N, Tesauro M, Lauro D, Cardillo C, Quon MJ. Citrus polyphenol hesperidin stimulates production of nitric oxide in endothelial cells while improving endothelial function and reducing inflammatory markers in patients with metabolic syndrome. The Journal of Clinical Endocrinology & Metabolism. 2011;96(5):E782-E792. DOI: 10.1210/jc.2010-2879
61. Morand C, Dubray C, Milenkovic D, Lioger D, Martin JF, Scalbert A, Mazur A. Hesperidin contributes to the vascular protective effects of orange juice: a randomized crossover study in healthy volunteers. The American Journal of Clinical Nutrition. 2011;93(1):73-80. DOI: 10.3945/ajcn.110.004945
62. Demonty I, Lin Y, Zebregs YE, Vermeer MA, van der Knaap HC, Jäkel M, Trautwein EA. The citrus flavonoids hesperidin and naringin do not affect serum cholesterol in moderately hypercholesterolemic men and women. Journal of Nutrition. 2010;140(9):1615-1620. DOI: 10.3945/jn.110.124735
63. Mahmoud AM, Ashour MB, Abdel-Moneim A, Ahmed O. Hesperidin and naringin attenuate hyperglycemia-mediated oxidative stress and proinflammatory cytokine production in high fat fed/streptozotocin-induced type 2 diabetic rats. Journal of Diabetes and its Complications. 2012;26(6):483-490. DOI: 10.1016/j.jdiacomp.2012.06.001
64. Akiyama S, Katsumata S, Suzuki K, Ishimi Y, Wu J, Uehara M. Dietary hesperidin exerts hypoglycemic and hypolipidemic effects in streptozotocin-induced marginal type 1 diabetic rats. Journal of Clinical Biochemistry & Nutrition. 2012;46(1):87-92. DOI: 10.3164/jcbn.09-8
65. Gandhi C, Upaganalawar A, Balaraman R. Protection against in vivo focal myocardial ischemia/reperfusion injury-induced arrhythmias and apoptosis by hesperidin. Free Radical Research. 2009;43(9):817-827. DOI: 10.1080/10715760903071656
66. Selvara P, Pugalend KV. Hesperidin, a flavanone glycoside, on lipid peroxidation and antioxidant status in experimental myocardial ischemic rats. Redox Report. 2010;15(5):217-223. DOI: 10.1179/135100010X12826446921509
67. Yamamoto M, Jokura H, Hashizume K, Ominami H, Shibuya Y, Suzuki A, Hase T, Shimotoyodome A. Hesperidin metabolite hesperetin-7-O-glucuronide, but not hesperetin-3’-O-glucuronide, exerts hypotensive, vasodilatory, and anti-inflammatory activities. Food & Function. 2016;7(1):118-126. DOI: 10.1039/c5fo00771b
68. Yamamoto M, Suzuki A, Jokura H, Yamamoto N, Hase T. Glucosyl hesperidin prevents endothelial dysfunction and oxidative stress in spontaneously hypertensive rats. Nutrition. 2008;24(5):470-476. DOI: 10.1016/j.nut.2008.01.010
69. Ohtsuki K, Abe A, Mitsuzumi H, Kondo M, Uemura K, Iwasaki Y, Kondo Y. Glucosyl hesperidin improves serum cholesterol composition and inhibits hypertrophy in vasculature. Journal of Nutritional Sciences and Vitaminology. 2003;49(6):447-450. DOI: 10.3177/jnsv.49.447
70. Akiyama S, Katsumata S, Suzuki K, Nakaya Y, Ishimi Y, Uehara M. Hypoglycemic and hypolipidemic effects of hesperidin and cyclodextrin-clathrated hesperetin in Goto-Kakizaki rats with type 2 diabetes. Bioscience, Biotechnology & Biochemistry. 2009;73(12):2779-2782. DOI: 10.1271/bbb.90576
71. Kumar B, Gupta SK, Srinivasan BP, Nag TC, Srivastava S, Saxena R, Jha KA. Hesperetin rescues retinal oxidative stress, neuroinflammation and apoptosis in diabetic rats. Microvascular Research. 2013;87:65-74. DOI: 10.1016/j.mvr.2013.01.002
72. Wang DM, Li SQ, Zhu XY, Wang Y, Wu WL, Zhang XJ. Protective effects of hesperidin against amyloid-beta (Abeta) induced neurotoxicity through the voltage dependent anion channel 1 (VDAC1)-mediated mitochondrial apoptotic pathway in PC12 cells. Neurochemical Research. 2013;38(5):1034-1044. DOI: 10.1007/s11064-013-1013-4
