يعرض 1 - 7 نتائج من 7 نتيجة بحث عن '"Д. М. Монаков"', وقت الاستعلام: 0.77s تنقيح النتائج
  1. 1
    دورية أكاديمية

    المساهمون: The study was conducted within the scope of Moscow Government Grant No. 2212-5 (dated 2022), sponsoring the research centre of the S.P. Botkin State Clinical Hospital, Исследование проведено в рамках Гранта Правительства Москвы № 2212-5 (от 2022 года), спонсирующего исследовательский центр ГБУЗ ГКБ им. С.П. Боткина ДЗМ

    المصدر: Urology Herald; Том 11, № 2 (2023); 5-17 ; Вестник урологии; Том 11, № 2 (2023); 5-17 ; 2308-6424 ; 10.21886/2308-6424-2023-11-2

    وصف الملف: application/pdf

    العلاقة: https://www.urovest.ru/jour/article/view/715/486Test; Santucci RA, Joyce GF, Wise M. Male urethral stricture disease. J Urol. 2007;177(5):1667-74. DOI:10.1016/j.juro.2007.01.041; Alwaal A, Blaschko SD, McAninch JW, Breyer BN. Epidemiology of urethral strictures. Transl Androl Urol. 2014;3(2):209-13. DOI:10.3978/j.issn.2223-4683.2014.04.07; Palminteri E, Berdondini E, Verze P, De Nunzio C, Vitarelli A, Carmignani L. Contemporary urethral stricture characteristics in the developed world. Urology. 2013;81(1):191-6. DOI:10.1016/j.urology.2012.08.062; Baskin LS, Constantinescu SC, Howard PS, McAninch JW, Ewalt DH, Duckett JW, Snyder HM, Macarak EJ. Biochemical characterization and quantitation of the collagenous components of urethral stricture tissue. J Urol. 1993;150(2 Pt 2):642-7. DOI:10.1016/s0022-5347(17)35572-6; Wood DN, Andrich DE, Greenwell TJ, Mundy AR. Standing the test of time: the long-term results of urethroplasty. World J Urol. 2006;24(3):250-4. DOI:10.1007/s00345-006-0057-3; Altun E. MR Imaging of the Penis and Urethra. Magn Reson Imaging Clin N Am. 2019;27(1):139-150. DOI:10.1016/j.mric.2018.09.006; Frankiewicz M, Markiet K, Krukowski J, Szurowska E, Matuszewski M. MRI in patients with urethral stricture: a systematic review. Diagn Interv Radiol. 2021;27(1):134-146. DOI:10.5152/dir.2020.19515; Патент РФ на изобретение RU 2749869 C2/17.06.2021. Бюл. №17. Велиев Е.И., Богданов А.Б., Катибов М.И., Полякова А.С. Способ формирования анастомоза на вентральной поверхности уретры без пересечения спонгиозного тела при стриктурах бульбозного отдела уретры. Ссылка активна на 19.02.2023.; Bogdanov AB, Veliev EI, Sokolov EA, Metelev AY, Ivkin EE, Tomilov AA, Veliev RA, Marchenko VV, Monakov DM, Katibov MI, Afyouni AS, Furr J, Okhunov Z, Sabanegh E. Nontransecting Anastomotic Urethroplasty Via Ventral Approach Without Full Mobilization of the Corpus Spongiosum Dorsal Semicircumference. Urology. 2021;152:136-141. DOI:10.1016/j.urology.2020.10.074; Osman Y, El-Ghar MA, Mansour O, Refaie H, El-Diasty T. Magnetic resonance urethrography in comparison to retrograde urethrography in diagnosis of male urethral strictures: is it clinically relevant? Eur Urol. 2006;50(3):587-93; discussion 594. DOI:10.1016/j.eururo.2006.01.015; Sung DJ, Kim YH, Cho SB, Oh YW, Lee NJ, Kim JH, Chung KB, Moon du G, Kim JJ. Obliterative urethral stricture: MR urethrography versus conventional retrograde urethrography with voiding cystourethrography. Radiology. 2006;240(3):842-8. DOI:10.1148/radiol.2403050590; Oh MM, Jin MH, Sung DJ, Yoon DK, Kim JJ, Moon du G. Magnetic resonance urethrography to assess obliterative posterior urethral stricture: comparison to conventional retrograde urethrography with voiding cystourethrography. J Urol. 2010;183(2):603-7. DOI:10.1016/j.juro.2009.10.016; Банчик Э.Л., Митусов В.В., Домбровский В.И., Коган М.И. Динамическая магнитно-резонансная томография в диагностике заболеваний уретры у мужчин (комплекс импульсных последовательностей). Вестник рентгенологии и радиологии. 2013;(4):33-40.; Домбровский В.И., Коган М.И., Банчик Э.Л., Митусов В.В. Роль магнитно-резонансной томографии в диагностике стриктурной болезни уретры у мужчин. Урология. 2015;(2):24-30.; El-Ghar MA, Osman Y, Elbaz E, Refiae H, El-Diasty T. MR urethrogram versus combined retrograde urethrogram and sonourethrography in diagnosis of urethral stricture. Eur J Radiol. 2010;74(3):e193-8. DOI:10.1016/j.ejrad.2009.06.008; Rastogi R, Joon P, Pushkarna A, Agarwal A, Wani AM, Bhagat PK, Gupta Y, Sharma S, Das PK, Parashar S, Sinha P, Chaudhary M, Pratap V. Compar¬ative role of sonourethrography (SUG) and magnetic resonance urethrography (MRU) in anterior male urethral strictures. Ann Clin Lab Res. 2016; 4(4):1-4. DOI:10.21767/2386-5180.1000140; Mikolaj F, Karolina M, Oliwia K, Jakub K, Adam K, Mariusz B, Patrycja N, Marcin M. Retrograde urethrography, sonouretrography and magnetic resonance urethrography in evaluation of male urethral strictures. Should the novel methods become the new standard in radiological diagnosis of urethral stricture disease? Int Urol Nephrol. 2021;53(12):2423-2435. DOI:10.1007/s11255-021-02994-5; Jordan GH, Eltahawy EA, Virasoro R. The technique of vessel sparing excision and primary anastomosis for proximal bulbous urethral reconstruction. J Urol. 2007;177(5):1799-802. DOI:10.1016/j.juro.2007.01.036; Gur U, Jordan GH. Vessel-sparing excision and primary anastomosis (for proximal bulbar urethral strictures). BJU Int. 2008;101(9):1183-95. DOI:10.1111/j.1464-410X.2008.07619.x; Andrich DE, Mundy AR. Non-transecting anastomotic bulbar urethroplasty: a preliminary report. BJU Int. 2012;109(7):1090-4. DOI:10.1111/j.1464-410X.2011.10508.x; https://www.urovest.ru/jour/article/view/715Test

  2. 2
    دورية أكاديمية

    المساهمون: The study was not sponsored, Исследование не имело спонсорской поддержки

    المصدر: Urology Herald; Том 11, № 2 (2023); 203-214 ; Вестник урологии; Том 11, № 2 (2023); 203-214 ; 2308-6424 ; 10.21886/2308-6424-2023-11-2

