Showing 1 - 20 results of 139 for search '"спермограмма"', query time: 0.79s Refine Results
  1. 1
  2. 2
  3. 3
    Academic Journal

    Source: Marine Medicine; Vol 10, No 4 (2024); 26-32 ; Морская медицина; Vol 10, No 4 (2024); 26-32 ; 2587-7828 ; 2413-5747

    File Description: application/pdf

  4. 4
    Academic Journal

    Contributors: 1

    Source: Marine Medicine; Vol 10, No 1 (2024); 54-63 ; Морская медицина; Vol 10, No 1 (2024); 54-63 ; 2587-7828 ; 2413-5747

    File Description: application/pdf

  5. 5
    Academic Journal

    Source: Andrology and Genital Surgery; Том 25, № 2 (2024); 54-60 ; Андрология и генитальная хирургия; Том 25, № 2 (2024); 54-60 ; 2412-8902 ; 2070-9781

    File Description: application/pdf

    Relation: https://agx.abvpress.ru/jour/article/view/758/581; Carlsen E, Giwercman A, Keiding N, Skakkebaek NE. Evidence for decreasing quality of semen during past 50 years. BMJ 1992;305:609–13. https://doi.org/10.1136/bmj.305.6854.609.; Levine H, Jørgensen N, Martino-Andrade A, Mendiola J, Weksler-Derri D, Jolles M, et al. Temporal trends in sperm count: a systematic review and meta-regression analysis of samples collected globally in the 20th and 21st centuries. Hum Reprod Update 2023;29:157–76. https://doi.org/10.1093/humupd/dmac035.; Cipriani S, Ricci E, Chiaffarino F, Esposito G, Dalmartello M, La Vecchia C, et al. Trend of change of sperm count and concentration over the last two decades: A systematic review and meta‐regression analysis. Andrology 2023;11:997–1008. https://doi.org/10.1111/andr.13396.; Korneyev IA. Annual trends in semen parameters among men attending a fertility center between 2016 and 2022. Urology Reports (St - Petersburg) 2023;13:23–9. https://doi.org/10.17816/uroved296569.; Agarwal A, Mulgund A, Hamada A, Chyatte MR. A unique view on male infertility around the globe. Reproductive Biology and Endocrinology 2015;13:37. https://doi.org/10.1186/s12958-015-0032-1.; Xie Y, Mirzaei M, Kahrizi MS, Shabestari AM, Riahi SM, Farsimadan M, et al. SARS-CoV-2 effects on sperm parameters: a meta-analysis study. J Assist Reprod Genet 2022;39:1555–63. https://doi.org/10.1007/s10815-022-02540-x.; Драпкина Ю.С., Долгушина Н.В., Шатылко Т.В., Николаева М.А., Менжинская И.В., Иванец Т.Ю., et al. Вакцина Гам-КОВИД-Вак (Спутник V) не оказывает негативного влияния на сперматогенез у мужчин. Акушерство и Гинекология 2021:88–94. https://doi.org/https://doi.org/10.18565/aig.202L7.88-94.; Punjani N, Alawamlh OA, Kim SJ, Salter CA, Wald G, Feliciano M, Williams N, Dudley V, Goldstein M. Changes in Semen Analysis over Time: A Temporal Trend Analysis of 20 Years of Subfertile Non-Azoospermic Men. World J Mens Health. 2023 Apr;41(2):382-389. doi:10.5534/wjmh.210201. Epub 2022 Jun 13.; Nordkap L, Joensen UN, Blomberg Jensen M, Jørgensen N. Regional differences and temporal trends in male reproductive health disorders: Semen quality may be a sensitive marker of environmental exposures. Mol Cell Endocrinol 2012;355:221–30. https://doi.org/10.1016/j.mce.2011.05.048.; Jensen TK, Heitmann BL, Jensen MB, Halldorsson TI, Andersson A-M, Skakkebæk NE, et al. High dietary intake of saturated fat is associated with reduced semen quality among 701 young Danish men from the general population. Am J Clin Nutr 2013;97:411–8. https://doi.org/10.3945/ajcn.112.042432.; Afeiche MC, Williams PL, Gaskins AJ, Mendiola J, Jørgensen N, Swan SH, et al. Meat Intake and Reproductive Parameters Among Young Men. Epidemiology 2014;25:323–30. https://doi.org/10.1097/EDE.0000000000000092.; LaPointe S, Mehta A, Gaskins AJ. Diet and recreational drug use in relation to male reproductive health. Fertil Steril 2023. https://doi.org/10.1016/j.fertnstert.2023.10.013.; Gollenberg AL, Liu F, Brazil C, Drobnis EZ, Guzick D, Overstreet JW, et al. Semen quality in fertile men in relation to psychosocial stress. Fertil Steril 2010;93:1104–11. https://doi.org/10.1016/j.fertnstert.2008.12.018.; Sharma R, Harlev A, Agarwal A, Esteves SC. Cigarette Smoking and Semen Quality: A New Meta-analysis Examining the Effect of the 2010 World Health Organization Laboratory Methods for the Examination of Human Semen. Eur Urol 2016;70:635–45. https://doi.org/10.1016/j.eururo.2016.04.010.; Kleshchev M, Osadchuk L, Osadchuk A. Age-Related Changes in Sperm Morphology and Analysis of Multiple Sperm Defects. Frontiers in Bioscience-Scholar 2023;15:12. https://doi.org/10.31083/j.fbs1503012.; Griffin DK. Is the Y chromosome disappearing?—Both sides of the argument. Chromosome Research 2012;20:35–45. https://doi.org/10.1007/s10577-011-9252-1.; Esteves SC, Roque M, Bedoschi G, Haahr T, Humaidan P. Intracytoplasmic sperm injection for male infertility and consequences for offspring. Nat Rev Urol 2018;15:535–62. https://doi.org/10.1038/s41585-018-0051-8.; https://agx.abvpress.ru/jour/article/view/758

  6. 6
    Academic Journal

    Source: Andrology and Genital Surgery; Том 25, № 1 (2024); 147-158 ; Андрология и генитальная хирургия; Том 25, № 1 (2024); 147-158 ; 2412-8902 ; 2070-9781