73. Tamilselvam K, Braidy N, Manivasagam T, Essa MM, Prasad NR, Karthikeyan S, Thenmozhi AJ, Selvaraju S, Guillemin GJ. Neuroprotective effects of hesperidin, a plant flavanone, on rotenone-induced oxidative stress and apoptosis in a cellular model for Parkinson’s disease. Oxidative Medicine and Cellular Longevity. 2013;2013:102741. DOI: 10.1155/2013/102741
74. Souza LC, de Gomes MG, Goes AT, Del Fabbro L, Filho CB, Boeira SP, Jesse CR. Evidence for the involvement of the serotonergic 5-HT(1A) receptors in the antidepressant-like effect caused by hesperidin in mice. Progress in Neuro-Psychopharmacology & Biological Psychiatry. 2013;40:103-109. DOI: 10.1016/j.pnpbp.2012.09.003
75. Nandakumar N, Balasubramanian MP. Hesperidin protects renal and hepatic tissues against free radical-mediated oxidative stress during DMBA-induced experimental breast cancer. Journal of Environmental Pathology, Toxicology and Oncology. 2011;30(4):283-300. DOI: 10.1615/jenvironpatholtoxicoloncol.v30.i4.20
76. Saiprasad G, Chitra P, Manikandan R, Sudhandiran G. Hesperidin alleviates oxidative stress and downregulates the expressions of proliferative and inflammatory markers in azoxymethane-induced experimental colon carcinogenesis in mice. Inflammation Research. 2013;62(4):425-440. DOI: 10.1007/s00011-013-0595-2
77. Dong W, Wei X, Zhang F, Hao J, Huang F, Zhang C, Liang W. A dual character of flavonoids in influenza A virus replication and spread through modulating cell-autonomous immunity by MAPK signaling pathways. Science Reports. 2014;4:7237. DOI: 10.1038/srep07237
78. Russo M, Moccia S, Spagnuolo C, Tedesco I, Russo GL. Roles of flavonoids against coronavirus infection. Chemico-Biological Interactions. 2020;328:109211. DOI: 10.1016/j.cbi.2020.109211
79. Haggag YA, El-Ashmawy NE, Okasha KM. Is hesperidin essential for prophylaxis and treatment of COVID-19 infection? Medical Hypotheses. 2020;144:109957. DOI: 10.1016/j.mehy.2020.109957
80. Guy RK, DiPaola RS, Romanelli F, Dutch RE. Rapid repurposing of drugs for COVID-19. Science (New York, N.Y.). 2020;368(6493):829-830. DOI: 10.1126/science.abb9332
81. Magro G. COVID-19: review on latest available drugs and therapies against SARS-CoV-2. Coagulation and inflammation cross-talking. Virus Research. 2020;286:198070. DOI: 10.1016/j.virusres.2020.198070
82. Johnson RM, Vinetz JM. Dexamethasone in the management of COVID-19. BMJ. 2020;370:m2648. DOI: 10.1136/bmj.m2648
83. Debiaggi M, Tateo F, Pagani L, Luini M, Romero E. Effects of propolis flavonoids on virus infectivity and replication. Microbiologica. 1990;13(3):207-213. PMID: 2125682
84. Huang L, Shi Y, Gong B, Jiang L, Liu X, Yang J, Tang J, You C, Jiang Q, Long B, Zeng T, Luo M, Zeng F, Zeng F, Wang S, Yang X, Yang Z. Blood single cell immune profiling reveals the interferon-MAPK pathway mediated adaptive immune response for COVID-19. medRxiv. 2020. Posted 2020 Mar 17. DOI: 2020.2003.2015.20033472
85. Wu C, Liu Y, Yang Y, Zhang P, Zhong W, Wang Y, Wang Q, Xu Y, Li M, Li X, Zheng M, Chen L, Li H. Analysis of therapeutic targets for SARS-CoV-2 and discovery of potential drugs by computational methods. Acta Pharmaseutica Sinica B. 2020;10(5):766-788. DOI: 10.1016/j.apsb.2020.02.008
86. Российские клинические рекомендации по диагностике и лечению хронических заболеваний вен. Флебология. 2018;12(3):146-240. [Russian clinical guidelines for the diagnosis and treatment of chronic venous diseases. Phlebologiya. 2018;12(3):146-240. (In Russ.).] DOI: 10.17116/flebo20187031146
87. Rabe E, Guex J, Puskas A, Scuderi A, Fernandez Quesada F; VCP Coordinators. Epidemiology of chronic venous disorders in geographically diverse populations: results from the Vein Consult Program. International Angiology. 2012;31(2):105-115. PMID: 22466974