    وصف الملف: application/pdf

    العلاقة: https://www.urovest.ru/jour/article/view/735/481Test; Chang SS, Bochner BH, Chou R, Dreicer R, Kamat AM, Lerner SP, Lotan Y, Meeks JJ, Michalski JM, Morgan TM, Quale DZ, Rosenberg JE, Zietman AL, Holzbeierlein JM. Treatment of Non-Metastatic Muscle-Invasive Bladder Cancer: AUA/ASCO/ASTRO/SUO Guideline. J Urol. 2017;198(3):552-559. Erratum in: J Urol. 2017;198(5):1175. DOI:10.1016/j.juro.2017.04.086; Chang SS, Boorjian SA, Chou R, Clark PE, Daneshmand S, Konety BR, Pruthi R, Quale DZ, Ritch CR, Seigne JD, Skinner EC, Smith ND, McKiernan JM. Diagnosis and Treatment of Non-Muscle Invasive Bladder Cancer: AUA/SUO Guideline. J Urol. 2016;196(4):1021-9. DOI:10.1016/j.juro.2016.06.049; Gul ZG, Katims AB, Winoker JS, Wiklund P, Waingankar N, Mehrazin R. Robotic assisted radical cystectomy versus open radical cystectomy: a review of what we do and don't know. Transl Androl Urol. 2021;10(5):2209-2215. DOI:10.21037/tau.2019.11.32; Parekh DJ, Reis IM, Castle EP, Gonzalgo ML, Woods ME, Svatek RS, Weizer AZ, Konety BR, Tollefson M, Krupski TL, Smith ND, Shabsigh A, Barocas DA, Quek ML, Dash A, Kibel AS, Shemanski L, Pruthi RS, Montgomery JS, Weight CJ, Sharp DS, Chang SS, Cookson MS, Gupta GN, Gorbonos A, Uchio EM, Skinner E, Venkatramani V, Soodana-Prakash N, Kendrick K, Smith JA Jr, Thompson IM. Robot-assisted radical cystectomy versus open radical cystectomy in patients with bladder cancer (RAZOR): an open-label, randomised, phase 3, non-inferiority trial. Lancet. 2018;391(10139):2525-2536. DOI:10.1016/S0140-6736(18)30996-6; Menon M, Hemal AK, Tewari A, Shrivastava A, Shoma AM, Abol-Ein H, Ghoneim MA. Robot-assisted radical cystectomy and urinary diversion in female patients: technique with preservation of the uterus and vagina. J Am Coll Surg. 2004;198(3):386-93. DOI:10.1016/j.jamcollsurg.2003.11.010; Beecken WD, Wolfram M, Engl T, Bentas W, Probst M, Blaheta R, Oertl A, Jonas D, Binder J. Robotic-assisted laparoscopic radical cystectomy and intra-abdominal formation of an orthotopic ileal neobladder. Eur Urol. 2003;44(3):337-9. DOI:10.1016/s0302-2838(03)00301-4; Клинические рекомендации «Рак мочевого пузыря». Ссылка активна на 09.12.2022. https://oncology-association.ru/wp-content/uploads/2022/03/rak-mochevogo-puzyrya.pdfTest; Hirobe M, Tanaka T, Shindo T, Ichihara K, Hotta H, Takahashi A, Kato R, Yanase M, Matsukawa M, Itoh N, Kunishima Y, Taguchi K, Horita H, Masumori N. Complications within 90 days after radical cystectomy for bladder cancer: results of a multicenter prospective study in Japan. Int J Clin Oncol. 2018;23(4):734-741. DOI:10.1007/s10147-018-1245-z; Lee CU, Kang M, Kim TJ, Na JP, Sung HH, Jeon HG, Seo SI, Jeon SS, Lee HM, Jeong BC. Predictors of postoperative complications after robot-assisted radical cystectomy with extracorporeal urinary diversion. Cancer Manag Res. 2019;11:5055-5063. DOI:10.2147/CMAR.S199432; Bochner BH, Dalbagni G, Sjoberg DD, Silberstein J, Keren Paz GE, Donat SM, Coleman JA, Mathew S, Vickers A, Schnorr GC, Feuerstein MA, Rapkin B, Parra RO, Herr HW, Laudone VP. Comparing Open Radical Cystectomy and Robot-assisted Laparoscopic Radical Cystectomy: A Randomized Clinical Trial. Eur Urol. 2015;67(6):1042-1050. DOI:10.1016/j.eururo.2014.11.043; Wijburg CJ, Hannink G, Michels CTJ, Weijerman PC, Issa R, Tay A, Decaestecker K, Wiklund P, Hosseini A, Sridhar A, Kelly J, d'Hondt F, Mottrie A, Klaver S, Edeling S, Dell'Oglio P, Montorsi F, Rovers MM, Witjes JA. Learning Curve Analysis for Intracorporeal Robot-assisted Radical Cystectomy: Results from the EAU Robotic Urology Section Scientific Working Group. Eur Urol Open Sci. 2022;39:55-61. DOI:10.1016/j.euros.2022.03.004; Dindo D, Demartines N, Clavien PA. Classification of surgical complications: a new proposal with evaluation in a cohort of 6336 patients and results of a survey. Ann Surg. 2004;240(2):205-13. DOI:10.1097/01.sla.0000133083.54934.ae; Borghesi M, Schiavina R, Antonelli A, Buizza C, Celia A, Parma P, De Concilio B, Mengoni F, Romagnoli D, Saraceni G, Brunocilla E, Porreca A. Peri-Operative Outcomes after Open and Robot-Assisted Radical Cystectomy by Using an Advanced Bipolar Seal and Cut Technology (Caiman®): A Prospective, Comparative, and Multi-Institutional Study. Curr Urol. 2019;12(2):64-69. DOI:10.1159/000489421; Chang SS, Smith JA Jr, Wells N, Peterson M, Kovach B, Cookson MS. Estimated blood loss and transfusion requirements of radical cystectomy. J Urol. 2001;166(6):2151-4. PMID: 11696725.; Оганян В.А., Грицкевич А.А., Симонов А.Д., Полотбек Ж., Мирошкина И.В., Костин А.А. Робот-ассистированная цистэктомия с ортотопической везикопластикой при раке мочевого пузыря. Экспериментальная и клиническая урология. 2022;15(3)44-54 DOI:10.29188/2222-8543-2022-15-3-44-54; https://www.urovest.ru/jour/article/view/735Test

  3. 3
    دورية أكاديمية

    المصدر: Urology Herald; Том 11, № 1 (2023); 171-176 ; Вестник урологии; Том 11, № 1 (2023); 171-176 ; 2308-6424 ; 10.21886/2308-6424-2023-11-1

    وصف الملف: application/pdf

    العلاقة: https://www.urovest.ru/jour/article/view/683/459Test; Аполихин О.И., Москвин С.В. Лазерная терапия при мужском бесплодии. Ч. 1. Этиология и патогенез. Экспериментальные исследования. Урология. 2017;(5):115-123. DOI:10.18565/urology.2017.5.115-123; Аполихин О.И., Красняк С.С. Влияние алкоголя на мужскую репродуктивную систему. Общественное здоровье. 2021;1(2):62-69. DOI:10.21045/2782-1676-2021-1-2-62-69; Боровец С.Ю., Москвин С.В. Методики лазерной терапии при мужском бесплодии. Вестник урологии. 2017;5(3):49-61. DOI:10.21886/2308-6424-2017-5-3-49-61; Гамидов С.И., Овчинников Р.И., Попова А.Ю., Наумов Н.П., Гасанов Н.Г. 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; World Health Organization (WHO). WHO laboratory manual for the examination and processing of human semen, sixth edition. Geneva: World Health Organization; 2021.; World Health Organization (WHO). WHO laboratory manual for the examination and processing of human semen. 5th ed. Geneva: WHO; 2010.; Patel AS, Leong JY, Ramasamy R. Prediction of male infertility by the World Health Organization laboratory manual for assessment of semen analysis: A systematic review. Arab J Urol. 2017;16(1):96-102. DOI:10.1016/j.aju.2017.10.005; Esteves SC, Zini A, Aziz N, Alvarez JG, Sabanegh ES Jr, Agarwal A. Critical appraisal of World Health Organization's new reference values for human semen characteristics and effect on diagnosis and treatment of subfertile men. Urology. 2012;79(1):16-22. DOI:10.1016/j.urology.2011.08.003; Alshahrani S, Aldossari K, Al-Zahrani J, Gabr AH, Henkel R, Ahmad G. Interpretation of semen analysis using WHO 1999 and WHO 2010 reference values: Abnormal becoming normal. Andrologia. 2018;50(2). DOI:10.1111/and.12838; Esteves SC. Clinical relevance of routine semen analysis and controversies surrounding the 2010 World Health Organization criteria for semen examination. Int Braz J Urol. 2014;40(4):443-53. DOI:10.1590/S1677-5538.IBJU.2014.04.02; Murray KS, James A, McGeady JB, Reed ML, Kuang WW, Nangia AK. The effect of the new 2010 World Health Organization criteria for semen analyses on male infertility. Fertil Steril. 2012;98(6):1428-31. DOI:10.1016/j.fertnstert.2012.07.1130; Guzick DS, Overstreet JW, Factor-Litvak P, Brazil CK, Nakajima ST, Coutifaris C, Carson SA, Cisneros P, Steinkampf MP, Hill JA, Xu D, Vogel DL; National Cooperative Reproductive Medicine Network. Sperm morphology, motility, and concentration in fertile and infertile men. N Engl J Med. 2001;345(19):1388-93. DOI:10.1056/NEJMoa003005; Nzisa J. The Evolution of the Worl Health Organization (WHO) Manual on Semen Analysis. EMJ Repro Health. 2021;7(1):23-26. URL: https://doi.org/10.33590/emjreprohealth/21F0810Test; Agarwal A, Parekh N, Panner Selvam MK, Henkel R, Shah R, Homa ST, Ramasamy R, Ko E, Tremellen K, Esteves S, Majzoub A, Alvarez JG, Gardner DK, Jayasena CN, Ramsay JW, Cho CL, Saleh R, Sakkas D, Hotaling JM, Lundy SD, Vij S, Marmar J, Gosalvez J, Sabanegh E, Park HJ, Zini A, Kavoussi P, Micic S, Smith R, Busetto GM, Bakırcıoğlu ME, Haidl G, Balercia G, Puchalt NG, Ben-Khalifa M, Tadros N, Kirkman-Browne J, Moskovtsev S, Huang X, Borges E, Franken D, Bar-Chama N, Morimoto Y, Tomita K, Srini VS, Ombelet W, Baldi E, Muratori M, Yumura Y, La Vignera S, Kosgi R, Martinez MP, Evenson DP, Zylbersztejn DS, Roque M, Cocuzza M, Vieira M, Ben-Meir A, Orvieto R, Levitas E, Wiser A, Arafa M, Malhotra V, Parekattil SJ, Elbardisi H, Carvalho L, Dada R, Sifer C, Talwar P, Gudeloglu A, Mahmoud AMA, Terras K, Yazbeck C, Nebojsa B, Durairajanayagam D, Mounir A, Kahn LG, Baskaran S, Pai RD, Paoli D, Leisegang K, Moein MR, Malik S, Yaman O, Samanta L, Bayane F, Jindal SK, Kendirci M, Altay B, Perovic D, Harlev A. Male Oxidative Stress Infertility (MOSI): Proposed Terminology and Clinical Practice Guidelines for Management of Idiopathic Male Infertility. World J Mens Health. 2019;37(3):296-312. DOI:10.5534/wjmh.190055; https://www.urovest.ru/jour/article/view/683Test

  4. 4
    دورية أكاديمية

    المصدر: Andrology and Genital Surgery; Том 24, № 4 (2023); 67-72 ; Андрология и генитальная хирургия; Том 24, № 4 (2023); 67-72 ; 2412-8902 ; 2070-9781