    File Description: application/pdf

    Relation: https://agx.abvpress.ru/jour/article/view/746/577; Gual-Frau J, Abad C, Amengual MJ et al. Oral antioxidant treatment partly improves integrity of human sperm DNA in infertile grade I varicocele patients. Hum Fertil (Camb). 2015 Sep;18(3):225-9. doi:10.3109/14647273.2015.1050462. Epub 2015 Jun 19. PMID: 26090928.; Balercia G, Regoli F, Armeni T et al. Placebo-controlled double-blind randomized trial on the use of L-carnitine, L-acetylcarnitine, or combined L-carnitine and L-acetylcarnitine in men with idiopathic asthenozoospermia. Fertil Steril. 2005 Sep;84(3):662-71. doi:10.1016/j.fertnstert.2005.03.064. PMID: 16169400.; Busetto GM, Agarwal A, Virmani A et al. Effect of metabolic and antioxidant supplementation on sperm parameters in oligo-astheno-teratozoospermia, with and without varicocele: A double-blind placebo-controlled study. Andrologia. 2018 Apr;50(3). doi:10.1111/and.12927. Epub 2018 Jan 7. PMID: 29315686.; Адъювантная антиоксидантная терапия у больных бесплодием при варикоцеле / С. И. Гамидов, Р. И. Овчинников, А. Ю. Попова [и др.] // Урология. – 2017. – № 2-S2. – С. 64-72. – DOI 10.18565/urol.2017.2(suppl.2)64-72.; Корнеев И.А. Мужское бесплодие, метаболический синдром и ожирение / И. А. Корнеев, И. А. Мацуева // Урологические ведомости. 2021. Т. 11, № 2. С. 153–162. – DOI:10.17816/uroved61509.; Engin-Ustun Y, Yílmaz N, Akgun N et al. Body Mass Index Effects Kruger’s Criteria in Infertile Men. Int J Fertil Steril. 2018 Jan;11(4):258-262. doi:10.22074/ijfs.2018.4888. Epub 2017 Oct 12. PMID: 29043700; PMCID: PMC5641456.; Abdali D, Samson SE, Grover AK. How effective are antioxidant supplements in obesity and diabetes? Med Princ Pract. 2015;24(3):201-15. doi:10.1159/000375305. Epub 2015 Mar 14. PMID: 25791371; PMCID: PMC5588240.; Jeulin C, Lewin LM. Role of free L-carnitine and acetyl-Lcarnitine in post-gonadal maturation of mammalian spermatozoa. Hum Reprod Update. 1996 Mar-Apr;2(2):87-102. doi:10.1093/humupd/2.2.87. PMID: 9079406.; Showell MG, Brown J, Yazdani A, Stankiewicz MT, Hart RJ. Antioxidants for male subfertility. Cochrane Database Syst Rev. 2011 Jan 19;(1):CD007411. doi:10.1002/14651858.CD007411. pub2. Update in: Cochrane Database Syst Rev. 2014;12:CD007411. PMID: 21249690.; Li Y, Xie Y, Qiu C, Yu B, Yang F, Cheng Y, Zhong W, Yuan J. Effects of L-carnitine supplementation on glucolipid metabolism: a systematic review and meta-analysis. Food Funct. 2023 Mar 6;14(5):2502-2517. doi:10.1039/d2fo02930h. PMID: 36815696.; Choi M, Park S, Lee M. L-Carnitine’s Effect on the Biomarkers of Metabolic Syndrome: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Nutrients. 2020 Sep 12;12(9):2795. doi:10.3390/nu12092795. PMID: 32932644; PMCID: PMC7551203.; Martínez-Soto JC, Domingo JC, Cordobilla B et al. Dietary supplementation with docosahexaenoic acid (DHA) improves seminal antioxidant status and decreases sperm DNA fragmentation. Syst Biol Reprod Med. 2016 Dec;62(6):387-395. doi:10.1080/19396368.2016.1246623. Epub 2016 Oct 28. PMID: 27792396.; González-Ravina C, Aguirre-Lipperheide M, Pinto F et al. Effect of dietary supplementation with a highly pure and concentrated docosahexaenoic acid (DHA) supplement on human sperm function. Reprod Biol. 2018 Sep;18(3):282-288. doi:10.1016/j.repbio.2018.06.002. Epub 2018 Jun 20. PMID: 29934046.; Попова А.Ю., Гамидов С.И., Овчинников Р.И. и др. Опыт применения докозагексаеновой кислоты (БрудиПлюс) у пациентов с повышенным индексом фрагментации ДНК сперматозоидов в Научном центре акушерства, гинекологии и перинатологии им. акад. В.И. Кулакова. Андрология и генитальная хирургия. 2015;16(2):51-55. https://doi.org/10.17650/2070-9781-2015-16-2-51-55; Виноградов И.В., Живулько А.Р. Докозагексаеновая кислота в лечении мужского бесплодия, вызванного высоким уровнем фрагментации ДНК сперматозоидов. Андрология и генитальная хирургия. 2020;21(4):89-97. https://doi.org/10.17650/2070-9781-2020-21-4-89-97; Nashivochnikova NA, Krupin VN, Leanovich VE. [The role of antioxidants in the therapy of metabolic syndrome in men]. Urologiia. 2023 Sep;(4):90-97. Russian. PMID: 37850287.; Zhang JJ, Wu ZB, Cai YJ et al. L-carnitine ameliorated fastinginduced fatigue, hunger, and metabolic abnormalities in patients with metabolic syndrome: a randomized controlled study. Nutr J. 2014 Nov 26;13:110. doi:10.1186/1475-2891-13-110. PMID: 25424121; PMCID: PMC4258024.; Talenezhad N, Mohammadi M, Ramezani-Jolfaie N et al. Effects of l-carnitine supplementation on weight loss and body composition: A systematic review and meta-analysis of 37 randomized controlled clinical trials with dose-response analysis. Clin Nutr ESPEN. 2020 Jun;37:9-23. doi:10.1016/j.clnesp.2020.03.008. Epub 2020 Apr 18. PMID: 32359762.; Alberti KG, Eckel RH, Grundy SM et al. Harmonizing the metabolic syndrome: a joint interim statement of the International Diabetes Federation Task Force on Epidemiology and Prevention; National Heart, Lung, and Blood Institute; American Heart Association; World Heart Federation; International Atherosclerosis Society; and International Association for the Study of Obesity. Circulation. 2009 Oct 20;120(16):1640-5. doi:10.1161/CIRCULATIONAHA.109.192644. Epub 2009 Oct 5. PMID: 19805654.; Корнеев И.А. Персонифицированный подход к назначению антиоксидантной терапии мужчинам, состоящим в бесплодном браке / И. А. Корнеев // Клинический разбор в общей медицине. 2023. Т. 4, № 4. С. 82–88. DOI:10.47407/kr2023.4.4.00260.; Корнеев И.А. Изучение показателей эякулята и результатов лечения с применением вспомогательных репродуктивных технологий мужчин с избыточным весом и ожирением, состоящих в бесплодном браке / И. А. Корнеев // Урологические ведомости. 2022. Т. 12, № 1. С. 41-48. DOI:10.17816/uroved99867.; Saleh AAEW, Amin EM, Elfallah AA et al. Insulin resistance and idiopathic infertility: A potential possible link. Andrologia. 2020 Dec;52(11):e13773. doi:10.1111/and.13773. Epub 2020 Aug 20. PMID: 32816339.; Verit A, Verit FF, Oncel H et al. Is there any effect of insulin resistance on male reproductive system? Arch Ital Urol Androl. 2014 Mar 28;86(1):5-8. doi:10.4081/aiua.2014.1.5. PMID: 24704923.; Ma J, Han RY, Mei XA et al. [Correlation of insulin resistance with male reproductive hormone levels and semen parameters]. Zhonghua Nan Ke Xue. 2018 Aug;24(8):695-699. Chinese. PMID: 30173427.; La Vignera S, Condorelli RA, Duca Y et al. FSH treatment for normogonadotropic male infertility: a synergistic role for metformin? Eur Rev Med Pharmacol Sci. 2019 Jul;23(13): 5994-5998. doi:10.26355/eurrev_201907_18346. PMID: 31298351.; Xu Y, Jiang W, Chen G, Zhu W et al. L-carnitine treatment of insulin resistance: A systematic review and meta-analysis. Adv Clin Exp Med. 2017 Mar-Apr;26(2):333-338. doi:10.17219/acem/61609. PMID: 28791854.; Zamani M, Pahlavani N, Nikbaf-Shandiz M et al. The effects of L-carnitine supplementation on glycemic markers in adults: A systematic review and dose-response meta-analysis. Front Nutr. 2023 Jan 10;9:1082097. doi:10.3389/fnut.2022.1082097. PMID: 36704801; PMCID: PMC9871499.; Гамидов С.И. Мужское здоровье и ожирение Э диагностика и терапевтические подходы / С.И. Гамидов, Т.В. Шатылко, Н.Г. Гасанов // Ожирение и метаболизм. 2019. Т. 16, № 3. С. 29–36. DOI:10.14341/omet10314.; Епанчинцева Е.А., Селятицкая В.Г., Корнеев И.А., Бабенко А.Ю. Влияние ингибиторов ароматазы на мужскую фертильность: обзор литературы. Андрология и генитальная хирургия. 2023;24(4):49-58. https://doi.org/10.17650/2070-9781-2023-24-4-49-58; Abbott K, Burrows TL, Acharya S et al. Dietary supplementation with docosahexaenoic acid rich fish oil increases circulating levels of testosterone in overweight and obese men. Prostaglandins Leukot Essent Fatty Acids. 2020 Dec;163:102204. doi:10.1016/j.plefa.2020.102204. Epub 2020 Nov 12. PMID: 33221700.; Kelley DS, Adkins Y, Woodhouse LR et al. Docosahexaenoic acid supplementation improved lipocentric but not glucocentric markers of insulin sensitivity in hypertriglyceridemic men. Metab Syndr Relat Disord. 2012 Feb;10(1):32-8. doi:10.1089/met.2011.0081. Epub 2011 Oct 14. PMID: 21999398.; Sahebkar A. Effect of L-carnitine Supplementation on Circulating C-reactive Protein Levels: A Systematic Review and Meta-Analysis. J Med Biochem. 2015 Apr;34(2):151-159. doi:10.2478/jomb-2014-0030. Epub 2015 Mar 3. PMID: 28356827; PMCID: PMC4922328.; Savica V, Santoro D, Mazzaglia G et al. L-carnitine infusions may suppress serum C-reactive protein and improve nutritional status in maintenance hemodialysis patients. J Ren Nutr. 2005 Apr;15(2):225-30. doi:10.1053/j.jrn.2004.10.002. PMID: 15827896.; Elisia I, Yeung M, Kowalski S et al. Omega 3 supplementation reduces C-reactive protein, prostaglandin E2 and the granulocyte/ lymphocyte ratio in heavy smokers: An open-label randomized crossover trial. Front Nutr. 2022 Dec 1;9:1051418. doi:10.3389/fnut.2022.1051418. PMID: 36532545; PMCID: PMC9751896.; Wang TM, Hsieh SC, Chen JW et al. Docosahexaenoic acid and eicosapentaenoic acid reduce C-reactive protein expression and STAT3 activation in IL-6-treated HepG2 cells. Mol Cell Biochem. 2013 May;377(1-2):97-106. doi:10.1007/s11010-013-1574-1. Epub 2013 Jan 30. PMID: 23361365.; Структурные нарушения хроматина сперматозоидов. Патофизиологические аспекты. Клиническая значимость / М.Н. Коршунов, Е.С. Коршунова, П.С. Кызласов [и др.] // Вестник урологии. 2021. Т. 9, № 1. С. 95-104. DOI:10.21886/2308-6424-2021-9-1-95-104.; Фрагментация ДНК сперматозоидов: клиническая значимость, причины, методы оценки и коррекции / С.Ю. Боровец, В.А. Егорова, А.М. Гзгзян, С. Х. и др.// Урологические ведомости. 2020. Т. 10, № 2. С. 173–180. DOI:10.17816/uroved102173-180.; Лечение бесплодия, ассоциированного с высоким уровнем фрагментации ДНК сперматозоидов / Ю.В. Олефир, М.Н. Коршунов, А.Р. Живулько и др.// Экспериментальная и клиническая урология. 2022. Т. 15, № 1. С. 112–119. DOI:10.29188/2222-8543-2022-15-1-112-119.; Кореньков Д.Г., Павлов А.Л., Казимзаде Э.Д. Влияние препарата БЕСТФертил на репродуктивную функцию у мужчин с идиопатическим бесплодием. Андрология и генитальная хирургия. 2018;19(4):54-59. https://DOI.org/10.17650/2070-9781-2018-19-4-54-59; Гамидов С.И., Попова А.Ю., Гасанов Н.Г. и др. Оценка влияния комплекса «БЕСТФертил» на показатели спермограммы, оксидативного стресса и фрагментации ДНК сперматозоидов у мужчин с бесплодием. Андрология и генитальная хирургия. 2019;20(1):91-98. https://doi.org/10.17650/2070-9781-2019-20-1-91-98; Шатылко Т.В., Гамидов С.И., Попова А.Ю. Оценка влияния антиоксидантного комплекса БЕСТФертил на показатели спермограммы и выраженность астенического синдрома у мужчин, перенесших новую коронавирусную инфекцию (COVID-19). Андрология и генитальная хирургия. 2021;22(4):68-76. https://doi.org/10.17650/1726-9784-2021-22-4-68-76; Оксидативный стресс сперматозоидов: клиническое значение и коррекция / С.И. Гамидов, Т.В. Шатылко, А.Ю. Попова [и др.] // Медицинский совет. 2021. № 3. С. 19–27. DOI 10.21518/2079-701X-2021-3-19-27.; https://agx.abvpress.ru/jour/article/view/746

  7. 7
    Academic Journal

    Authors: Rykova, O.V.

    Source: INTERNATIONAL JOURNAL OF ENDOCRINOLOGY; Том 15, № 3 (2019); 246-252
    Международный эндокринологический журнал-Mìžnarodnij endokrinologìčnij žurnal; Том 15, № 3 (2019); 246-252
    Міжнародний ендокринологічний журнал-Mìžnarodnij endokrinologìčnij žurnal; Том 15, № 3 (2019); 246-252

    File Description: application/pdf

  8. 8
    Academic Journal

    Authors: Rykova, O.V.

    Source: INTERNATIONAL JOURNAL OF ENDOCRINOLOGY; Том 15, № 1 (2019); 44-49
    Международный эндокринологический журнал-Mìžnarodnij endokrinologìčnij žurnal; Том 15, № 1 (2019); 44-49
    Міжнародний ендокринологічний журнал-Mìžnarodnij endokrinologìčnij žurnal; Том 15, № 1 (2019); 44-49

    File Description: application/pdf

  9. 9
  10. 10
  11. 11
  12. 12
    Academic Journal

    Source: Urology Herald; Том 11, № 1 (2023); 125-133 ; Вестник урологии; Том 11, № 1 (2023); 125-133 ; 2308-6424 ; 10.21886/2308-6424-2023-11-1