88. Criqui M, Jamosmos M, Fronek A, Denenberg J, Langer R, Bergan J, Golomb B. Chronic venous disease in an ethnically diverse population: the San Diego population study. American Journal of Epidemiology. 2003;158(5):448-456. DOI: 10.1093/aje/kwg166
89. Камаев А.А., Булатов В.Л., Вахратьян П.Е., Волков А.М., Волков А.С., Гаврилов Е.К., Головина В.И., Ефремова О.И., Иванов О.О., Илюхин Е.А., Каторкин С.Е., Кончугова Т.В., Кравцов П.Ф., Максимов С.В., Мжаванадзе Н.Д., Пиханова Ж.М., Прядко С.И., Смирнов А.А., Сушков С.А., Чаббаров Р.Г., Шиманко А.И., Якушкин С.Н., Апханова Т.В., Деркачев С.Н., Золотухин И.А., Калинин Р.Е., Кириенко А.И., Кульчицкая Д.Б., Пелевин А.В., Петриков А.С., Рачин А.П., Селиверстов Е.И., Стойко Ю.М., Сучков И.А. Клинические рекомендации. Варикозное расширение вен. Флебология. 2022;16(1):41-108. [Kamayev AA, Bulatov VL, Vakhratyan PE, Volkov AM, Volkov AS, Gavrilov EK, Golovina VI, Yefremova OI, Ivanov OO, Ilyukhin EA, Katorkin SE, Konchugova TV, Kravtsov PF, Maksimov SV, Mzhavanadze N.D, Pikhanova ZhM, Pryadko SI, Smirnov AA, Sushkov SA, Chabbarov RG, Shimanko AI, Yakushkin SN, Apkhanova TV, Derkachev SN, Zolotukhin IA, Kalinin RE, Kiriyenko AI, Kulchitskaya DB, Pelevin AV, Petrikov AS, Rachin AP, Seliverstov EI, Stoyko YuM, Suchkov IA. Clinical recommendations. Varicose veins. Phlebologiya. 2022;16(1):41-108. (In Russ.).] DOI: 10.17116/flebo20221601141
90. Шевченко Ю.Л., Стойко Ю.М., Гудымович В.Г., Никитина А.М., Трифонов С.И. Дисфункции эндотелия у больных варикозной болезнью нижних конечностей и ее возможные коррекции. Ангиология и сосудистая хирургия. 2010;16(4):99-107. [Shevchenko YuL, Stoyko YuM, Gudymovich VG, Nikitina AM, Trifonov SI. Endothelial dysfunction in patients with lower limb varicose disease and possibilities correction thereof. Angiology and Vascular Surgery. 2010;16(4):99-107. (In Russ.).]
91. Стойко Ю.М., Гудымович В.Г. Мониторинг качества жизни у больных варикозной болезнью вен нижних конечностей с использованием препарата венарус. Хирургия. Журнал им. Н.И. Пирогова. 2010;6:46-51. [Stoyko YuM, Gudymovich VG. Monitoring the quality of life in patients with varicose disease of the lower extremities using venarus drug. Pirogov Russian Journal of Surgery. 2010;6:46-51. (In Russ.).]
92. Гудымович В.Г., Стойко Ю.М., Яковлева Н.М., Никитина А.М. Флеботропная терапия препаратом венарус у больных с хронической венозной недостаточностью нижних конечностей. Ангиология и сосудистая хирургия. 2013;19(4):88-91. [Gudymovich VG, Stoyko YuM, Yakovleva NM, Nikitina AM. Phlebotrophic therapy with venarus in patients suffering from lower limb chronic venous insufficiency. Angiology and Vascular Surgery. 2013;19(4):88-91. (In Russ.).]
93. Eberhardt RT, Raffetto JD. Chronic venous insufficiency. Circulation. 2014;130(4):333-346. DOI: 10.1161/CIRCULATIONAHA.113.006898
94. Гурфинкель Ю.И., Сасонко М.Л., Талов Н.А. Коррекция параметров микроциркуляции крови и функции эндотелия при хронической венозной недостаточности нижних конечностей. Ангиология и сосудистая хирургия. 2017;23(2):89-95. [Gurfinkel YuI, Sasonko ML, Talov NA. Correction of blood microcirculation parameters and endothelial function in chronic venous insufficiency of lower limbs. Angiology and Vascular Surgery. 2017;23(2):89-95. (In Russ.).]
95. Калинин Р.Е., Сучков И.А., Камаев А.А., Звягина В.И., Крылов А.А. Эндотелиотропные эффекты венотонизирующих препаратов при лечении больных с варикозной болезнью. Ангиология и сосудистая хирургия. 2018;24(4):72-74. [Kalinin RE, Suchkov IA, Kamayev AA, Zvyagina VI, Krylov AA. Endotheliotropic effects of venotonic drugs in treatment of patients with varicose veins. Angiology and Vascular Surgery. 2018;24(4):72-74. (In Russ.).]