    وصف الملف: application/pdf

    العلاقة: https://agx.abvpress.ru/jour/article/view/706/551Test; Krawetz S.A. Paternal contribution: new insights and future challenges. Nat Revi Genet 2005;6(8):633–42. DOI:10.1038/nrg1654; Рогозин Д.С. Мужская фертильность: обзор литературы января – марта 2021 года. Вестник урологии 2021;9(2):142–9. DOI:10.21886/2308-6424-2021-9-2-142-149; Коршунов М.Н., Коршунова Е.С., Кызласов П.С. и др. Структурные нарушения хроматина сперматозоидов. Патофизиологические аспекты. Клиническая значимость. Вестник урологии 2021;9(1):95–104. DOI:10.21886/2308-6424-2021-9-1-95-104; Руднева С.А., Брагина Е.Е., Арифулин Е.А. и др. Фрагментация ДНК в сперматозоидах и ее взаимосвязь с нарушением сперматогенеза. Андрология и генитальная хирургия 2014;15(4):26–33. DOI:10.17650/2070-9781-2014-4-26-33; Рыжков А.И., Шорманов И.С., Соколова С.Ю. Фрагментация ДНК сперматозоидов. Есть ли связь с основными параметрами спермы и возрастом? Экспериментальная и клиническая урология 2020;(4):58–64. DOI:10.29188/2222-8543-2020-13-4-58-64; Авадиева Н.Э. Применение ДНК фрагментации спермы в андрологической практике. Вестник урологии 2019;7(1):7–11. DOI:10.21886/2308-6424-2019-7-1-7-11; Agarwal A., Majzoub A., Esteves S.C. et al. Clinical utility of sperm DNA fragmentation testing: practice recommendations based on clinical scenarios. Transl Androl Urol 2016;5(6):935–50. DOI:10.21037/tau.2016.10.03; Aitken R.J. Oxidative stress and the etiology of male infertility. J Assist Reprod Genet 2016;33(12):1691–2. DOI:10.1007/s10815-016-0791-4; Aitken R.J. DNA damage in human spermatozoa; important contributor to mutagenesis in the offspring. Transl Androl Urol 2017;6(Suppl 4):S761–4. DOI:10.21037/tau.2017.09.13; Bui A.D., Sharma R., Henkel R., Agarwal A. Reactive oxygen species impact on sperm DNA and its role in male infertility. Andrologia 2018;50(8):e13012. DOI:10.1111/and.13012; Esteves S.C., Gosálvez J., López-Fernández C. et al. Diagnostic accuracy of sperm DNA degradation index (DDSi) as a potential noninvasive biomarker to identify men with varicocele-associated infertility. Int Urol Nephrol 2015;47(9):1471–7. DOI:10.1007/s11255-015-1053-6; Rima D., Shiv B.K., Bhavna C. et al. Oxidative stress induced damage to paternal genome and impact of meditation and yoga – can it reduce incidence of childhood cancer? Asian Pac J Cancer Prev 2016;17(9):4517–25.; Коршунов М.Н., Коршунова Е.С., Даренков С.П. Клиническая эффективность использования тестикулярных сперматозоидов в программах репродуктивных технологий при высоком показателе ДНК-фрагментации. Урологические ведомости 2017;7(5):57–8.; 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; Simon L., Brunborg G., Stevenson M. et al. Clinical significance of sperm DNA damage in assisted reproduction outcome. Hum Reprod 2010;25(7):1594–608. DOI:10.1093/humrep/deq103; Simon L., Emery B.R., Carrell D.T. Review: diagnosis and impact of sperm DNA alterations in assisted reproduction. Best Pract Res Clin Obstet Gynaecol 2017;44:38–56. DOI:10.1016/j.bpobgyn.2017.07.003; Simon L., Lutton D., McManus J., Lewis S.E. Sperm DNA damage measured by the alkaline Comet assay as an independent predictor of male infertility and in vitro fertilization success. Fertil Steril 2011;95(2):652–7. DOI:10.1016/j.fertnstert.2010.08.019; Simon L., Murphy K., Shamsi M.B. et al. Paternal influence of sperm DNA integrity on early embryonic development. Hum Reprod 2014;29(11):2402–12. DOI:10.1093/humrep/deu228; Simon L., Proutski I., Stevenson M. et al. Sperm DNA damage has a negative association with live-birth rates after IVF. Reprod BioMed Online 2013;26(1):68–78. DOI:10.1016/j.rbmo.2012.09.019; Santi D., Spaggiari G., Simoni M. Sperm DNA fragmentation index as a promising predictive tool for male infertility diagnosis and treatment management – meta-analyses. Reprod Biomed Online 2018;37(3):315–26. DOI:10.1016/j.rbmo.2018.06.023; Spanò M., Kolstad A.H., Larsen S.B. et al. The applicability of the flow cytometric sperm chromatin structure assay in epidemiological studies. Asclepios. Hum Reprod 1998;13(9):2495–505. DOI:10.1093/humrep/13.9.2495; Evenson D.P., Jost L.K., Marshall D. et al. Utility of the sperm chromatin structure assay as a diagnostic and prognostic tool in the human fertility clinic. Hum Reprod 1999;14(4):1039–49. DOI:10.1093/humrep/14.4.1039; Evenson D.P., Wixon R.L. Environmental toxicants cause sperm DNA fragmentation as detected by the Sperm Chromatin Structure Assay (SCSA®). Toxicol Appl Pharmacol 2005;207(2 Suppl):532–7. DOI:10.1016/j.taap.2005.03.021; Malić Vončina S., Golob B., Ihan A. et al. Sperm DNA fragmentation and mitochondrial membrane potential combined are better for predicting natural conception than standard sperm parameters. Fertil Steril 2016;105(3):637–44.e.1. DOI:10.1016/j.fertnstert.2015.11.037; Vandekerckhove F.W., De Croo I., Gerris J. et al. Sperm chromatin dispersion test before sperm preparation is predictive of clinical pregnancy in cases of unexplained infertility treated with intrauterine insemination and induction with clomiphene citrate. Front Med (Lausanne) 2016;3:63. DOI:10.3389/fmed.2016.00063; Bungum M., Humaidan P., Axmon A. et al. Sperm DNA integrity assessment in prediction of assisted reproduction technology outcome. Hum Reprod 2007;22(1):174–9. DOI:10.1093/humrep/del326; Bungum M., Humaidan P., Spano M. et al. 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; Duran E.H., Morshedi M., Taylor S., Oehninger S. Sperm DNA quality predicts intrauterine insemination outcome: a prospective cohort study. Hum Reprod 2002;17(12):3122–8. DOI:10.1093/humrep/17.12.3122; Rilcheva V.S., Ayvazova N.P., Ilieva L.O. et al. Sperm DNA integrity test and assisted reproductive technology (Art) outcome. J Biomed Clin Res 2016;9:21–9. DOI:10.1515/jbcr-2016-0003; Chen Q., Zhao J.Y., Xue X., Zhu G.X. The association between sperm DNA fragmentation and reproductive outcomes following intrauterine insemination, a meta-analysis. Reprod Toxicol 2019;86:50–5. DOI:10.1016/j.reprotox.2019.03.004; Sugihara A., Van Avermaete F., Roelant E. et al. The role of sperm DNA fragmentation testing in predicting intra-uterine insemination outcome: a systematic review and meta-analysis. Eur J Obstet Gynecol Reprod Biol 2020;244:8–15. DOI:10.1016/j.ejogrb.2019.10.005; Li Z., Wang L., Cai J., Huang H. Correlation of sperm DNA damage with IVF and ICSI outcomes: a systematic review and meta-analysis. J Assist Reprod Genet 2006;23(9–10):367–76. DOI:10.1007/s10815-006-9066-9; Zhao J., Zhang Q., Wang Y., Li Y. Whether sperm deoxyribonucleic acid fragmentation has an effect on pregnancy and miscarriage after in vitro fertilization/intracytoplasmic sperm injection: a systematic review and meta-analysis. Fertil Steril 2014;102(4):998–1005e.8. DOI:10.1016/j.fertnstert.2014.06.033; Osman A., Alsomait H., Seshadri S. et al. The effect of sperm DNA fragmentation on live birth rate after IVF or ICSI: a systematic review and meta-analysis. Reprod Biomed Online 2015;30:120–7. DOI:10.1016/j.rbmo.2014.10.018; Deng C., Li T., Xie Y. et al. Sperm DNA fragmentation index influences assisted reproductive technology outcome: a systematic review and meta-analysis combined with a retrospective cohort study. Andrologia 2019;51(6):e13263. DOI:10.1111/and.13263; Lewis S.E.M. The place of sperm DNA fragmentation testing in current day fertility management. Middle East Fertil Soc J 2013;18(2):78–82. DOI:10.1016/j.mefs.2013.01.010; Meseguer M., Santiso R., Garrido N. et al. Effect of sperm DNA fragmentation on pregnancy outcome depends on oocyte quality. Fertil Steril 2011;95(1):124–8. DOI:10.1016/j.fertnstert.2010.05.055; Zini A., Jamal W., Cowan L., Al-Hathal N. Is sperm DNA damage associated with IVF embryo quality? A systematic review. J Assist Reprod Genet 2011;28(5):391–7. DOI:10.1007/s10815-011-9544-6; Moskovtsev S.I., Jarvi K., Mullen J.B. et al. Testicular spermatozoa have statistically significantly lower DNA damage compared with ejaculated spermatozoa in patients with unsuccessful oral antioxidant treatment. Fertil Steril 2010;93(4):1142–6. DOI:10.1016/j.fertnstert.2008.11.005; Greco E., Scarselli F., Iacobelli M. et al. Efficient treat ment of infertility due to sperm DNA damage by ICSI with testicular spermatozoa. Hum Reprod 2005;20(1):226–30. DOI:10.1093/humrep/deh590; Arafa M., AlMalki A., AlBadr M. et al. ICSI outcome in patients with high DNA fragmentation: testicular versus ejaculated spermatozoa. Andrologia 2018;50(1). DOI:10.1111/and.12835; Zhang J., Xue H., Qiu F. et al. Testicular spermatozoon is superior to ejaculated spermatozoon for intracytoplasmic sperm injection to achieve pregnancy in infertile males with high sperm DNA damage. Andrologia 2019;51(2):e13175. DOI:10.1111/and.13175; Pabuccu E.G., Caglar G.S., Tangal S. et al. Testicular versus ejaculated spermatozoa in ICSI cycles of nor mozoospermic men with high sperm DNA fragmen tation and previous ART failures. Andrologia 2017;49(2). DOI:10.1111/and.12609; Hayden R.P., Wright D.L., Toth T.L., Tanrikut C. Selective use of percutaneous testis biopsy to optimize IVF-ICSI outcomes: a case series. Fertil Res Pract 2016;2:7. DOI:10.1186/s40738-016-0020-y; Esteves S.C., Sánchez-Martín F., Sánchez-Martín P. et al. Comparison of reproductive outcome in oligozoospermic men with high sperm DNA fragmentation undergoing intracytoplasmic sperm injection with ejaculated and testicular sperm. Fertil Steril 2015;104(6):1398–405. DOI:10.1016/j.fertnstert.2015.08.028; Mehta A., Bolyakov A., Schlegel P.N., Paduch D.A. Higher pregnancy rates using testicular sperm in men withsevere oligospermia. Fertil Steril 2015;104(6):1382–7. DOI:10.1016/j.fertnstert.2015.08.008; Bradley C.K., McArthur S.J., Gee A.J. et al. Intervention improves assisted conception intracytoplasmic sperm injection outcomes for patients with high levels of sperm DNA fragmentation: a retrospective analysis. Andrology 2016;4(5):903–10. DOI:10.1111/andr.12215; Alharbi M., Hamouche F., Phillips S. et al. Use of testicular sperm in couples with SCSA-defined high sperm DNA fragmentation and failed intracytoplasmic sperm injection using ejaculated sperm. Asian J Androl 2020;22(4):348–53. DOI:10.4103/aja.aja_99_19; Esteves S.C., Roque M., Bradley C.K., Garrido N. Reproductive outcomes of testicular versus ejaculated sperm for intracytoplasmic sperm injection among men with high levels of DNA fragmentation in semen: systematic review and meta-analysis. Fertil Steril 2017;108(3):456–67.e1. DOI:10.1016/j.fertnstert.2017.06.018.; https://agx.abvpress.ru/jour/article/view/706Test