    File Description: application/pdf

    Relation: https://www.urovest.ru/jour/article/view/677/453; Zhu N, Zhang D, Wang W, Li X, Yang B, Song J, Zhao X, Huang B, Shi W, Lu R, Niu P, Zhan F, Ma X, Wang D, Xu W, Wu G, Gao GF, Tan W; China Novel Coronavirus Investigating and Research Team. A Novel Coronavirus from Patients with Pneumonia in China, 2019. N Engl J Med. 2020;382(8):727-733. DOI:10.1056/NEJMoa2001017; Zhou P, Yang XL, Wang XG, Hu B, Zhang L, Zhang W, Si HR, Zhu Y, Li B, Huang CL, Chen HD, Chen J, Luo Y, Guo H, Jiang RD, Liu MQ, Chen Y, Shen XR, Wang X, Zheng XS, Zhao K, Chen QJ, Deng F, Liu LL, Yan B, Zhan FX, Wang YY, Xiao GF, Shi ZL. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature. 2020;579(7798):270- 273. Erratum in: Nature. 2020;588(7836):E6. DOI:10.1038/s41586-020-2012-7; Lu R, Zhao X, Li J, Niu P, Yang B, Wu H, Wang W, Song H, Huang B, Zhu N, Bi Y, Ma X, Zhan F, Wang L, Hu T, Zhou H, Hu Z, Zhou W, Zhao L, Chen J, Meng Y, Wang J, Lin Y, Yuan J, Xie Z, Ma J, Liu WJ, Wang D, Xu W, Holmes EC, Gao GF, Wu G, Chen W, Shi W, Tan W. Genomic characterisation and epidemiology of 2019 novel coronavirus: implications for virus origins and receptor binding. Lancet. 2020;395(10224):565- 574. DOI:10.1016/S0140-6736(20)30251-8; Jalaleddine N, Bouzid A, Hachim M, Sharif-Askari NS, Mahboub B, Senok A, Halwani R, Hamoudi RA, Al Heialy S. ACE2 polymorphisms impact COVID-19 severity in obese patients. Sci Rep. 2022;12(1):21491. DOI:10.1038/s41598-022-26072-7; Wang Z, Xu X. scRNA-seq Profiling of Human Testes Reveals the Presence of the ACE2 Receptor, A Target for SARSCoV-2 Infection in Spermatogonia, Leydig and Sertoli Cells. Cells. 2020;9(4):920. DOI:10.3390/cells9040920; Bechmann N, Maccio U, Kotb R, Dweik RA, Cherfane M, Moch H, Bornstein SR, Varga Z. COVID-19 Infections in Gonads: Consequences on Fertility? Horm Metab Res. 2022;54(8):549-555. DOI:10.1055/a-1891-6621; Pan F, Xiao X, Guo J, Song Y, Li H, Patel DP, Spivak AM, Alukal JP, Zhang X, Xiong C, Li PS, Hotaling JM. No evidence of severe acute respiratory syndrome-coronavirus 2 in semen of males recovering from coronavirus disease 2019. Fertil Steril. 2020;113(6):1135-1139. DOI:10.1016/j.fertnstert.2020.04.024; Song C, Wang Y, Li W, Hu B, Chen G, Xia P, Wang W, Li C, Diao F, Hu Z, Yang X, Yao B, Liu Y. Absence of 2019 novel coronavirus in semen and testes of COVID-19 patients†. Biol Reprod. 2020;103(1):4-6. DOI:10.1093/biolre/ioaa050; Guo L, Zhao S, Li W, Wang Y, Li L, Jiang S, Ren W, Yuan Q, Zhang F, Kong F, Lei J, Yuan M. Absence of SARS-CoV-2 in semen of a COVID-19 patient cohort. Andrology. 2021;9(1):42-47. DOI:10.1111/andr.12848; Sanders JM, Monogue ML, Jodlowski TZ, Cutrell JB. Pharmacologic Treatments for Coronavirus Disease 2019 (COVID-19): A Review. JAMA. 2020;323(18):1824-1836. DOI:10.1001/jama.2020.6019; Tang X, Wu C, Li X, Song Y, Yao X, Wu X, Duan Y, Zhang H, Wang Y, Qian Z, Cui J, Lu J. On the origin and continuing evolution of SARS-CoV-2. Natl Sci Rev. 2020;7(6):1012-1023. DOI:10.1093/nsr/nwaa036; Jackson CB, Farzan M, Chen B, Choe H. Mechanisms of SARS-CoV-2 entry into cells. Nat Rev Mol Cell Biol. 2022;23(1):3-20. DOI:10.1038/s41580-021-00418-x; Atzrodt CL, Maknojia I, McCarthy RDP, Oldfield TM, Po J, Ta KTL, Stepp HE, Clements TP. A Guide to COVID-19: a global pandemic caused by the novel coronavirus SARS-CoV-2. FEBS J. 2020;287(17):3633-3650. DOI:10.1111/febs.15375; Harrison AG, Lin T, Wang P. Mechanisms of SARS-CoV-2 Transmission and Pathogenesis. Trends Immunol. 2020;41(12):1100-1115. DOI:10.1016/j.it.2020.10.004; Lin B, Ferguson C, White JT, Wang S, Vessella R, True LD, Hood L, Nelson PS. Prostate-localized and androgen-regulated expression of the membrane-bound serine protease TMPRSS2. Cancer Res. 1999;59(17):4180-4. PMID: 10485450; Senapati S, Banerjee P, Bhagavatula S, Kushwaha PP, Kumar S. Contributions of human ACE2 and TMPRSS2 in determining host-pathogen interaction of COVID-19. J Genet. 2021;100(1):12. DOI:10.1007/s12041-021-01262-w; Huang C, Jiang Y, Yan J. Comparative analyses of ACE2 and TMPRSS2 gene: Implications for the risk to which vertebrate animals are susceptible to SARS-CoV-2. J Med Virol. 2021;93(9):5487-5504. DOI:10.1002/jmv.27073; Singh H, Choudhari R, Nema V, Khan AA. ACE2 and TMPRSS2 polymorphisms in various diseases with special reference to its impact on COVID-19 disease. Microb Pathog. 2021;150:104621. DOI:10.1016/j.micpath.2020.104621; Xu J, Qi L, Chi X, Yang J, Wei X, Gong E, Peh S, Gu J. Orchitis: a complication of severe acute respiratory syndrome (SARS). Biol Reprod. 2006;74(2):410-6. DOI:10.1095/biolreprod.105.044776; Ибишев Х.С., Мамедов Э.А., Гусова З.Р., Паленный А.И., Прокоп Я.О. Показатели тестостерона в сыворотке крови и гемодинамики тестикул до и после инфицирования SARS-COV-2 (пилотное исследование). Урология. 2021;(5):5-9. DOI:10.18565/urology.2021.5.5-9; Auerbach JM, Khera M. Testosterone's Role in COVID-19. J Sex Med. 2021;18(5):843-848. DOI:10.1016/j.jsxm.2021.03.004 22. Okçelik S. COVID-19 pneumonia causes lower testosterone levels. Andrologia. 2021;53(1):e13909. DOI:10.1111/and.13909; Montaño LM, Sommer B, Solís-Chagoyán H, RomeroMartínez BS, Aquino-Gálvez A, Gomez-Verjan JC, Calixto E, González-Avila G, Flores-Soto E. Could Lower Testosterone in Older Men Explain Higher COVID-19 Morbidity and Mortalities? Int J Mol Sci. 2022;23(2):935. DOI:10.3390/ijms23020935; Yang M, Chen S, Huang B, Zhong JM, Su H, Chen YJ, Cao Q, Ma L, He J, Li XF, Li X, Zhou JJ, Fan J, Luo DJ, Chang XN, Arkun K, Zhou M, Nie X. Pathological Findings in the Testes of COVID-19 Patients: Clinical Implications. Eur Urol Focus. 2020;6(5):1124-1129. DOI:10.1016/j.euf.2020.05.009; Тодоров С.С., Ибишев Х.С., Васильев О.Н., Прокоп Я.О. Морфологические изменения тестикул пациентов с идиопатической инфертильностью, перенесших новую коронавирусную инфекцию (COVID-19). Вестник урологии. 2022;10(2):72-77. DOI:10.21886/2308-6424-2022-10-2-72-77; Erbay G, Sanli A, Turel H, Yavuz U, Erdogan A, Karabakan M, Yaris M, Gultekin MH. Short-term effects of COVID-19 on semen parameters: A multicenter study of 69 cases. Andrology. 2021;9(4):1060-1065. DOI:10.1111/andr.13019; Holtmann N, Edimiris P, Andree M, Doehmen C, BastonBuest D, Adams O, Kruessel JS, Bielfeld AP. Assessment of SARS-CoV-2 in human semen-a cohort study. Fertil Steril. 2020;114(2):233-238. DOI:10.1016/j.fertnstert.2020.05.028; Best JC, Kuchakulla M, Khodamoradi K, Lima TFN, Frech FS, Achua J, Rosete O, Mora B, Arora H, Ibrahim E, Ramasamy R. Evaluation of SARS-CoV-2 in Human Semen and Effect on Total Sperm Number: A Prospective Observational Study. World J Mens Health. 2021;39(3):489-495. DOI:10.5534/wjmh.200192; Guo TH, Sang MY, Bai S, Ma H, Wan YY, Jiang XH, Zhang YW, Xu B, Chen H, Zheng XY, Luo SH, Xie XF, Gong CJ, Weng JP, Shi QH. Semen parameters in men recovered from COVID-19. Asian J Androl. 2021;23(5):479-483. DOI:10.4103/aja.aja_31_21; Hajizadeh Maleki B, Tartibian B. COVID-19 and male reproductive function: a prospective, longitudinal cohort study. Reproduction. 2021;161(3):319-331. DOI:10.1530/REP-20-0382; Donders GGG, Bosmans E, Reumers J, Donders F, Jonckheere J, Salembier G, Stern N, Jacquemyn Y, Ombelet W, Depuydt CE. Sperm quality and absence of SARS-CoV-2 RNA in semen after COVID-19 infection: a prospective, observational study and validation of the SpermCOVID test. Fertil Steril. 2022;117(2):287-296. DOI:10.1016/j.fertnstert.2021.10.022; Wang M, Hu J, Huang B, Yang Q, Liu S, Li Z, Yang L, Xi Q, Zhu L, Jin L. Investigating the impact of SARS-CoV-2 infection on basic semen parameters and in vitro fertilization/ intracytoplasmic sperm injection outcomes: a retrospective cohort study. Reprod Biol Endocrinol. 2022;20(1):46. DOI:10.1186/s12958-022-00918-1; Carli G, Nichele I, Ruggeri M, Barra S, Tosetto A. Deep vein thrombosis (DVT) occurring shortly after the second dose of mRNA SARS-CoV-2 vaccine. Intern Emerg Med. 2021;16(3):803-804. DOI:10.1007/s11739-021-02685-0; Jeet Kaur R, Dutta S, Charan J, Bhardwaj P, Tandon A, Yadav D, Islam S, Haque M. Cardiovascular Adverse Events Reported from COVID-19 Vaccines: A Study Based on WHO Database. Int J Gen Med. 2021;14:3909-3927. DOI:10.2147/IJGM.S324349; Hippisley-Cox J, Patone M, Mei XW, Saatci D, Dixon S, Khunti K, Zaccardi F, Watkinson P, Shankar-Hari M, Doidge J, Harrison DA, Griffin SJ, Sheikh A, Coupland CAC. Risk of thrombocytopenia and thromboembolism after covid-19 vaccination and SARS-CoV-2 positive testing: self-controlled case series study. BMJ. 2021;374:n1931. DOI:10.1136/bmj.n1931; Elimam A, Al-Dhahir S. Pfizer COVID-19 vaccine-induced peritonitis. Clin Med (Lond). 2022;22(Suppl 4):53. DOI:10.7861/clinmed.22-4-s53; Barbari A. COVID-19 Vaccine Concerns: Fact or Fiction? Exp Clin Transplant. 2021;19(7):627-634. DOI:10.6002/ect.2021.0056; Kricorian K, Civen R, Equils O. COVID-19 vaccine hesitancy: misinformation and perceptions of vaccine safety. Hum Vaccin Immunother. 2022;18(1):1950504. DOI:10.1080/21645515.2021.1950504; Prabani KIP, Weerasekara I, Damayanthi HDWT. COVID-19 vaccine acceptance and hesitancy among patients with cancer: a systematic review and meta-analysis. Public Health. 2022;212:66-75. DOI:10.1016/j.puhe.2022.09.001; Navin MC, Oberleitner LM, Lucia VC, Ozdych M, Afonso N, Kennedy RH, Keil H, Wu L, Mathew TA. COVID-19 Vaccine Hesitancy Among Healthcare Personnel Who Generally Accept Vaccines. J Community Health. 2022;47(3):519-529. DOI:10.1007/s10900-022-01080-w; Schaler L, Wingfield M. COVID-19 vaccine - can it affect fertility? Ir J Med Sci. 2022;191(5):2185-2187. DOI:10.1007/s11845-021-02807-9; Gonzalez DC, Nassau DE, Khodamoradi K, Ibrahim E, Blachman-Braun R, Ory J, Ramasamy R. Sperm Parameters Before and After COVID-19 mRNA Vaccination. JAMA. 2021;326(3):273-274. DOI:10.1001/jama.2021.9976; Сорокина Т.М., Брагина Е.Е., Сорокина Е.А., Курило Л.Ф., Штаут М.И., Седова А.О., Черных В.Б. Оценка и сравнительный анализ спермиологических показателей у мужчин до и после вакцинации препаратом «Спутник V» (Гам-КОВИД-Вак). Андрология и генитальная хирургия. 2021;22(4):45-53. DOI:10.17650/1726-9784-2021-22-4-45-53; Xia W, Zhao J, Hu Y, Fang L, Wu S. Investigate the effect of COVID-19 inactivated vaccine on sperm parameters and embryo quality in in vitro fertilization. Andrologia. 2022;54(6):e14483. DOI:10.1111/and.14483; https://www.urovest.ru/jour/article/view/677