96. Роднянский Д.В., Фокин А.А. Диосминсодержащие флеботропные препараты при варикозной экземе. Ангиология и сосудистая хирургия. 2019;25(3):88-92. [Rodnyanskiy DV, Fokin AA. Diosmin-containing phlebotropic drugs in varicose eczema. Angiology and Vascular Surgery. 2019;25(3):88-92. (In Russ.).] DOI: 10.33529/ANGI02019303
97. Сонькин И.Н., Шайков Е.В., Крылов Д.В., Булатов В.Л., Ремизова А.С., Резванцев М.В. Эффективность применения препарата венарус в лечении больных с посттромботической болезнью нижних конечностей. Ангиология и сосудистая хирургия. 2014;20(4):77-83. [Sonkin IN, Shaykov EV, Krylov DV, Bulatov VL, Remizova AS, Rezvantsev MV. Efficacy of venarus in treatment of patients with post-thrombotic syndrome of lower limbs. Angiology and Vascular Surgery. 2014;20(4):77-83. (In Russ.).]
98. Игнатьев И.М. Открытое проспективное рандомизированное исследование результатов применения препарата венарус при посттромботической болезни. Ангиология и сосудистая хирургия. 2018;24(1):97-101. [Ignatyev IM. Open prospective randomized study of the results of using venarus in post-thrombotic syndrome. Angiology and Vascular Surgery. 2018;24(1):97-101. (In Russ.).] PMID: 29688200
99. Петриков А.С., Дудин Д.В., Попкова Л.Н. Развитие посттромботической болезни на фоне пролонгированной антитромботической терапии с учетом содержания маркеров воспаления, гемостаза и эндотелиальной дисфункции. Флебология. 2020;14(4):275 284. [Petrikov AS, Dudin DV, Popkova LN. Connection of post-thrombotic syndrome development with markers of inflammation, coagulation, and endothelial dysfunction in patients on prolonged anticoagulation. Phlebologiya. 2020;14(4):275-284. (In Russ.).] DOI: 10.17116/flebo202014041275
100. Петриков А.С., Дудин Д.В., Дронов С.В., Эйрих В.Р., Шойхет Я.Н. Прогресс реканализации глубоких вен нижних конечностей на фоне пролонгированной антитромботической терапии и содержание D-димеров, C-реактивного белка и гомоцистеина. Флебология. 2020;14(3):196 204. [Petrikov AS, Dudin DV, Dronov SV, Eyrikh VR, Shoykhet YaN. Lower extremity deep vein recanalization following prolonged antithrombotic therapy and D-dimer, C-reactive protein and homocysteine levels. Phlebologiya. 2020;14(3):196-204. (In Russ.).] DOI: 10.17116/flebo202014031196
101. Лобастов К.В., Счастливцев И.В., Барганджия А.Б. Риск развития посттромботической болезни при использовании прямых оральных антикоагулянтов: систематический обзор литературы и мета-анализ. Хирургия. Журнал им. Н.И. Пирогова. 2022;2:89 99. [Lobastov KV, Schastlivtsev IV, Bargandgiya AB. Risk of post-thrombotic syndrome following direct oral anticoagulant intake: a systematic review and meta-analysis. Pirogov Russian Journal of Surgery. 2022;2:89-99. (In Russ.).] DOI: 10.17116/hirurgia202202189
102. Сонькин И.Н., Крылов Д.В., Мельник В.Ю., Атабеков А.И. Выраженность проявлений посттромботической болезни после применения различных вариантов консервативного лечения тромбоза глубоких вен нижних конечностей. Флебология. 2018;12(3):118-122. [Sonkin IN, Krylov DV, Melnik VYu, Atabekov A.I. The severity of manifestations of the post-thrombotic syndrome following different variants of conservative treatment of deep vein thrombosis in the lower extremities. Phlebologiya. 2018;12(3):118-122. (In Russ.).]
103. Зудин А.М, Засорина М.А, Вихерт Т.А, Гонсалес А.К, Тарковский А.А. Ультразвуковая оценка изменений венозной гемодинамики у больных с посттромботической болезнью при непрерывном приеме флеботоников. Ангиология и сосудистая хирургия. 2014;20(2):52-57. [Zudin AM, Zasorina MA, Vikhert TA, Gonzalez AK, Tarkovskiy AA. Ultrasound assessment of alterations in venous haemodynamics in patients with post-thrombotic disease permanently taking phlebotonics. Angiology and Vascular Surgery. 2014;20(2):52-57. (In Russ.).]
  

[ Содержание выпуска N 1 | Выпуски журнала | Список журналов ]