  5. 5
    دورية أكاديمية

    المصدر: Andrology and Genital Surgery; Том 24, № 4 (2023); 155-160 ; Андрология и генитальная хирургия; Том 24, № 4 (2023); 155-160 ; 2412-8902 ; 2070-9781

    وصف الملف: application/pdf

    العلاقة: https://agx.abvpress.ru/jour/article/view/716/561Test; Williams M., Jezior J. Management of combat-related urological trauma in the modern era. Nat Rev Urol 2013;10(9):504–12. DOI:10.1038/nrurol.2013.148; Serkin F.B., Soderdahl D.W., Hernandez J. et al. Combat urologic trauma in US military overseas contingency operations. J Trauma 2010;69(Suppl 1):S175–8. DOI:10.1097/TA.0b013e3181e45cd1; Goldman C., Shaw N., du Plessis D. et al. Gunshot wounds to the penis and scrotum: a narrative review of management in civilian and military settings. Transl Androl Urol 2021;10(6):2596–608. DOI:10.21037/tau-20-1175; Hudak S.J., Morey A.F., Rozanski T.A., Fox CW Jr. Battlefield urogenital injuries: changing patterns during the past century. Urology 2005;65(6):1041–6. DOI:10.1016/j.urology.2004.11.031; Vucković I., Tucak A., Gotovac J. et al. Croatian experience in the treatment of 629 urogenital war injuries. J Trauma 1995;39(4):733–6. DOI:10.1097/00005373-199510000-00023; Tucak A., Lukacević T., Kuvezdić H. et al. Urogenital wounds during the war in Croatia in 1991/1992. J Urol 1995;153(1):121–2. DOI:10.1097/00005392-199501000-00043; Al-Azzawi I.S., Koraitim M.M. Lower genitourinary trauma in modern warfare: the experience from civil violence in Iraq. Injury 2014;45(5):885–9. DOI:10.1016/j.injury.2014.01.005; Al-Azzawi I.S., Koraitim M.M. Urethral and penile war injuries: the experience from civil violence in Iraq. Arab J Urol 2014;12(2):149–54. DOI:10.1016/j.aju.2013.11.002; Phonsombat S., Master V.A., McAninch J.W. Penetrating external genital trauma: a 30-year single institution experience. J Urol 2008;180(1):192–5; discussion 195–6. DOI:10.1016/j.juro.2008.03.041; Golovko S., Gybalo R., Lurin I. et al. Penetrating gunshot wounds to the penis: a case report of combat patient injured in the war in Ukraine. Int J Emerg Med 2023;16(1):5. DOI:10.1186/s12245-023-00481-5; Cerwinka W.H., Block N.L. Civilian gunshot injuries of the penis: the Miami experience. Urology 2009;73(4):877–80. DOI:10.1016/j.urology.2008.10.057; Kunkle D.A., Lebed B.D., Mydlo J.H., Pontari M.A. Evaluation and management of gunshot wounds of the penis: 20-year experience at an urban trauma center. J Trauma 2008;64(4):1038–42. DOI:10.1097/TA.0b013e3180342036; Balzano F.L., Hudak S.J. Military genitourinary injuries: past, present, and future. Transl Androl Urol 2018;7(4):646–52. DOI:10.21037/tau.2018.04.05; Шанава Г.Ш., Мосоян М.С., Грабский А.М., Арзуманян К.Г. Особенности извлечения инородных тел из нижних мочевыводящих путей. Урологические ведомости 2021;11(3):213–8. DOI:10.17816/uroved72148; https://agx.abvpress.ru/jour/article/view/716Test

  6. 6
    دورية أكاديمية

    المصدر: Andrology and Genital Surgery; Том 24, № 2 (2023); 49-57 ; Андрология и генитальная хирургия; Том 24, № 2 (2023); 49-57 ; 2412-8902 ; 2070-9781