  13. 13
    Academic Journal

    Source: Urology Herald; Том 11, № 3 (2023); 16-22 ; Вестник урологии; Том 11, № 3 (2023); 16-22 ; 2308-6424 ; 10.21886/2308-6424-2023-11-3

    File Description: application/pdf

    Relation: https://www.urovest.ru/jour/article/view/752/489; Gorelick JI, Goldstein M. Loss of fertility in men with varicocele. Fertil Steril. 1993;59(3):613-6. PMID: 8458466.; Котов С.В., Корочкин Н.Д., Клименко А.А. Рецидивное варикоцеле. Вестник урологии. 2021;9(2):132-141. DOI:10.21886/2308-6424-2021-9-2-132-141; Sharlip ID, Jarow JP, Belker AM, Lipshultz LI, Sigman M, Thomas AJ, Schlegel PN, Howards SS, Nehra A, Damewood MD, Overstreet JW, Sadovsky R. Best practice policies for male infertility. Fertil Steril. 2002;77(5):873-82. DOI:10.1016/s0015-0282(02)03105-9; Abdel-Meguid TA, Al-Sayyad A, Tayib A, Farsi HM. Does varicocele repair improve male infertility? An evidence-based perspective from a randomized, controlled trial. Eur Urol. 2011;59(3):455-61. DOI:10.1016/j.eururo.2010.12.008; Кызласов П.С., Абуев Г.Г., Мустафаев А.Т., Боков А.И. Опыт применения новой малоинвазивной методики оперативного лечения варикоцеле. Вестник урологии. 2023;11(1):52-58. DOI:10.21886/2308-6424-2023-11-1-52-58; Ficarra V, Cerruto MA, Liguori G, Mazzoni G, Minucci S, Tracia A, Gentile V. Treatment of varicocele in subfertile men: The Cochrane Review--a contrary opinion. Eur Urol. 2006;49(2):258-63. DOI:10.1016/j.eururo.2005.11.023; Yamamoto M, Hibi H, Hirata Y, Miyake K, Ishigaki T. Effect of varicocelectomy on sperm parameters and pregnancy rate in patients with subclinical varicocele: a randomized prospective controlled study. J Urol. 1996;155(5):1636-8. PMID: 8627841.; Breznik R, Vlaisavljević V, Borko E. Treatment of varicocele and male fertility. Arch Androl. 1993;30(3):157-60. DOI:10.3109/01485019308987750; Bungum M, Humaidan P, Spano M, Jepson K, Bungum L, Giwercman A. The predictive value of sperm chromatin structure assay (SCSA) parameters for the outcome of intrauterine insemination, IVF and ICSI. Hum Reprod. 2004;19(6):1401-8. DOI:10.1093/humrep/deh280; Рогозин Д.С. Мужская фертильность: обзор публикаций июля — сентября 2021 года. Вестник урологии. 2021;9(4):147-155. DOI:10.21886/2308-6424-2021-9-4-147-155; Ding H, Tian J, Du W, Zhang L, Wang H, Wang Z. Open non-microsurgical, laparoscopic or open microsurgical varicocelectomy for male infertility: a meta-analysis of randomized controlled trials. BJU Int. 2012;110(10):1536-42. DOI:10.1111/j.1464-410X.2012.11093.x; Persad E, O'Loughlin CA, Kaur S, Wagner G, Matyas N, Hassler-Di Fratta MR, Nussbaumer-Streit B. Surgical or radiological treatment for varicoceles in subfertile men. Cochrane Database Syst Rev. 2021;4(4):CD000479. DOI:10.1002/14651858.CD000479.pub6; WHO laboratory manual for the examination and processing of human semen, sixth edition. Geneva: World Health Organization; 2021. ISBN: 978-92-4-003078-7 (electronic version); WHO Manual for the Standardized Investigation, Diagnosis and Management of the Infertile Male. Cambridge: Cambridge University Press; 2000. ISBN: 9780521774741; Agarwal A, Prabakaran S, Allamaneni SS. Relationship between oxidative stress, varicocele and infertility: a meta-analysis. Reprod Biomed Online. 2006;12(5):630-3. DOI:10.1016/s1472-6483(10)61190-x; Agarwal A, Deepinder F, Cocuzza M, Agarwal R, Short RA, Sabanegh E, Marmar JL. Efficacy of varicocelectomy in improving semen parameters: new meta-analytical approach. Urology. 2007;70(3):532-8. DOI:10.1016/j.urology.2007.04.011; Boman JM, Libman J, Zini A. Microsurgical varicocelectomy for isolated asthenospermia. J Urol. 2008;180(5):2129-32. DOI:10.1016/j.juro.2008.07.046; Schatte EC, Hirshberg SJ, Fallick ML, Lipschultz LI, Kim ED. Varicocelectomy improves sperm strict morphology and motility. J Urol. 1998;160(4):1338-40. PMID: 9751350.; Божедомов В.А., Шомаруфов А.Б., Божедомова Г.Е., Охоботов Д.А., Камалов Д.М., Сорокин Н.И., Камалов А.А. Варикоцеле и репродуктивная функция: возможности коррекции патозооспермии (данные проспективного сравнительного исследования). Урология. 2021;(2):62-68. DOI:10.18565/urology.2021.2.62-68; Shomarufov AB, Bozhedomov VA, Akilov FA, Mukhtarov ST, Giyasov SI, Abbosov SA, Kamalov AA. Prediction of reproductive function recovery after microsurgical varicocelectomy in men from infertile couples: Clinical and laboratory predictors. Andrologia. 2021;53(8):e14101. DOI:10.1111/and.14101; Almekaty K, Zahran MH, Zoeir A, Minhas S, Salem K. The role of artery-preserving varicocelectomy in subfertile men with severe oligozoospermia: a randomized controlled study. Andrology. 2019;7(2):193-198. DOI:10.1111/andr.12580; Enatsu N, Yamaguchi K, Chiba K, Miyake H, Fujisawa M. Clinical outcome of microsurgical varicocelectomy in infertile men with severe oligozoospermia. Urology. 2014;83(5):1071-4. DOI:10.1016/j.urology.2014.01.029; https://www.urovest.ru/jour/article/view/752

  14. 14
    Academic Journal

    Source: Andrology and Genital Surgery; Том 24, № 1 (2023); 150-156 ; Андрология и генитальная хирургия; Том 24, № 1 (2023); 150-156 ; 2412-8902 ; 2070-9781

    File Description: application/pdf

    Relation: https://agx.abvpress.ru/jour/article/view/645/512; Aitken R.J., Best F.S.M., Richardson D.W. et al. An analysis of sperm function in cases of unexplained infertility: conventional criteria, movement characteristics, and fertilizing capacity. Fertil Steril 1982;38(2):212–21. DOI:10.1016/S0015-0282(16)46462-9; Agarwal A., Saleh R.A. Role of oxidants in male infertility: rationale, significance, and treatment. Urol Clin North Am 2002;29(4):817–27. DOI:10.1016/s0094-0143(02)00081-2; Sharma R.K., Agarwal A. Role of reactive oxygen species in male infertility. Urology 1996;48(6):835–50. DOI:10.1016/s0090-4295(96)00313-5; Sanocka D., Jedrzejczak P., Szumała-Kaekol A. et al. Male genital tract inflammation: the role of selected interleukins in regulation of pro-oxidant and antioxidant enzymatic substances in seminal plasma. J Androl 2003;24(3):448–55. DOI:10.1002/j.1939-4640.2003.tb02693.x; Koçak I., Yenisey C., Dündar M. et al. Relationship between seminal plasma interleukin-6 and tumor necrosis factor alpha levels with semen parameters in fertile and infertile men. Urol Res 2002;30(4):263–7. DOI:10.1007/s00240-002-0269-y; Gruschwitz M.S., Brezinschek R., Brezinschek H.P. Cytokine levels in the seminal plasma of infertile males. J Androl 1996;17(2):158–63.; Elfassy Y., Bongrani A., Levy P. et al. Relationships between metabolic status, seminal adipokines, and reproductive functions in men from infertile couples. Eur J Endocrinol 2020;182(1):67–7. DOI:10.1530/EJE-19-0615; Grande G., Milardi D., Baroni S. et al. Identification of seminal markers of male accessory gland inflammation: from molecules to proteome. Am J Reprod Immunol 2018;80(2):e12992. DOI:10.1111/aji.12992; Seshadri S., Bates M., Vince G., Jones D.I.L. The role of cytokine expression in different subgroups of subfertile men. Am J Reprod Immunol 2009;62(5):275–82. DOI:10.1111/j.1600-0897.2009.00736.x; https://agx.abvpress.ru/jour/article/view/645

  15. 15
    Academic Journal

    Source: Andrology and Genital Surgery; Том 24, № 4 (2023); 49-58 ; Андрология и генитальная хирургия; Том 24, № 4 (2023); 49-58 ; 2412-8902 ; 2070-9781