    وصف الملف: application/pdf

    العلاقة: https://agx.abvpress.ru/jour/article/view/660/520Test; Jensen C.F.S., Østergren P., Dupree J.M. et al. Varicocele and male infertility. Nat Rev Urol 2017;14(9):523–33. DOI:10.1038/nrurol.2017.98; Peterson A.C., Lance R.S., Ruiz H.E. Outcomes of varicocele ligation done for pain. J Urol 1998;159(5):1565–7. DOI:10.1097/00005392-199805000-00043; Shridharani A., Lockwood G., Sandlow J. Varicocelectomy in the treatment of testicular pain: a review. Curr Opin Urol 2012;22(6):499–506. DOI:10.1097/MOU.0b013e328358f69f; Котов С.В., Корочкин Н.Д., Клименко А.А. Рецидивное варикоцеле. Вестник урологии 2021;9(2):132–41. DOI:10.21886/2308-6424-2021-9-2-132-141; Жуков О.Б., Капто А.А., Михайленко Д.С., Евдокимов В.В. Варикозная болезнь органов таза мужчины. Андрология и генитальная хирургия 2016;17(4):72–7. DOI:10.17650/2070-9781-2016-17-4-72-77; Yaman O., Ozdiler E., Anafarta K., Göğüş O. Effect of microsurgical subinguinal varicocele ligation to treat pain. Urology 2000;55(1):107–8. DOI:10.1016/S0090-4295(99)00374-X; Schlegel P.N., Goldstein M. Alternate indications for varicocele repair: non-obstructive azoospermia, pain, androgen deficiency and progressive testicular dysfunction. Fertil Steril 2011;96(6):1288–93. DOI:10.1016/j.fertnstert.2011.10.033; Muthuveloe D.W., During V., Ashdown D. et al. The effectiveness of varicocele embolisation for the treatment of varicocele related orchalgia. Springer Plus 2015;4(1):392. DOI:10.1186/s40064-015-1177-2; Elzanaty S., Johansen C.E. Microsurgical varicocele repair on men with grade III lesions and chronic dull scrotal pain: a pilot study. Curr Urol 2015;8(1):29–31. DOI:10.1159/000365685; Han D.Y., Yang Q.Y., Chen X. et al. Who will benefit from surgical repair for painful varicocele: a meta-analysis. Int Urol Nephrol 2016;48(7):1071–8. DOI:10.1007/s11255-016-1246-7; Karademir K., Senkul T., Baykal K. et al. Evaluation of the role of varicocelectomy including external spermatic vein ligation in patients with scrotal pain. Int J Urol 2005;12(5):484–8. DOI:10.1111/j.1442-2042.2005.01063.x; Maghraby H.A. Laparoscopic varicocelectomy for painful varicoceles: merits and outcomes. J Endourol 2002;16(2):107–10. DOI:10.1089/089277902753619627; Parekattil S.J., Brahmbhatt J.V. Robotic approaches for male infertility and chronic orchialgia microsurgery. Curr Opin Urol 2011;21(6):493–9. DOI:10.1097/MOU.0b013e32834bb783; Chawla A., Kulkarni G., Kamal K., Zini A. Microsurgical varicocelectomy for recurrent or persistent varicoceles associated with orchalgia. Urology 2005;66(5):1072–4. DOI:10.1016/j.urology.2005.05.052; Reşorlu B., Kara C., Sahin E., Unsal A. et al. The significance of age on success of surgery for patients with varicocele. Int Urol Nephrol 2010;42(2):351–6. DOI:10.1007/s11255-009-9589-y; Park H.J., Lee S.S., Park N.C. Predictors of pain resolution after varicocelectomy for painful varicocele. Asian J Androl 2011;13(5):754–8. DOI:10.1038/aja.2010.87; Kim S.O., Jung H., Park K. Outcomes of microsurgical subinguinal varicocelectomy for painful varicoceles. J Androl 2012;33(5):872–5. DOI:10.2164/jandrol.111.014993; Chen L.K., Chen S.S. Risk factors for developing pain in normospermic patients with varicocoele. Int J Androl 2012;35:176–80. DOI:10.1111/j.1365-2605.2011.01215.x; Handel L.N., Shetty R., Sigman M. The relationship between varicoceles and obesity. J Urol 2006;176(5):2138–40; discussion 2140. DOI:10.1016/j.juro.2006.07.023; Soylemez H., Atar M., Ali Sancaktutar A. et al. Varicocele among healthy young men in Turkey; prevalence and relationship with body mass index. Int Braz J Urol 2012;38(1):116–21. DOI:10.1590/S1677-55382012000100016; Bungum M., Humaidan P., Axmon A. et al. Sperm DNA integrity assessment in prediction of assisted reproduction technology outcome. Hum Reprod 2007;22(1):174–9. DOI:10.1093/humrep/del326; Collins J.A., Barnhart K.T., Schlegel P.N. Do sperm DNA integrity tests predict pregnancy with in vitro fertilization? Fertil Steril 2008;89(4):823–31. DOI:10.1016/j.fertnstert.2007.04.055; Giwercman A., Lindstedt L., Larsson M. et al. Sperm chromatin structure assay as an independent predictor of fertility in vivo: a case-control study. Int J Androl 2010;33(1):e221–7. DOI:10.1111/j.1365-2605.2009.00995.x; Henkel R., Hajimohammad M., Stalf T. et al. Influence of deoxyribonucleic acid damage on fertilization and pregnancy. Fertil Steril 2004;81(4):965–72. DOI:10.1016/j.fertnstert.2003.09.044; Virro M.R., Larson-Cook K.L., Evenson D.P. Sperm chromatin structure assay (SCSA) parameters are related to fertilization, blastocyst development, and ongoing pregnancy in in vitro fertilization and intracytoplasmic sperm injection cycles. Fertil Steril 2004;81(5):1289–95. DOI:10.1016/j.fertnstert.2003.09.063; Lopes S., Sun J.G., Jurisicova A. et al. Sperm deoxyribonucleic acid fragmentation is increased in poor-quality semen samples and correlates with failed fertilization in intracytoplasmic sperm injection. Fertil Steril 1998;69(3):528–32. DOI:10.1016/S0015-0282(97)00536-0; De Iuliis G.N., Thomson L.K., Mitchell L.A. et al. DNA damage in human spermatozoa is highly correlated with the efficiency of chromatin remodeling and the formation of 8-hydroxy-2’- deoxyguanosine, a marker of oxidative stress. Biol Reprod 2009;81(3):517–24. DOI:10.1095/biolreprod.109.076836; Sakkas D., Seli E., Bizzaro D. et al. Abnormal spermatozoa in the ejaculate: abortive apoptosis and faulty nuclear remodelling during spermatogenesis. Reprod Biomed Online 2003;7(4):428–32. DOI:10.1016/S1472-6483(10)61886-X; Tremellen K. Oxidative stress and male infertility – a clinical perspective. Hum Reprod Update 2008;14(3):243–58. DOI:10.1093/humupd/dmn004; Sun J.G., Jurisicova A., Casper R.F. Detection of deoxyribonucleic acid fragmentation in human sperm: correlation with fertilization in vitro. Biol Reprod 1997;56(3):602–7. DOI:10.1095/biolreprod56.3.602; Allamaneni S.S., Naughton C.K., Sharma R.K. et al. Increased seminal reactive oxygen species levels in patients with varicoceles correlate with varicocele grade but not with testis size. Fertil Steril 2004;82(6):1684–6. DOI:10.1016/j.fertnstert.2004.04.071; Mancini A., Meucci E., Milardi D. et al. Seminal antioxidant capacity in pre- and postoperative varicocele. J Androl 2004;25(1):44–9. DOI:10.1002/j.1939-4640.2004.tb02757.x; Saleh R.A., Agarwal A., Sharma R.K. et al. Evaluation of nuclear DNA damage in spermatozoa from infertile men with varicocele. Fertil Steril 2003;80(6):1431–6. DOI:10.1016/S0015-0282(03)02211-8; Schoor R.A., Elhanbly S.M., Niederberger C. The pathophysiology of varicocele-associated male infertility. Curr Urol Rep 2001;2(6):432–6. DOI:10.1007/s11934-001-0035-7; Naughton C.K., Nangia A.K., Agarwal A. Pathophysiology of varicoceles in male infertility. Hum Reprod Update 2001;7(5):473–81. DOI:10.1093/humupd/7.5.473; Nallella K.P., Allamaneni S.S., Pasqualotto F.F. et al. Relationship of interleukin-6 with semen characteristics and oxidative stress in patients with varicocele. Urology 2004;64(5):1010–3. DOI:10.1016/j.urology.2004.05.045; Гамидов С.И., Овчинников Р.И., Попова А.Ю. и др. Адъювантная антиоксидантная терапия у больных бесплодием при варикоцеле. Урология 2017;2(suppl):64–72. DOI:10.18565/urol.2017.2-supplement.64-72; Köksal I.T., Tefekli A., Usta M. et al. The role of reactive oxygen species in testicular dysfunction associated with varicocele. BJU Int 2000;86(4):549–52. DOI:10.1046/j.1464-410X.2000.00755.x; Blumer C.G., Restelli A.E., Giudice P.T. et al. Effect of varicocele on sperm function and semen oxidative stress. BJU Int 2012;109(2):259–65. DOI:10.1111/j.1464-410X.2011.10240.x; Romeo C., Ientile R., Santoro G. et al. Nitric oxide production is increased in the spermatic veins of adolescents with left idiophatic varicocele. J Pediatr Surg 2001;36(2):389–93. DOI:10.1053/jpsu.2001.20724; Pasqualotto F.F., Sundaram A., Sharma R.K. et al. Semen quality and oxidative stress scores in fertile and infertile patients with varicocele. Fertil Steril 2008;89(3):602–7. DOI:10.1016/j.fertnstert.2007.03.057; Mostafa T., Anis T.H., El-Nashar A. et al. Varicocelectomy reduces reactive oxygen species levels and increases antioxidant activity of seminal plasma from infertile men with varicocele. Int J Androl 2001;24(5):261–5. DOI:10.1046/j.1365-2605.2001.00296.x; Cervellione R.M., Cervato G., Zampieri N. et al. Effect of varicocelectomy on the plasma oxidative stress parameters. J Pediatr Surg 2006;41(2):403–6. DOI:10.1016/j.jpedsurg.2005.11.018; Hurtado de Catalfo G.E., Ranieri-Casilla A., Marra F.A. et al. Oxidative stress biomarkers and hormonal profile in human patients undergoing varicocelectomy. Int J Androl 2007;30(6):519–30. DOI:10.1111/j.1365-2605.2007.00753.x; Chen S.S., Huang W.J., Chang L.S., Wei Y.H. Attenuation of oxidative stress after varicocelectomy in subfertile patients with varicocele. J Urol 2008;179(2):639–42. DOI:10.1016/j.juro.2007.09.039; Sakamoto Y., Ishikawa T., Kondo Y. et al. The assessment of oxidative stress in infertile patients with varicocele. BJU Int 2008;101(12):1547–52. DOI:10.1111/j.1464-410X.2008.07517.x; Dada R., Shamsi M.B., Venkatesh S. et al. Attenuation of oxidative stress & DNA damage in varicocelectomy: implications in infertility management. Indian J Med Res 2010;132(6):728–30.; Rodriguez Peña M, Alescio L, Russell A, et al. Predictors of improved seminal parameters and fertility after varicocele repair in young adults. Andrologia 2009;41(5):277–81. DOI:10.1111/j.1439-0272.2009.00919.x; Lacerda J.I., Del Giudice P.T., da Silva B.F. et al. Adolescent varicocele: improved sperm function after varicocelectomy. Fertil Steril 2011;95(3):994–9. DOI:10.1016/j.fertnstert.2010.10.031; Smith R., Kaune H., Parodi D. et al. Increased sperm DNA damage in patients with varicocele: relationship with seminal oxidative stress. Hum Reprod 2006;21(4):986–93. DOI:10.1093/humrep/dei429; Blumer C.G., Fariello R.M., Restelli A.E. et al. Sperm nuclear DNA fragmentation and mitochondrial activity in men with varicocele. Fertil Steril 2008;90(5):171–22. DOI:10.1016/j.fertnstert.2007.09.007; Chen C.H., Lee S.S., Chen D.C. et al. Apoptosis and kinematics of ejaculated spermatozoa in patients with varicocele. J Androl 2004;25(3):348–53. DOI:10.1002/j.1939-4640.2004.tb02799.x; Bertolla R.P., Cedenho A.P., Hassun Filho P.A. et al. Sperm nuclear DNA fragmentation in adolescents with varicocele. Fertil Steril 2006;85(3):625–8. DOI:10.1016/j.fertnstert.2005.08.032; Zini A, Dohle G. Are varicoceles associated with increased deoxyribonucleic acid fragmentation? Fertil Steril 2011;96(6):1283–7. DOI:10.1016/j.fertnstert.2011.10.016; Zini A., Azhar R., Baazeem A., Gabriel M.S. Effect of microsurgical varicocelectomy on human sperm chromatin and DNA integrity: a prospective trial. Int J Androl 2011;34(1):14–9. DOI:10.1111/j.1365-2605.2009.01048.x; Werthman P., Wixon R., Kasperson K., Evenson D.P. Significant decrease in sperm deoxyribonucleic acid fragmentation after varicocelectomy. Fertil Steril 2008;90(5):1800–4. DOI:10.1016/j.fertnstert.2006.09.019; Marmar J.L., Agarwal A., Prabakaran S. et al. Reassessing the value of varicocelectomy as a treatment for male subfertility with a new meta-analysis. Fertil Steril 2007;88(3):639–48. DOI:10.1016/j.fertnstert.2006.12.008; Smit M., Romijn J.C., Wildhagen M.F. et al. Decreased sperm DNA fragmentation after surgical varicocelectomy is associated with increased pregnancy rate. J Urol 2010;183(1):270–4. DOI:10.1016/j.juro.2009.08.161; Baker K., McGill J., Sharma R. et al. Pregnancy after varicocelectomy: impact of postoperative motility and DFI. Urology 2013;81(4):760–6. DOI:10.1016/j.urology.2012.12.005; Leung L., Ho K.L., Tam P.C., Yiu M.K. Subinguinal microsurgical varicocelectomy for male factor subfertility: ten-year experience. Hong Kong Med J 2013;19(4):334–40. DOI:10.12809/hkmj133884; Practice Committee of American Society for Reproductive Medicine. Report on varicocele and infertility. Fertil Steril 2008;90(5 Suppl):S247–9. DOI:10.1016/j.fertnstert.2008.08.050; Agarwal A., Deepinder F., Cocuzza M. et al. Efficacy of varicocelectomy in improving semen parameters: new metaanalytical approach. Urology 2007;70(3):532–8. DOI:10.1016/j.urology.2007.04.011; French D.B., Desai N.R., Agarwal A. Varicocele repair: does it still have a role in infertility treatment? Curr Opin Obstet Gynecol 2008;20(3):269–74. DOI:10.1097/GCO.0b013e3282fcc00c; Acar H., Kilinc M., Guven S. et al. Comparison of semen profile and frequency of chromosome aneuploidies in sperm nuclei of patients with varicocele before and after varicocelectomy. Andrologia 2009;41(3):157–62. DOI:10.1111/j.1439-0272.2008.00907.x; Mansour Ghanaie M., Asgari S.A., Dadrass N. et al. Effects of varicocele repair on spontaneous first trimester miscarriage: a randomized clinical trial. Urol J 2012;9(2):505–13.; Nasr-Esfahani M.H., Abasi H., Razavi S. et al. Varicocelectomy: semen parameters and protamine deficiency. Int J Androl 2009;32(2):115–22. DOI:10.1111/j.1365-2605.2007.00822.x; Pirke K.M., Vogt H.J., Sintermann R., Spyra B. Testosterone in peripheral plasma, spermatic vein and in testicular tissue under basal conditions and after HCG-stimulation in patients with varicocele. Andrologia 1983;15(6):637–41. DOI:10.1111/j.1439-0272.1983.tb00179.x; Abdel-Meguid T.A., Farsi H.M., Al-Sayyad A. et al. Effects of varicocele on serum testosterone and changes of testosterone after varicocelectomy: a prospective controlled study. Urology 2014;84(5):1081–7. DOI:10.1016/j.urology.2014.05.029; Tanrikut C., Goldstein M., Rosoff J.S. et al. Varicocele as a risk factor for androgen deficiency and effect of repair. BJU Int 2011;108(9):1480–4. DOI:10.1111/j.1464-410X.2010.10030.x; Goldstein M., Eid J.F. Elevation of intratesticular and scrotal skin surface temperature in men with varicocele. J Urol 1989;142(3):743–5. DOI:10.1016/S0022-5347(17)38874-2; Li F., Yue H., Yamaguchi K. et al. Effect of surgical repair on testosterone production in infertile men with varicocele: a metaanalysis. Int J Urol 2012;19(2):149–54. DOI:10.1111/j.1442-2042.2011.02890.x; Hsiao W., Rosoff J.S., Pale J.R. et al. Older age is associated with similar improvements in semen parameters and testosterone after subinguinal microsurgical varicocelectomy. J Urol 2011;185(2):620–5. DOI:10.1016/j.juro.2010.09.114; Zohdy W., Ghazi S., Arafa M. Impact of varicocelectomy on gonadal and erectile functions in men with hypogonadism and infertility. J Sex Med 2011;8(3):885–93. DOI:10.1111/j.1743-6109.2010.01974.x; Sathya Srini V., Belur Veerachari S. Does varicocelectomy improve gonadal function in men with hypogonadism and infertility? Analysis of a prospective study. Int J Endocrinol 2011;2011:916380. DOI:10.1155/2011/916380; Hsiao W., Rosoff J.S., Pale J.R. et al. Varicocelectomy is associated with increases in serum testosterone independent of clinical grade. Urology 2013;81(6):1213–7. DOI:10.1016/j.urology.2013.01.060; Esteves S.C. Clinical management of infertile men with nonobstructive azoospermia. Asian J Androl 2015;17(3):459–70. DOI:10.4103/1008-682X.148719; Попова А.Ю., Гамидов С.И., Овчинников Р.И. и др. Варикоцеле и необструктивная азооспермия – с чего начать? Андрология и генитальная хирургия 2017;18(4):77–80. DOI:10.17650/2070-9781-2017-18-4-77-80; Gat Y., Bachar G.N., Everaert K. et al. Induction of spermatogenesis in azoospermic men after internal spermatic vein embolization for the treatment of varicocele. Hum Reprod 2005;20(4):1013–7. DOI:10.1093/humrep/deh706; Esteves S.C., Miyaoka R., Agarwal A. Sperm retrieval techniques for assisted reproduction. Int Braz J Urol 2011;37(5):570–83. DOI:10.1590/S1677-55382011000500002; Matthews G.J., Matthews E.D., Goldstein M. Induction of spermatogenesis and achievement of pregnancy after microsurgical varicocelectomy in men with azoospermia and severe oligoasthenospermia. Fertil Steril 1998;70(1):71–5. DOI:10.1016/S0015-0282(98)00108-3; Poulakis V., Ferakis N., de Vries R. et al. Induction of spermatogenesis in men with azoospermia or severe oligoteratoasthenospermia after antegrade internal spermatic vein sclerotherapy for the treatment of varicocele. Asian J Androl 2006;8(5):613–9. DOI:10.1111/j.1745-7262.2006.00157.x; Abdel-Meguid T.A., Al-Sayyad A., Tayib A., Farsi H.M. 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; Kiraç M., Deniz N., Biri H. The effect of microsurgical varicocelectomy on semen parameters in men with non-obstructive azoospermia. Curr Urol 2013;6(3):136–40. DOI:10.1159/000343527; Schlegel P.N., Kaufmann J. Role of varicocelectomy in men with nonobstructive azoospermia. Fertil Steril 2004;81(6):1585–8. DOI:10.1016/j.fertnstert.2003.10.036; Tung M.C., Huang W.J., Chen K.K. Modified subinguinal varicocelectomy for painful varicocele and varicocele-associated infertility. J Chin Med Assoc 2004;67(6):296–300.; Pasqualotto F.F., Sobreiro B.P., Hallak J. et al. Induction of spermatogenesis in azoospermic men after varicocelectomy repair: an update. Fertil Steril 2006;85(3):635–9. DOI:10.1016/j.fertnstert.2005.08.043; Esteves S.C., Glina S. Recovery of spermatogenesis after microsurgical subinguinal varicocele repair in azoospermic men based on testicular histology. Int Braz J Urol 2005;31(6):541–8. DOI:10.1590/S1677-55382005000600005; Esteves S.C., Miyaoka R., Roque M., Agarwal A. Outcome of varicocele repair in men with nonobstructive azoospermia: systematic review and meta-analysis. Asian J Androl 2016;18(2):246–53. DOI:10.4103/1008-682X.169562; Ustuner M., Yilmaz H., Yavuz U. et al. Varicocele repair improves testicular histology in men with nonobstructive azoospermia. Biomed Res Int 2015;2015:709452. DOI:10.1155/2015/709452; Yamamoto M., Hibi H., Hirata Y. et al. 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.; Kroese A.C., de Lange N.M., Collins J., Evers J.L. Surgery or embolization for varicoceles in subfertile men. Cochrane Database Syst Rev 2012;10:CD000479. DOI:10.1002/14651858.CD000479.pub5; Kim K.H., Lee J.Y., Kang D.H. et al. Impact of surgical varicocele repair on pregnancy rate in subfertile men with clinical varicocele and impaired semen quality: a meta-analysis of randomized clinical trials. Korean J Urol 2013;54(10):703–9. DOI:10.4111/kju.2013.54.10.703; Kirby E.W., Wiener L.E., Rajanahally S. et al. Undergoing varicocele repair before assisted reproduction improves pregnancy rate and live birth rate in azoospermic and oligospermic men with a varicocele: a systematic review and meta-analysis. Fertil Steril 2016;106(6):1338–43. DOI:10.1016/j.fertnstert.2016.07.1093; https://agx.abvpress.ru/jour/article/view/660Test