    File Description: application/pdf

    Relation: https://agx.abvpress.ru/jour/article/view/704/549; Levine H., Jørgensen N., Martino-Andrade A. et al. Temporal trends in sperm count: a systematic review and meta-regression analysis of samples collected globally in the 20th and 21st centuries. Hum Reprod Update 2022;29(2):157–76. DOI:10.1093/humupd/dmac035; Мужское бесплодие. Клинические рекомендации. Российское общество урологов, 2021. Доступно по: https://cr.minzdrav.gov.ru/recomend/5_2 (дата обращения: 02.11.2022). .; Лебедев Г.С., Голубев Н.А., Шадеркин И.А. и др. Мужское бесплодие в Российской Федерации: статистические данные за 2000–2018 годы. Экспериментальная и клиническая урология 2019;(4):4–12. DOI:10.29188/2222-8543-2019-11-4-4-12; Andrology. Male reproductive health and dysfunction. Ed. by E. Nieschlag, H.M. Behre, S. Nieschlag. Berlin: Springer Verlag, 2010. 629 р.; Jung J.H., Seo J.T. Empirical medical therapy in idiopathic male infertility: promise or panacea? Clin Exp Reprod Med 2014;41(3):108–14. DOI:10.5653/cerm.2014.41.3.108; Gundewar T., Kuchakulla M., Ramasamy R. A paradoxical decline in semen parameters in men treated with clomiphene citrate: a systematic review. Andrologia 2021;53(1):e13848. DOI:10.1111/and.13848; Huijben M., Huijsmans R.L.N., Lock M.T.W.T. et al. Clomiphene citrate for male infertility: a systematic review and meta-analysis. Andrology 2023;11(6):987–96. DOI:10.1111/andr.13388; Schlegel P.N., Sigman M., Collura B. et al. Diagnosis and treatment of infertility in men: AUA/ASRM Guideline PART II. J Urol 2021;205(1):44–51. DOI:10.1097/JU.0000000000001520; EAU guidelines on sexual and reproductive health. European Association of Urology Arnhem, Netherlands. Available at: https://uroweb.org/guidelines/sexual-and-reproductive-health/chapter/male-infertility (accessed: 02.11.2022).; Rastrelli G., Corona G., Mannucci E., Maggi M. Factors affecting spermatogenesis upon gonadotropin-replacement therapy: a meta-analytic study. Andrology 2014;2(6):794–808. DOI:10.1111/andr.262; Dostalova P., Zatecka E., Dvorakova-Hortova K. Of oestrogens and sperm: a review of the roles of oestrogens and oestrogen receptors in male reproduction. Int J Mol Sci 2017;18(5):904. DOI:10.3390/ijms18050904; Kelch R.P., Jenner M.R., Weinstein R. et al. Estradiol and testosterone secretion by human, simian, and canine testes, in males with hypogonadism and in male pseudohermaphrodites with the feminizing testes syndrome. J Clin Investig 1972;51(4):824–30. DOI:10.1172/JCI106877; Lubahn D.B., Moyer J.S., Golding T.S. et al. Alteration of reproductive function but not prenatal sexual development after insertional disruption of the mouse estrogen receptor gene. Proc Natl Acad Sci U S A 1993;90(23):11162–6. DOI:10.1073/pnas.90.23.11162; Hirata S., Shoda T., Kato J., Hoshi K. Isoform/variant mRNAs for sex steroid hormone receptors in humans. Trends Endocrinol Metab 2003;14(3):124–9. DOI:10.1016/S1043-2760(03)00028-6; Dumasia K., Kumar A., Kadam L., Balasinor N.H. Effect of estrogen receptor-subtype-specific ligands on fertility in adult male rats. J Endocrinol 2015;225(3):169–80. DOI:10.1530/JOE-15-0045; Dumasia K., Kumar A., Deshpande S. et al. Differential roles of estrogen receptors, ESR1 and ESR2, in adult rat spermatogenesis. Mol Cell Endocrinol 2016;428:89–100. DOI:10.1016/j.mce.2016.03.024; Upadhyay R.D., Kumar A.V., Ganeshan M., Balasinor N.H. Tubulobulbar complex: cytoskeletal remodeling to release spermatozoa. Reprod Biol Endocrinol 2012;10:27. DOI:10.1186/1477-7827-10-27; Lucas T.F.G., Lazari M.F.M., Porto C.S. Differential role of the estrogen receptors ESR1 and ESR2 on the regulation of proteins involved with proliferation and differentiation of Sertoli cells from 15-day-old rats. Mol Cell Endocrinol 2014;382(1):84–96. DOI:10.1016/j.mce.2013.09.015; Yanagimachi R. Mammalian fertilization. In: The physiology of reproduction. Ed. by E. Knobil, J.D. Neill. New York: Raven Press, 1994. Pp. 189–317.; Ded L., Sebkova N., Cerna M. et al. In vivo exposure to 17βestradiol triggers premature sperm capacitation in cauda epididymis. Reproduction 2013;145(3):255–63. DOI:10.1530/REP-12-0472; Справочник Видаль. Инструкция по применению препарата Летрозол. Доступно по: https://www.vidal.ru/drugs/letrozole__29301?ysclid=lddgkcwi22729917312 (дата обращения: 12.04.2022); Справочник Видаль. Инструкция по применению препарата Анастрозол. Доступно по: https://www.vidal.ru/drugs/anastrozole-1?ysclid=lddgpk4z9z942508447 (дата обращения 12.04.2022); Vigersky R.A., Glass A.R. Effects of delta 1-testolactone on the pituitary-testicular axis in oligospermic men. J Clin Endocrinol Metab 1981;52(5):897–902. DOI:10.1210/jcem-52-5-897; Pavlovich C.P., King P., Goldstein M., Schlegel P.N. Evidence of a treatable endocrinopathy in infertile men. J Urol 2001;165(3):837–41. DOI:10.1016/S0022-5347(05)66540-8; Shuling L., Sie Kuei M.L., Saffari S.E. et al. Do men with normal testosterone-oestradiol ratios benefit from letrozole for the treatment of male infertility? Reprod Biomed Online 2019;38(1):39–45. DOI:10.1016/j.rbmo.2018.09.016; Schlegel P.N. Aromatase inhibitors for male infertility. Fertil Steril 2012;98(6):1359–62. DOI:10.1016/j.fertnstert.2012.10.023; Clark R.V., Sherins R.J. Treatment of men with idiopathic oligozoospermic infertility using the aromatase inhibitor, testolactone. Results of a double-blinded, randomized, placebo-controlled trial with crossover. J Androl 1989;10(3):240–7. DOI:10.1002/j.1939-4640.1989.tb00094.x; Del Giudice F., Busetto G.M., De Berardinis E. et al. A systematic review and meta-analysis of clinical trials implementing aromatase inhibitors to treat male infertility. Asian J Androl 2020;22(4):360–7. DOI:10.4103/aja.aja_101_19; Guo B., Li J.J., Ma Y.L. et al. Efficacy and safety of letrozole or anastrozole in the treatment of male infertility with low testosterone-estradiol ratio: a meta-analysis and systematic review. Andrology 2022;10(5):894–909. DOI:10.1111/andr.13185; Tian F., Diao R., Zhen W. Clinical treatment on the low testosterone estradiol ratio of patients with oligoasthenospermia. Chinese J Coal Ind Med 2016.; Kooshesh L., Bahmanpour S., Zeighami S., Nasr-Esfahani M.H. Effect of Letrozole on sperm parameters, chromatin status and ROS level in idiopathic oligo/astheno/teratozoospermia. Reprod Biol Endocrinol 2020;18(1):47. DOI:10.1186/s12958-020-00591-2; Murad M. Aromatase inhibitors in infertile patients: effects on seminal parameters, serum and seminal plasma testosterone levels, and estradiol levels during short-term follow-up. Turk J Med Sci 2009;39(4):519–24.; Павлова З.Ш., Аметов А.С., Голодников И.И., Камалов А.А. Нормализация баланса эстрогенов и андрогенов у мужчин с гиперэстрогенией при использовании препарата класса ингибиторов ароматазы – летрозол. Акушерство, гинекология и репродукция. 2022;16(1):16–28. DOI:10.17749/2313-7347/ob.gyn.rep.2022.282; González-Marín C., Gosálvez J., Roy R. Types, causes, detection and repair of DNA fragmentation in animal and human sperm cells. Int J Mol Sci 2012;13(11):14026–52. DOI:10.3390/ijms131114026; Andrabi S. Mammalian sperm chromatin structure and assessment of DNA fragmentation. J Assist Reprod Genet 2007;24(12):561–9. DOI:10.1007/s10815-007-9177-y; Nili H.A., Mozdarani H., Aleyasin A. Correlation of sperm DNA damage with protamine deficiency in Iranian subfertile men. Reprod Biomed Online 2009;18(4):479–85. DOI:10.1016/s1472-6483(10)60123-x; Roca J. Topoisomerase II: a fitted mechanism for the chromatin landscape. Nucleic Acids Res 2008;37(3):721–30. DOI:10.1093/nar/gkn994; Bakshi R., Galande S., Bali P. et al. Developmental and hormonal regulation of type II DNA topoisomerase in rat testis. J Mol Endocrinol 2001;26(3):193–206. DOI:10.1677/jme.0.0260193; Muratori M., Baldi E. Effects of FSH on sperm DNA fragmentation: review of clinical studies and possible mechanisms of action. Front Endocrinol (Lausanne) 2018;9:734. DOI:10.3389/fendo.2018.00734; Colacurci N., De Leo V., Ruvolo G. et al. Recombinant FSH improves sperm DNA damage in male infertility: a phase II clinical trial. Front Endocrinol (Lausanne) 2018;9:383. DOI:10.3389/fendo.2018.00383; Ruvolo G., Fornaro F., Bosco L. et al. r-FSH administration in idiopathic oligoasthenoteratozoospermic patients (iOAT) reduces the apoptotic rate in sperm cells. Fertil Steril 2009;92(3):S72–S3. DOI:10.1016/j.fertnstert.2009.07.281; Витязева И.И., Боголюбова С.В., Брагина Е.Е., Арифулин Е.А. Возможность получения сперматозоидов у мужчин с немозаичной формой синдрома Клайнфельтера в программах экстракорпорального оплодотворения. Обзор литературы и описание случая. Андрология и генитальная хирургия 2014;15(3):16–25. DOI:10.17650/2070-9781-2014-3-16-25; Почерников Д.Г., Стрельников А.И., Винокуров Е.Ю. Способ коррекции гиперэстрадиолемии и нормогонадотропного гипогонадизма у мужчин. Патент РФ № RU 2474424 C2 от 10.02.2013. Доступно по: https://patents.s3.yandex.net/RU2474424C2_20130210.pdf; Гамидов С.И., Тажетдинов О.Х. Способ прогнозирования эффективности ингибиторов ароматазы в лечении идиопатического бесплодия у мужчин с ожирением. Патент РФ № RU 2503008 C1 от 27.12.2013. Доступно по: https://patents.s3.yandex.net/RU2503008C1_20131227.pdf; Тажетдинов О.Х. Особенности диагностики и лечения бесплодия у мужчин с ожирением. Автореф. дис. … канд. мед. наук. М., 2012. 27 с.; Ожирение и избыточный вес. Информационный бюллетень ВОЗ. Доступно по: https://www.who.int/ru/news-room/fact-sheets/detail/obesity-and-overweight (дата обращения: 26.09.2021).; Епанчинцева Е.А., Селятицкая В.Г., Свиридова М.А., Лутов Ю.В. Медико-социальные факторы риска бесплодия у мужчин. Андрология и генитальная хирургия 2016;17(3):47–53. DOI:10.17650/2070-9781-2016-17-3-47-53; Ng M., Fleming T., Robinson M. et al. Global, regional, and national prevalence of overweight and obesity in children and adults during 1980–2013: a systematic analysis for the global burden of disease study 2013. Lancet 2014;384(9945):766–81. DOI:10.1016/S0140-6736(14)60460-8; Campbell J.M., Lane M., Owens J.A., Bakos H.W. Paternal obesity negatively affects male fertility and assisted reproduction outcomes: a systematic review and meta-analysis. Reprod Biomed Online 2015;31(5):593–604. DOI:10.1016/j.rbmo.2015.07.012; Aly J.M., Polotsky A.J. Paternal diet and obesity: effects on reproduction. Semin Reprod Med 2017;35(4):313–7. DOI:10.1055/s-0037-1602593; Campbell J.M., McPherson N.O. Influence of increased paternal BMI on pregnancy and child health outcomes independent of maternal effects: a systematic review and meta-analysis. Obes Res Clin Pract 2019;13(6):511–21. DOI:10.1016/j.orcp.2019.11.003; Bieniek J.M., Kashanian J.A., Deibert C.M. et al. Influence of increasing body mass index on semen and reproductive hormonal parameters in a multi-institutional cohort of subfertile men. Fertil Steril 2016;106(5):1070–5. DOI:10.1016/j.fertnstert.2016.06.041; Sermondade N., Faure C., Fezeu L. et al. BMI in relation to sperm count: an updated systematic review and collaborative meta-analysis. Hum Reprod Update 2013;19(3):221–31. DOI:10.1093/humupd/dms050; Guo D., Wu W., Tang Q. et al. The impact of BMI on sperm parameters and the metabolite changes of seminal plasma concomitantly. Oncotarget 2017;8(30):48619–34. DOI:10.18632/oncotarget.14950; Sepidarkish M., Maleki-Hajiagha A., Maroufizadeh S. et al. The effect of body mass index on sperm DNA fragmentation: a systematic review and meta-analysis. Int J Obes (Lond) 2020;44(3):549–58. DOI:10.1038/s41366-020-0524-8; Le W., Su S.H., Shi L.H. et al. Effect of male body mass index on clinical outcomes following assisted reproductive technology: a meta-analysis. Andrologia 2016;48(4):406–24. DOI:10.1111/and.12461; Leisegang K., Henkel R., Agarwal A. Obesity and metabolic syndrome associated with systemic inflammation and the impact on the male reproductive system. Am J Reprod Immunol 2019;82(5):e13178. DOI:10.1111/aji.13178; https://agx.abvpress.ru/jour/article/view/704