  7. 7
    دورية أكاديمية

    المصدر: Andrology and Genital Surgery; Том 24, № 1 (2023); 24-35 ; Андрология и генитальная хирургия; Том 24, № 1 (2023); 24-35 ; 2412-8902 ; 2070-9781

    وصف الملف: application/pdf

    العلاقة: https://agx.abvpress.ru/jour/article/view/633/500Test; Ефремов Е.А., Касатонова Е.В. Актуальные и перспективные методы лечения идиопатического мужского бесплодия. Андрология и генитальная хирургия 2022;23(3):48–53. DOI:10.17650/2070-9781-2022-23-3-48-53; Ефремов Е.А., Касатонова Е.В., Красняк С.С. и др. Роль антиоксидантов в улучшении параметров эякулята и клинических исходов у бесплодных пар. Экспериментальная и клиническая урология 2017;2:61–5.; Louis J.F., Thoma M.E., Sørensen D.N. et al. The prevalence of couple infertility in the United States from a male perspective: evidence from a nationally representative sample. Andrology 2013;1(5):741–8. DOI:10.1111/j.2047-2927.2013.00110.x; 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; Jungwirth А., Diemer T., Kopa Z. et al. Male infertility. EAU Guideline. 2020. Available at: https://uroweb.org/guideline/male-infertility/#8Test.; Hamada A., Esteves S.C., Nizza M., Agarwal A. Unexplained male infertility: diagnosis and management. Int Braz J Urol 2012;38(5):576–94. DOI:10.1590/s1677-55382012000500002; Esteves S.C., Agarwal A. Novel concepts in male infertility. Int Braz J Urol 2011;37(1):5–15. DOI:10.1590/s1677-55382011000100002; Esteves S.C., Agarwal A., Cho C.L., Majzoub A. A Strengths-Weaknesses-Opportunities-Threats (SWOT) analysis on the clinical utility of sperm DNA fragmentation testing in specific male infertility scenarios. Trans Androl Urol 2017;6(Suppl 4):S734–S60. DOI:10.21037/tau.2017.08.20; Santi D., Spaggiari G., Simoni M. Sperm DNA fragmentation index as a promising predictive tool for male infertility diagnosis and treatment management – meta-analyses. Reprod Biomed Online 2018;37(3):315–26. DOI:10.1016/j.rbmo.2018.06.023; Коршунов М.Н., Коршунова Е.С., Кызласов П.С. и др. Структурные нарушения хроматина сперматозоидов. Патофизиологические аспекты. Клиническая значимость. Вестник урологии 2021;9(1):95–104. DOI:10.21886/2308-6424-2021-9-1-95-104; Gill K., Jakubik J., Rosiak-Gill A. et al. Utility and predictive value of human standard semen parameters and sperm DNA dispersion for fertility potential. Int J Environ Res Public Health 2019;16(11):2004. DOI:10.3390/ijerph16112004; Dada R. Sperm DNA damage diagnostics: when and why. Transl Androl Urol 2017;6(Suppl 4):S691–S4. DOI:10.21037/tau.2017.05.26; Esteves S.C., Zini A., Coward R.M. et al. Sperm DNA fragmentation testing: summary evidence and clinical practice recommendations. Andrologia 2021;53(2):e13874. DOI:10.1111/and.13874; Руднева С.А., Брагина Е.Е., Арифулин Е.А. и др. Фрагментация ДНК в сперматозоидах и ее взаимосвязь с нарушением сперматогенеза. Андрология и генитальная хирургия 2014;15(4):26–33. DOI:10.17650/2070-9781-2014-4-26-33; Авадиева Н.Э. Применение ДНК фрагментации спермы в андрологической практике. Вестник урологии 2019;7(1):7–11. DOI:10.21886/2308-6424-2019-7-1-7-11; Cho C.L., Agarwal A., Majzoub A., Esteves S.C. Clinical utility of sperm DNA fragmentation testing: concise practice recommendations. Transl Androl Urol 2017;6(Suppl 4):S366–S73. DOI:10.21037/tau.2017.07.28; Esteves S.C., Santi D., Simoni M. An update on clinical and surgical interventions to reduce sperm DNA fragmentation in infertile men. Andrology 2020;8(1):53–81. DOI:10.1111/andr.12724; Boeri L., Capogrosso P., Ventimiglia E. et al. Heavy cigarette smoking and alcohol consumption are associated with impaired sperm parameters in primary infertile men. Asian J Androl 2019;21(5):478–85. DOI:10.4103/aja.aja_110_18; Sharma R., Harlev A., Agarwal A., Esteves S.C. 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(4):635–45. DOI:10.1016/j.eururo.2016.04.010; Mostafa R.M., Nasrallah Y.S., Hassan M.M. et al. The effect of cigarette smoking on human seminal parameters, sperm chromatin structure and condensation. Andrologia 2018;50(3):e12910. DOI:10.1111/and.12910; Fraga C.G., Motchnik P.A., Wyrobek A.J. et al. Smoking and low antioxidant levels increase oxidative damage to sperm DNA. Mutat Res 1996;351(2):199–203. DOI:10.1016/0027-5107(95)00251-0; Gunes S., Metin Mahmutoglu A., Arslan M.A., Henkel R. Smokinginduced genetic and epigenetic alterations in infertile men. Andrologia 2018;50(9):e13124. DOI:10.1111/and.13124; Ranganathan P., Rao K.A., Thalaivarasai Balasundaram S. Deterioration of semen quality and sperm-DNA integrity as influenced by cigarette smoking in fertile and infertile human male smokers – a prospective study. J Cell Biochem 2019;120(7):11784–93. DOI:10.1002/jcb.28458; Vande Loock K., Ciardelli R., Decordier I. et al. Preterm newborns show slower repair of oxidative damage and paternal smoking associated DNA damage. Mutagenesis 2012;27(5):573–80. DOI:10.1093/mutage/ges022; Aboulmaouahib S., Madkour A., Kaarouch I. et al. Impact of alcohol and cigarette smoking consumption in male fertility potential: looks at lipid peroxidation, enzymatic antioxidant activities and sperm DNA damage. Andrologia 2018;50(3):e12926. DOI:10.1111/and.12926; Lee K.M., Ward M.H., Han S. et al. Paternal smoking, genetic polymorphisms in CYP1A1 and childhood leukemia risk. Leuk Res 2009;33(2):250–58. DOI:10.1016/j.leukres.2008.06.031; Kumar S.B., Chawla B., Bisht S. et al. Tobacco use increases oxidative DNA damage in sperm – possible etiology of childhood cancer. Asian Pac J Cancer Prev 2015;16(16):6967–72. DOI:10.7314/apjcp.2015.16.16.6967; Verhaeghe F., Di Pizio P., Bichara C. et al. Cannabis consumption might exert deleterious effects on sperm nuclear quality in infertile men. Reprod Biomed Online 2020;40(2):270–80. DOI:10.1016/j.rbmo.2019.11.002; Radwan M., Jurewicz J., Merecz-Kot D. et al. Sperm DNA damage-the effect of stress and everyday life factors. Int J Impot Res 2016;28(4):148–54. DOI:10.1038/ijir.2016.15; Schmid T.E., Eskenazi B., Baumgartner A. et al. The effects of male age on sperm DNA damage in healthy non-smokers. Hum Reprod 2007;22(1):180–7. DOI:10.1093/humrep/del338; Jurewicz J., Hanke W., Radwan M., Bonde J.P. Environmental factors and semen quality. Int J Occup Med Environ Health 2009;22(4):305–29. DOI:10.2478/v10001-009-0036-1; Lafuente R., García-Blàquez N., Jacquemin B., Checa M.A. Outdoor air pollution and sperm quality. Fertil Steril 2016;106(4):880– 96. DOI:10.1016/j.fertnstert.2016.08.022; Radwan M., Jurewicz J., Polańska K. et al. Exposure to ambient air pollution – does it affect semen quality and the level of reproductive hormones? Ann Hum Biol 2016;43(1):50–6. DOI:10.3109/03014460.2015.1013986; Rubes J., Selevan S.G., Evenson D.P. et al. Episodic air pollution is associated with increased DNA fragmentation in human sperm without other changes in semen quality. Hum Reprod 2005;20(10):2776–83. DOI:10.1093/humrep/dei122; Jeng H.A., Pan C.H., Chao M.R. et al. Sperm quality and DNA integrity of coke oven workers exposed to polycyclic aromatic hydrocarbons. Int J Occup Med Environ Health 2016;29(6):915–26. DOI:10.13075/ijomeh.1896.00598; Rubes J., Selevan S.G., Sram R.J. et al. GSTM1 genotype influences the susceptibility of men to sperm DNA damage associated with exposure to air pollution. Mutat Res 2007;625(1–2):20–8. DOI:10.1016/j.mrfmmm.2007.05.012; Evenson D.P., Wixon R.L. Environmental toxicants cause sperm DNA fragmentation as detected by the Sperm Chromatin Structure Assay (SCSA®). Toxicol Appl Pharmacol 2005;207(2 Suppl);532–7. DOI:10.1016/j.taap.2005.03.021; Jamal F., Haque Q.S., Singh S., Rastogi S.K. The influence of organophosphate and carbamate on sperm chromatin and reproductive hormones among pesticide sprayers. Toxicol Ind Health 2016;32(8):1527–36. DOI:10.1177/0748233714568175; Miranda-Contreras L., Cruz I., Osuna J.A. et al. [Effects of occupational exposure to pesticides on semen quality of workers in an agricultural community of Merida state, Venezuela (In Spanish)]. Invest Clin 2015;56(2):123–36.; Sánchez-Peña L.C., Reyes B.E., López-Carrillo L. et al. Organophosphorous pesticide exposure alters sperm chromatin structure in Mexican agricultural workers. Toxicol Appl Pharmacol 2004;196(1):108–13. DOI:10.1016/j.taap.2003.11.023; Gandhi J., Hernandez R.J., Chen A. et al. Impaired hypothalamicpituitary-testicular axis activity, spermatogenesis, and sperm function promote infertility in males with lead poisoning. Zygote 2017;25(2):103–10. DOI:10.1017/S0967199417000028; Zhou D.D., Hao J.L., Guo K.M. et al. Sperm quality and DNA damage in men from Jilin Province, China, who are occupationally exposed to ionizing radiation. Genet Mol Res 2016;15(1):gmr.15018078. DOI:10.4238/gmr.15018078; Zhu W.J., Qiao J. [Male reproductive toxicity of bisphenol A (In Chinese)]. Zhonghua Nan Ke Xue 2015;21(11):1026–30.; Jurewicz J., Hanke W. Exposure to phthalates: reproductive outcome and children health. A review of epidemiological studies. Int J Occup Med Environ Health 2011;24(2):115–41. DOI:10.2478/s13382-011-0022-2; Bujan L., Walschaerts M., Brugnon F. et al. Impact of lymphoma treatments on spermatogenesis and sperm deoxyribonucleic acid: a multicenter prospective study from the CECOS network. Fertil Steril 2014;102(3):667–74.e.3. DOI:10.1016/j.fertnstert.2014.06.008; O’Flaherty C., Vaisheva F., Hales B.F. et al. Characterization of sperm chromatin quality in testicular cancer and Hodgkin’s lymphoma patients prior to chemotherapy. Hum Reprod 2008;23(5):1044–52. DOI:10.1093/humrep/den081; Smit M., van Casteren N.J., Wildhagen M.F. et al. Sperm DNA integrity in cancer patients before and after cytotoxic treatment. Hum Reprod 2010;18(8):77–83. DOI:10.1093/humrep/deq104; Ståhl O., Eberhard J., Jepson K. et al. Sperm DNA integrity in testicular cancer patients. Hum Reprod 2006;21(12):3199–205. DOI:10.1093/humrep/del292; Корнеев И.