  16. 16
  17. 17
    Academic Journal

    Source: Andrology and Genital Surgery; Том 24, № 1 (2023); 19-23 ; Андрология и генитальная хирургия; Том 24, № 1 (2023); 19-23 ; 2412-8902 ; 2070-9781

    File Description: application/pdf

    Relation: https://agx.abvpress.ru/jour/article/view/632/499; Lomboy J.R., Coward R.M. The Varicocele: clinical presentation, evaluation, and surgical management. Semin Intervent Radiol 2016;33(3):163–9. DOI:10.1055/s-0036-1586143; Marte A. The history of varicocele: from antiquity to the modern ERA. Int Braz J Urol 2018;44(3):563–76. DOI:10.1590/S1677-5538.IBJU.2017.0386; Watanabe M., Nagai A., Kusumi N. et al. Minimal invasiveness and effectivity of subinguinal microscopic varicocelectomy: a comparative study with retroperitoneal high and laparoscopic approaches. Int J Urol 2005;12(10):892–8. DOI:10.1111/j.1442-2042.2005.01142.x; Ghanem H., Anis T., El-Nashar A., Shamloul R. Subinguinal microvaricocelectomy versus retroperitoneal varicocelectomy: comparative study of complications and surgical outcome. Urology 2004;64(5):1005–9. DOI:10.1016/j.urology.2004.06.060; Cayan S., Kadioglu T.C., Tefekli A. et al. Comparison of results and complications of high ligation surgery and microsurgical high inguinal varicocelectomy in the treatment of varicocele. Urology 2000;55(5):750–4. DOI:10.1016/s0090-4295(99)00603-2; Cayan S., Shavakhabov S., Kadioğlu A. Treatment of palpable varicocele in infertile men: a meta-analysis to define the best technique. J Androl 2009;30(1):33–40. DOI:10.2164/jandrol.108.005967; Diegidio P., Jhaveri J.K., Ghannam S. et al. Review of current varicocelectomy techniques and their outcomes. BJU Int 2011;108(7):1157–72. DOI:10.1111/j.1464410X.2010.09959.x; Marmar J.L., DeBenedictis T.J., Praiss D. The management of varicoceles by microdissection of the spermatic cord at the external inguinal ring. Fertil Steril 1985;43(4):583–8. DOI:10.1016/s0015-0282(16)48501-8; Goldstein M., Gilbert B.R., Dicker A.P. et al. Microsurgical inguinal varicocelectomy of the testis: an artery and lymphatic sparing technique. J Urol 1992;148(6):1808–11. DOI:10.1016/S0022-5347(17)37035-0; Wein A.J., Kavoussi L.R., Partin A.W., Peters C.A. Campbell-Walsh Urology. 11th edn. Philadelphia: Saunders, 2016. 4168 p.; Ахвледиани Н.Д., Рева И.А., Чернушенко А.С. и др. Варикоцеле: роль в развитии мужского бесплодия и методики хирургического лечения (обзор литературы). Урология 2020;4:111–8. DOI:10.18565/urology.2020.4.111-118; Persad E., O’Loughlin C.A., Kaur S. et al. Surgical or radiological treatment for varicoceles in subfertile men. Cochrane Database Syst Rev 2021;4(4):CD000479. DOI:10.1002/14651858.CD000479.pub6; Harrison R.G. The distribution of the vasal and cremasteric arteries to the testis and their functional importance. J Anat 1949;83(Pt 3):267–84.; Woznitzer M., Roth J.A. Optical magnification and Doppler ultrasound probe for varicocelectomy. Urology 1983;22(1):24–6. DOI:10.1016/0090-4295(83)90339-4; Ramadan A.E.-S., Eldemiry M.I.M., Ramadan A.E.A.E.-S. Doppler-controlled varicocelectomy. Br J Urol 1984;56(4):432–3. DOI:10.1111/j.1464-410x.1984.tb05838.x; Cocuzza M., Pagani R., Coelho R. et al. The systematic use of intraoperative vascular Doppler ultrasound during microsurgical subinguinal varicocelectomy improves precise identification and preservation of testicular blood supply. Fertil Steril 2010;93(7):2396–9. DOI:10.1016/j.fertnstert.2009.01.088; Shindel A.W., Yan Y., Naughton C.K. Does the number and size of veins ligated at left-sided microsurgical subinguinal varicocelectomy affect semen analysis outcomes? Urology 2007;69(6):1176–80. DOI:10.1016/j.urology.2007.01.086; Guo L., Sun W., Shao G. et al. Outcomes of microscopic subinguinal varicocelectomy with and without the assistance of Doppler ultrasound: a randomized clinical trial. Urology 2015;86(5):922–8. DOI:10.1016/j.urology.2015.08.002; Niederberger C. Re: outcomes of microscopic subinguinal varicocelectomy with and without the assistance of Doppler ultrasound: a randomized clinical trial. J Urol 2016;195(6):1857. DOI:10.1016/j.juro.2016.03.035; Owen R.C., McCormick B.J., Figler B.D., Coward R.M. A review of varicocele repair for pain. Transl Androl Urol 2017;6(S1):S20–S29. DOI:10.21037/tau.2017.03.36; Lv K.L., Zhang Y.D., Zhuang J.T. et al. Subinguinal microsurgical varicocelectomy with intraoperative microvascular Doppler ultrasound leads to the pain-free outcome after surgery. J Xray Sci Technol 2017;25(5):839–46. DOI:10.3233/XST-17247; Juho Y.-C., Wu S.-T., Kao C.-C. et al. Anatomic mapping of the internal spermatic vein via subinguinal varicocelectomy with intraoperative vascular Doppler ultrasound. J Chin Med Assoc 2019;82(2):115–9. DOI:10.1097/JCMA.0000000000000012; Özkaptan O., Balaban M., Sevinc C. et al. O. Impact of intraoperative doppler ultrasound assistance during microsurgical varicocelectomy on operative outcome and sperm parameters. Andrologia 2020;52(7):e13641. DOI:10.1111/and.13641; https://agx.abvpress.ru/jour/article/view/632

  18. 18
    Academic Journal

    Source: Andrology and Genital Surgery; Том 24, № 3 (2023); 75-81 ; Андрология и генитальная хирургия; Том 24, № 3 (2023); 75-81 ; 2412-8902 ; 2070-9781

    File Description: application/pdf

    Relation: https://agx.abvpress.ru/jour/article/view/685/535; Ferlin A., Raicu F., Gatta V. et al. Male infertility: role of genetic background. Reprod Biomed Online 2007;14(6):734–45. DOI:10.1016/s1472-6483(10)60677-3; Brugh V.M. 3rd, Matschke H.M., Lipshultz L.I. Male factor infertility. Endocrinol Metab Clin North Am 2003;32(3):689–707. DOI:10.1016/s0889-8529(03)00047-1; Котов С.В., Ирицян М.М., Бадаква Г.В. Результаты применения комплекса ББЕСТФертил у пациентов после микрохирургической подпаховой варикоцелэктомии (операции Мармара). Урология 2022;1:50–4. DOI:10.18565/urology.2022.1.50-54; Diegidio P., Jhaveri J.K., Ghannam S. et al. Review of current varicocelectomy techniques and their outcomes. BJU Int 2011;108(7):1157–72. DOI:10.1111/j.1464-410X.2010.09959.x; Agarwal A., Roychoudhury S., Bjugstad K.B., Cho C.L. Oxidation-reduction potential of semen: what is its role in the treatment of male infertility? Ther Adv Urol 2016;8(5):302–18. DOI:10.1177/1756287216652779; Корнеев И.А. Мужское бесплодие при оксидативном стрессе: пути решения проблемы. Урология 2022;1:102–8. DOI:10.18565/urology.2022.1.102-108; Agarwal A., Gupta S., Sharma R. Measurement of DNA fragmentation in spermatozoa by TUNEL assay using bench top flow cytometer. In: Andrological evaluation of male infertility. Ed. by A. Agarwal, S. Gupta, R. Sharma. Springer, Cham, 2016. P. 181–203. DOI:10.1007/978-3-319-26797-5_24; Kovac J.R., Pastuszak A.W., Lamb D.J. The use of genomics, proteomics, and metabolomics in identifying biomarkers of male infertility. Fertil Steril 2013;99(4):998–1007. DOI:10.1016/j.fertnstert.2013.01.111; Тулаганов К.А., Садриддинов Х.Н., Ибрагимов У.К. Фертильность мужчин и биохимический состав спермоплазмы. Андрология и генитальная хирургия 2009;10(2):94.; Lenzi A., Picardo M., Gandini L., Dondero F. Lipids of the sperm plasma membrane: from polyunsaturated fatty acids considered as markers of sperm function to possible scavenger therapy. Hum Reprod Update 1996;2(3):246–56. DOI:10.1093/humupd/2.3.246; Koelmel J.P., Kroeger N.M., Ulmer C.Z. et al. LipidMatch: an automated workflow for rule-based lipid identification using untargeted high-resolution tandem mass spectrometry data. BMC Bioinformatics 2017;18(1):331. DOI:10.1186/s12859-017-1744-3; Team R.C. R: a language and environment for statistical computing. R Foundation for Statistical Computing, Vienna, Austria. Available at: https://www.r-project.org/; Team R.C. RStudio: Integrated Development for R. Available at: http://www.rstudio.com/; Poulos A., Voglmayr J.K., White I.G. Phospholipid changes in spermatozoa during passage through the genital tract of the bull. Biochim Biophys Acta 1973;306(2):194–202. DOI:10.1016/0005-2760(73)90225-7; Parks J.E., Graham J.K. Effects of cryopreservation procedures on sperm membranes. Theriogenology 1992;38(2):209–22. DOI:10.1016/0093-691x(92)90231-f; Watson P.F. The causes of reduced fertility with cryopreserved semen. Anim Reprod Sci 2000;60–61:481–92. DOI:10.1016/s0378-4320(00)00099-3; Evans H.C., Dinh T.T.N., Ugur M.R. et al. Lipidomic markers of sperm cryotolerance in cattle. Sci Rep 2020;10(1):20192. DOI:10.1038/s41598-020-77089-9; Amaral A., Castillo J., Estanyol J.M. et al. Human sperm tail proteome suggests new endogenous metabolic pathways. Mol Cell Proteomics 2013;12(2):330–42. DOI:10.1074/mcp.M112.020552; Esmaeili V., Shahverdi A.H., Moghadasian M.H., Alizadeh A.R. Dietary fatty acids affect semen quality: a review. Andrology 2015;3(3):450–61. DOI:10.1111/andr.12024; Zerbinati C., Caponecchia L., Rago R. et al. Fatty acids profiling reveals potential candidate markers of semen quality. Andrology 2016;4(6):1094–101. DOI:10.1111/andr.12236; Tavilani H., Doosti M., Nourmohammadi I. et al. Lipid composition of spermatozoa in normozoospermic and asthenozoospermic males. Prostaglandins Leukot Essent Fatty Acids 2007;77(1):45–50. DOI:10.1016/j.plefa.2007.07.001; Lopalco P., Vitale R., Cho Y.S. et al. Alteration of cholesterol sulfate/seminolipid ratio in semen lipid profile of men with oligoasthenozoospermia. Front Physiol 2019;10:1344. DOI:10.3389/fphys.2019.01344; Гамидов С.И., Шатылко Т.В., Тамбиев А.Х. и др. Липидомный профиль семенной плазмы при необструктивной азооспермии с остановкой созревания сперматозоидов. Вестник урологии 2021;9(4):30–9. DOI:10.21886/2308-6424-2021-9-4-30-39; Tasseva G., Bai H.D., Davidescu M. et al. Phosphatidylethanolamine deficiency in Mammalian mitochondria impairs oxidative phosphorylation and alters mitochondrial morphology. J Biol Chem 2013;288(6):4158–73. DOI:10.1074/jbc.M112.434183; Jiang F., Ryan M.T., Schlame M. et al. Absence of cardiolipin in the crd1 null mutant results in decreased mitochondrial membrane potential and reduced mitochondrial function. J Biol Chem 2000;275(29):22387–94. DOI:10.1074/jbc.M909868199; Horvath S.E., Daum G. Lipids of mitochondria. Prog Lipid Res 2013;52(4):590–614. DOI:10.1016/j.plipres.2013.07.002; Mejia E.M., Hatch G.M. Mitochondrial phospholipids: role in mitochondrial function. J Bioenerg Biomembr 2016;48(2):99–112. DOI:10.1007/s10863-015-9601-4; Ren M., Phoon C.K., Schlame M. Metabolism and function of mitochondrial cardiolipin. Prog Lipid Res 2014;55:1–16. DOI:10.1016/j.plipres.2014.04.001; Vance J.E. Phospholipid synthesis and transport in mammalian cells. Traffic 2015;16(1):1–18. DOI:10.1111/tra.12230; Bogdanov M., Dowhan W. Lipid-assisted protein folding. J Biol Chem 1999;274(52):36827–30. DOI:10.1074/jbc.274.52.36827; https://agx.abvpress.ru/jour/article/view/685

  19. 19
    Academic Journal

    Contributors: The work was carried out within the framework of the project “Multicenter research bioresource collection “Human Reproductive Health” No. 15.BRK.21.0008 of the Ministry of Science and Higher Education of the Russian Federation., Работа выполнена в рамках проекта «Многоцентровая исследовательская биоресурсная коллекция “Репродуктивное здоровье человека”» № 15.БРК.21.0008 Министерства науки и высшего образования Российской Федерации.

    Source: Andrology and Genital Surgery; Том 23, № 4 (2022); 81-89 ; Андрология и генитальная хирургия; Том 23, № 4 (2022); 81-89 ; 2412-8902 ; 2070-9781

    File Description: application/pdf

    Relation: https://agx.abvpress.ru/jour/article/view/619/491; Nielsen J., Wohlert M. Chromosome abnormalities found among 34,910 newborn children: results from a 13-year incidence study in Arhus, Denmark. Hum Genet 1991;87(1):81–3. DOI:10.1007/BF01213097; McKinlay Gardner R.J. et al. Robertsonian translocations. In: Gardner and sutherland’s chromosome abnormalities and genetic counseling. Ed. by R.J. McKinlay Gardner, D.J. Armour. 5th edn. Oxford monographs on medical genetics. New York: Oxford Academic, 2018. (online edn.). DOI:10.1093/med/9780199329007.003.0007; De Braekeleer M., Dao T.N. Cytogenetic studies in male infertility: a review. Hum Reprod 1991;6(2):245–50. PMID: 2056021.; Van Assche E., Bonduelle M., Tournaye H. et al. Cytogenetics of infertile men. Hum Reprod 1996;11(Suppl 4):1–24. DOI:10.1093/humrep/11.suppl_4.1; Ogur G., Van Assche E., Vegetti W. et al. Chromosomal segregation in spermatozoa of 14 Robertsonian translocation carriers. Mol Hum Reprod 2006;12(3):209–15. DOI:10.1093/molehr/gah253; Luciani J.M., Guichaoua M.R., Mattei A., Morazzani M.R. Pachytene analysis of a man with a 13q;14q translocation and infertility. Behavior of the trivalent and nonrandom association with the sex vesicle. Cytogenet Cell Genet 1984;38(1):14–22. DOI:10.1159/000132023; Rosenmann A., Wahrman J., Richler C. et al. Meiotic association between the XY chromosomes and unpaired autosomal elements as a cause of human male sterility. Cytogenet Cell Genet 1985;39(1):19–29. DOI:10.1159/000132098; Godo A., Blanco J., Vidal F. et al. Altered segregation pattern and numerical chromosome abnormalities interrelate in spermatozoa from Robertsonian translocation carriers. Reprod Biomed Online 2015;31(1):79–88. DOI:10.1016/j.rbmo.2015.04.003; Kovaleva N.V. Examination of rates and spectrums of robertsonian translocations in the general population and in patients with reproductive disorders. Russ J Genet 2018;54(4):489–93. DOI:10.1134/S1022795418040099; WHO laboratory manual for the examination of human semen and sperm-cervical mucus interaction. 4th edn. Cambridge: Cambridge University Press, 1999.; WHO laboratory manual for the examination and processing of human semen. 5th edn. Geneva, Swtizerland: WHO, 2010.; Antonelli A., Gandini L., Petrinelli P. et al. Chromosomal alterations and male infertility. J Endocrinol Invest 2000;23(10):677–83. DOI:10.1007/BF03343793; Ferfouri F., Selva J., Boitrelle F. et al. The chromosomal risk in sperm from heterozygous Robertsonian translocation carriers is related to the sperm count and the translocation type. Fertil Steril 2011;96(6):1337–43. DOI:10.1016/j.fertnstert.2011.09.008; Vozdova M., Oracova E., Kasikova K. et al. Balanced chromosomal translocations in men: relationships among semen parameters, chromatin integrity, sperm meiotic segregation and aneuploidy. J Assist Reprod Genet 2013;30(3):391–405. DOI:10.1007/s10815-012-9921-9; Pastuszek E., Kiewisz J., Kulwikowska P.M. et al. Sperm parameters and DNA fragmentation of balanced chromosomal rearrangements carriers. Folia Histochem Cytobiol 2015;53(4):314–21. DOI:10.5603/fhc.a2015.0032; Lamotte A., Martinez G., Devillard F. et al. Is sperm FISH analysis still useful for Robertsonian translocations? Meiotic analysis for 23 patients and review of the literature. Basic Clin Androl 2018;28:5. DOI:10.1186/s12610-018-0069-z; Mayeur A., Ahdad N., Hesters L. et al. Chromosomal translocations and semen quality: a study on 144 male translocation carriers. Reprod Biomed Online 2019;38(1):46–55. DOI:10.1016/j.rbmo.2018.10.003; Gatimel N., Moreau J., Parinaud J., Léandri R.D. Sperm morphology: assessment, pathophysiology, clinical relevance, and state of the art in 2017. Andrology 2017;5(5):845–62. DOI:10.1111/andr.12389; Олефир Ю.В., Монаков Д.М. Клиническое значение морфологии сперматозоидов в выборе метода лечения мужского бесплодия. Экспериментальная и клиническая урология 2021;14(3):127–32. DOI:10.29188/2222-8543-2021-14-3-127-132; Гамидов С.И., Овчинников Р.И., Попова А.Ю. и др. Эффективность программ вспомогательных репродуктивных технологий в зависимости от характера изменений спермограммы. Андрология и генитальная хирургия 2018;19(2):82–7. DOI:10.17650/2070-9781-2018-19-2-82-87; Гамидов С.И., Овчинников Р.И., Попова А.Ю. и др. Роль мужского фактора бесплодия в программе вспомогательных репродуктивных технологий (обзор литературы). Андрология и генитальная хирургия 2017;18(3):28–36. DOI:10.17650/2070- 9781-2017-18-3-28-36; Zhu S., Zhu Y., Zhang F. et al. FISH analysis of numerical chromosomal abnormalities in the sperm of robertsonian translocation der(13; 14)(q10;q10) carriers. Front Genet 2022;13:1010568. DOI:10.3389/fgene.2022.1010568; Johannisson R., Schwinger E., Wolff H.H. et al. The effect of 13;14 Robertsonian translocations on germ-cell differentiation in infertile males. Cytogenet Cell Genet 1993;63(3):151–5. DOI:10.1159/000133524; Page S.L., Shin J.C., Han J.Y. et al. Breakpoint diversity illustrates distinct mechanisms for Robertsonian translocation formation. Hum Mol Genet 1996;5(9):1279–88. DOI:10.1093/hmg/5.9.1279; Morel F., Douet-Guilbert N., Le Bris M.J., Amice V. et al. Chromosomal abnormalities in couples undergoing intracytoplasmic sperm injection. A study of 370 couples and review of the literature. Int J Androl 2004;27(3):178–82. DOI:10.1111/j.1365-2605.2004.00472.x; https://agx.abvpress.ru/jour/article/view/619

  20. 20
    Academic Journal

    Contributors: The work was carried out within the framework of the state assignment for Institute of Immunology and Physiology (topic № АААА-А18-118020590108-7)., Работа выполнена в рамках госзадания ФГБУН ИИФ УрО РАН (тема № АААА-А18-118020590108-7).

    Source: Obstetrics, Gynecology and Reproduction; Vol 15, No 6 (2021); 715-725 ; Акушерство, Гинекология и Репродукция; Vol 15, No 6 (2021); 715-725 ; 2500-3194 ; 2313-7347

    File Description: application/pdf

    Relation: https://www.gynecology.su/jour/article/view/1184/974; Barati E., Nikzad H., Karimian M. Oxidative stress and male infertility: current knowledge of pathophysiology and role of antioxidant therapy in disease management. Cell Mol Life Sci. 2020;77(1):93–113. https://doi.org/10.1007/s00018-019-03253-8.; Shiraishi K., Matsuyama H. Effects of medical comorbidity on male infertility and comorbidity treatment on spermatogenesis. Fertil Steril. 2018;110(6):1006–11.e2. https://doi.org/10.1016/j.fertnstert.2018.07.002.; Потехина Е.С., Михайлюк Е.В., Непомнящих А.С. Спермограмма как инструмент оценки мужской фертильности. Научное обозрение. 2020;(1):11–4. https://doi.org/10.17513/srms.1093.; Жуков О.Б., Евдокимов В.В., Брагина Е.Е. Улучшение качества жизни и морфофункциональных характеристик сперматозоидов у мужчин с хроническим абактериальным простатитом и программы прегравидарной подготовки к отцовству. Андрология и генитальная хирургия. 2017;18(1):102–8. https://doi.org/10.17650/2070-9781-2017-18-1-102-108.; Авадиева Н.Э. Применение ДНК фрагментации спермы в андрологической практике. Вестник урологии. 2019;7(1):7–11. https://doi.org/10.21886/2308-6424-2019-7-1-7-11.; Barratt C.L.R., Björndahl L., De Jonge C.J. et al. The diagnosis of male infertility: an analysis of the evidence to support the development of global WHO guidance-challenges and future research opportunities. Hum Reprod Update. 2017;23(6):660–80. https://doi.org/10.1093/humupd/dmx021.; Busetto G.M., Del Giudice F., Virmani A. et al. Body mass index and age correlate with antioxidant supplementation effects on sperm quality: Post hoc analyses from a double-blind placebo-controlled trial. Andrologia. 2020;52(3):e13523. https://doi.org/10.1111/and.13523.; Agarwal A., Rana M., Qiu E. et al. Role of oxidative stress, infection and inflammation in male infertility. Andrologia. 2018;50(11):e13126. https://doi.org/10.1111/and.13126.; Кузнецова Н.Н., Шамин М.В., Фарбирович В.Я. и др. Оценка репродуктивного потенциала молодых мужчин Кузбасса и роль хронического воспаления органов репродуктивного тракта как фактора снижения фертильности эякулята. Омский научный вестник. 2015;(1):71–3.; Овчинников Р.И. Гамидов С.И., Попова А.Ю. и др. Причины репродуктивных потерь у мужчин – фрагментация ДНК сперматозоидов. РМЖ. 2015;23(11):634–8.; Esteves S.C., Agarwal A. Afterword to varicocele and male infertility: current concepts and future perspectives. Asian J Androl. 2016;18(2):319–22. https://doi.org/10.4103/1008-682X.172820.; Alsaikhan B., Alrabeeah K., Delouya G., Zini A. Epidemiology of varicocele. Asian J Androl. 2016;18(2):179–81. https://doi.org/10.4103/1008-682X.172640.; Jensen C.F.S., Østergren P., Dupree J.M. et al. Varicocele and male infertility. Nat Rev Urol. 2017;14(9):523–33. https://doi.org/10.1038/nrurol.2017.98.; Majzoub A., Esteves S.C., Gosálvez J., Agarwal A. Specialized sperm function tests in varicocele and the future of andrology laboratory. Asian J Androl. 2016;18(2):205–12. https://doi.org/10.4103/1008-682X.172642.; Kathrins M. Historical investigations into varicocele pathophysiology and sperm migration. Fertil Steril. 2018;109(1):75–6. https://doi.org/10.1016/j.fertnstert.2017.11.00.; Kruger T. Critical appraisal of conventional semen analysis in the context of varicocele. Asian J Androl. 2016;18(2):202–4. https://doi.org/10.4103/1008-682X.168691.; Santana V.P., Miranda-Furtado C.L., de Oliveira-Gennaro F.G., Dos Reis R.M. Genetics and epigenetics of varicocele pathophysiology: an overview. J Assist Reprod Genet. 2017;34(7):839–47. https://doi.org/10.1007/s10815-017-0931-5.; Brannigan R.E. Introduction: Varicoceles: a contemporary perspective. Fertil Steril. 2017;108(3):361–3. https://doi.org/10.1016/j.fertnstert.2017.07.1161.; Ефремов Е.А., Касатонова Е.В., Мельник Я.И., Симаков В.В. Позднее отцовство: обзор повреждающих эякулят механизмов, рисков и стратегий их преодоления. Эффективная фармакотерапия. 2016;(11):16–33.; Сулима А.Н., Литвинов В.В., Клименко П.М. и др. Особенности мужской инфертильности как единственного фактора бесплодия супружеской пары в клинике ВРТ. Экспериментальная и клиническая урология. 2019;(4):68–73. https://doi.org/10.29188/2222-8543-2019-11-4-68-73.; Кириленко Е.А., Онопко В.Ф. Окислительный стресс и мужская фертильность: современный взгляд на проблему. Бюллетень ВСНЦ СО РАМН. 2017;2(2):102–8.; Осадчук Л.В., Клещев М.А., Типисова Е.В., Осадчук А.В. Показатели сперматогенеза, гормонального и метаболического статуса у мужчин разных возрастных групп на Европейском севере России. Физиология человека. 2019;45(3):107–14. https://doi.org/10.1134/S0131164619020073.; Shridharani A., Owen R.C., Elkelany O.O., Kim E.D. The significance of clinical practice guidelines on adult varicocele detection and management. Asian J Androl. 2016;18(2):269–75. https://doi.org/10.4103/1008-682X.172641.; Kumar M., Selvam P., Agarwal A. Sperm and seminal plasma proteomics: molecular changes associated with varicocele-mediated male infertility. World J Mens Health. 2020;38(4):472–83. https://doi.org/10.5534/wjmh.190018.; Oliva A., Multigner L. Chronic epididymitis and grade III varicocele and their associations with semen characteristics in men consulting for couple infertility. Asian J Androl. 2018;20(4):360–5. https://doi.org/10.4103/aja.aja_78_17.; Руководство ВОЗ по исследованию и обработке эякулята человека. 5-е издание. Пер. с англ. Н.П. Макарова, под ред. Л.Ф. Курило. М.: Изд-во Капитал принт, 2012. 305 с.; Шмидт А.А., Замятин С.А., Гончар И.С., Коровин А.Е. Факторы риска развития мужской инфертильности. Клиническая патофизиология. 2019;25(4):56–60.; Belardin L.B., Del Giudice P.T., Camargo M. et al. Alterations in the proliferative/apoptotic equilibrium in semen of adolescents with varicocele. J Assist Reprod Genet. 2016;33(12):1657–64. https://doi.org/10.1007/s10815-016-0808-z.; Glassberg K.I. My indications for treatment of the adolescent varicocele (and why?). Transl Androl Urol. 2014;3(4):402–12. https://doi.org/10.3978/j.issn.2223-4683.2014.12.09.; Cho C.L., Esteves S.C., Agarwal A. Novel insights into the pathophysiology of varicocele and its association with reactive oxygen species and sperm DNA fragmentation. Asian J Androl. 2016;18(2):186–93. https://doi.org/10.4103/1008-682X.170441.; Zhang Y., Ma T., Su Z. et al. Varicoceles affect semen quality of infertile men in Southern China: A cross-sectional study of 5447 cases. Medicine (Baltimore). 2017;96(31):e7707. https://doi.org/10.1097/MD.0000000000007707.; Santana V.P., Miranda-Furtado C.L., Pedroso D.C.C. et al. The relationship among sperm global DNA methylation, telomere length, and DNA fragmentation in varicocele: a cross-sectional study of 20 cases. Syst Biol Reprod Med. 2019;65(2):95–104. https://doi.org/10.1080/19396368.2018.1557762.; Андреев Р.Ю., Раснер П.И., Малхасян В.А. и др. Варикоцеле – что нам о нем известно? Московский хирургический журнал. 2019;(5):24–31. https://doi.org/10.17238/issn2072-3180.2019.5.24-31.; Esteves S.C. Novel concepts in male factor infertility: clinical and laboratory perspectives. J Assist Reprod Genet. 2016;33(10):1319–35.; Martins A.D., Agarwal A. Oxidation reduction potential: a new biomarker of male infertility. Panminerva Med. 2019;61(2):108–17. https://doi.org/10.23736/S0031-0808.18.03529-2.; Tahamtan S., Tavalaee M., Izadi T. et al. Reduced sperm telomere length in individuals with varicocele is associated with reduced genomic integrity. Sci Rep. 2019;9(1):4336. https://doi.org/10.1038/s41598-019-40707-2.; Pallotti F., Paoli D., Carlini T. et al. Varicocele and semen quality: a retrospective case-control study of 4230 patients from a single centre. J Endocrinol Invest. 2018;41(2):185–92. https://doi.org/10.1007/s40618-017-0713-z.; Баженов И.В., Филиппова Е.С. Роль окислительного стресса в патогенезе мужского бесплодия. Эффективная фармакотерапия. 2018;(29):50–8.; Калинченко С.Ю., Тюзиков И.А. Окислительный стресс и мужское бесплодие – взаимосвязанные пандемии XXI в. Современные фармакотерапевтические возможности патогенетической коррекции нарушений сперматогенеза препаратами L-карнитина/ацетил-L-карнитина. Эффективная фармакотерапия. 2017;(22):6–19.; Корнеев И.А. Терапия мужского бесплодия: анализ исследований. Медицинский совет. 2019;(13):99–104. https://doi.org/10.21518/2079-701Х-2019-13-99-104.; https://www.gynecology.su/jour/article/view/1184