А., Мацуева И.А. Мужское бесплодие, метаболический синдром и ожирение. Урологические ведомости 2021;11(2):153–62. DOI:10.17816/uroved61509; Faure C., Dupont C., Baraibar M.A. et al. In subfertile couple, abdominal fat loss in men is associated with improvement of sperm quality and pregnancy: a case-series. PLoS One 2014;9(2):e86300. DOI:10.1371/journal.pone.0086300; Jurewicz J., Radwan M., Sobala W. et al. Dietary patterns and their relationship with semen quality. Am J Mens Health 2018;12(3):575–83. DOI:10.1177/1557988315627139; Morrison C.D., Brannigan R.E. Metabolic syndrome and infertility in men. Best Pract Res Clin Obstet Gynaecol 2015;29(4):507–15. DOI:10.1016/j.bpobgyn.2014.10.006; Sharma R., Agarwal A., Harlev A., Esteves S.C. A meta-analysis to study the effects of body mass index on sperm DNA fragmentation index in reproductive age men. Fertil Steril 2017;108(3):e138–e9. DOI:10.1016/j.fertnstert.2017.07.417; 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. DOI:10.4103/1008-682X.170441; Agarwal A., Hamada A., Esteves S.C. Insight into oxidative stress in varicocele-associated male infertility: part 1. Nat Rev Urol 2012;9(12):678–90. DOI:10.1038/nrurol.2012.197; Hamada A., Esteves S.C., Agarwal A. Insight into oxidative stress in varicocele associated male infertility: part 2. Nat Rev Urol 2013;10(1):26–37. DOI:10.1038/nrurol.2012.198; Zini A., Dohle G. Are varicoceles associated with increased deoxyribonucleic acid fragmentation? Fertil Steril 2011;96(6):1283–7. DOI:10.1016/j.fertnstert.2011.10.016; Esteves S.C., Gosálvez J., López-Fernández C. et al. Diagnostic accuracy of sperm DNA degradation index (DDSi) as a potential noninvasive biomarker to identify men with varicocele-associated infertility. Int Urol Nephrol 2015;47(9):1471–7. DOI:10.1007/s11255-015-1053-6; Lira Neto F.T., Roque M., Esteves S.C. Effect of varicocelectomy on sperm deoxyribonucleic acid fragmentation rates in infertile men with clinical varicocele: a systematic review and meta-analysis. Fertil Steril 2021;116(3): 696–712. DOI:10.1016/j.fertnstert.2021.04.003; Roque M., Esteves S.C. Effect of varicocele repair on sperm DNA fragmentation: а review. Int Urol Nephrol 2018;50(4):583–603. DOI:10.1007/s11255-018-1839-4; Cantoro U., Polito M., Muzzonigro G. Reassessing the role of subclinical varicocele in infertile men with impaired semen quality: a prospective study. Urology 2015;85(4):826–30. DOI:10.1016/j.urology.2015.01.015; Kim H.J., Seo J.T., Kim K.J. et al. Clinical significance of subclinical varicocelectomy in male infertility: systematic review and metaanalysis. Andrologia 2016;48(6):654–61. DOI:10.1111/and.12495; García-Peiró A., Ribas-Maynou J., Oliver-Bonet M. et al. Multiple determinations of sperm DNA fragmentation show that varicocelectomy is not indicated for infertile patients with subclinical varicocele. BioMed Res Int 2014;2014:181396. DOI:10.1155/2014/181396; Ni K., Steger K., Yang H. et al. A comprehensive investigation of sperm DNA damage and oxidative stress injury in infertile patients with subclinical, normozoospermic, and astheno/oligozoospermic clinical varicocele. Andrology 2016;4(5):816–24. DOI:10.1111/andr.12210; Agarwal A., Rana M., Qiu E. et al. Role of oxidative stress, infection and inflammation in male infertility. Andrologia 2018;50(11):e13126. DOI:10.1111/and.13126; Боровец С.Ю., Рыбалов М.А., Горбачев А.Г., Аль-Шукри С.Х. Влияние препарата «Простатилен® АЦ» на фрагментацию ДНК сперматозоидов при лечении пациентов с хроническим абактериальным простатитом и сопутствующими нарушениями репродуктивной функции. Андрология и генитальная хирургия 2017;18(3):54–8. DOI:10.17650/2070-9781-2017- 18-3-54-58; Рогозин Д.С. Мужская фертильность: обзор литературы января – марта 2021 года. Вестник урологии 2021;9(2):142–9. DOI:10.21886/2308-6424-2021-9-2-142-149; Buck Louis G.M., Sundaram R., Schisterman E.F. et al. Semen quality and time to pregnancy: the longitudinal investigation of fertility and the environment study. Fertil Steril 2014;101(2):453–62. DOI:10.1016/j.fertnstert.2013.10.022; Carlini T., Paoli D., Pelloni M. et al. Sperm DNA fragmentation in Italian couples with recurrent pregnancy loss. Reprod Biomed Online 2017;34(1):58–65. DOI:10.1016/j.rbmo.2016.09.014; Рыжков А.И., Шорманов И.С., Соколова С.Ю. Фрагментация ДНК сперматозоидов. Есть ли связь с основными параметрами спермы и возрастом? Экспериментальная и клиническая урология 2020;4:58–64. DOI:10.29188/2222-8543-2020-13-4-58-64; Evenson D.P., Djira G., Kasperson K., Christianson J. Relationships between the age of 25,445 men attending infertility clinics and sperm chromatin structure assay (SCSA®) defined sperm DNA and chromatin integrity. Fertil Steril 2020;114(2):311–20. DOI:10.1016/j.fertnstert.2020.03.028; Sakkas D., Alvarez J.G. Sperm DNA fragmentation: mechanisms of origin, impact on reproductive outcome, and analysis. Fertil Steril 2010;93(4):1027–36. DOI:10.1016/j.fertnstert.2009.10.046; Rima D., Shiv B.K., Bhavna C. et al. Oxidative stress induced damage to paternal genome and impact of meditation and yoga – can it reduce incidence of childhood cancer? Asian Pac J Cancer Prev 2016;17(9):4517–25.; De Iuliis G.N., Thomson L.K., Mitchell L.A. et al. DNA damage in human spermatozoa is highly correlated with the efficiency of chromatin remodeling and the formation of 8-hydroxy-2’-deoxyguanosine, a marker of oxidative stress. Biol Reprod 2009;81(3): 517–24. DOI:10.1095/biolreprod.109.076836; Muratori M., Tamburrino L., Marchiani S. et al. Investigation on the origin of sperm DNA fragmentation: role of apoptosis, immaturity and oxidative stress. Mol Med 2015;21(1):109–22. DOI:10.2119/molmed.2014.00158; Agarwal A., Parekh N., Panner Selvam M.K. et al. Male Oxidative Stress Infertility (MOSI): proposed terminology and clinical practice guidelines for management of idiopathic male infertility. World J Mens Health 2019;37(3):296–312. DOI:10.5534/wjmh.190055; Chengyong W., Man Y., Mei L. et al. GSTM1 null genotype contributes to increased risk of male infertility: a meta-analysis. J Assist Reprod Genet 2012;29(8):837–45. DOI:10.1093/humrep/del338; Majzoub A., Agarwal A., Esteves S.C. Understanding sperm DNA fragmentation. Trans Androl Urol 2017;6(Suppl 4):S535–S8. DOI:10.21037/tau.2017.04.27; Champroux A., Torres-Carreira J., Gharagozloo P. et al. Mammalian sperm nuclear organization: resiliencies and vulnerabilities. Basic Clin Androl 2016;26:17. DOI:10.1186/s12610-016-0044-5; Gosálvez J., López-Fernández C., Fernández J.L. et al. Unpacking the mysteries of sperm DNA fragmentation: ten frequently asked questions. J Rep Biotech Fertil 2015;4. DOI:10.1177/2058915815594454; Oleszczuk K., Augustinsson L., Bayat N. et al. Prevalence of high DNA fragmentation index in male partners of unexplained infertile couples. Andrology 2013;1(3):357–60. DOI:10.1111/j.2047-2927.2012.00041.x; ESHRE Guideline Group on RPL, Bender Atik R., Christiansen O.B., Elson J. et al. ESHRE guideline: recurrent pregnancy loss. Hum Reprod Open 2018;2018(2):hoy004. DOI:10.1093/hropen/hoy004; Robinson L., Gallos I.D., Conner S.J. et al. The effect of sperm DNA fragmentation on miscarriage rates: a systematic review and meta-analysis. Hum Reprod Open 2012;27(10):2908–17. DOI:10.1093/humrep/des261; McQueeni D.B., Zhang J., Robins J.C. Sperm DNA fragmentation and recurrent pregnancy loss: a systematic review and metaanalysis. Fertil Steril 2019;112(1):54–60.e3. DOI:10.1016/j.fertnstert.2019.03.003; Tan J., Taskin O., Albert A., Bedaiwy M.A. Association between sperm DNA fragmentation and idiopathic recurrent pregnancy loss: a systematic review and meta-analysis. Reprod Biomed Online 2019;38(6):951–60. DOI:10.1016/j.rbmo.2018.12.029; Zidi-Jrah I., Hajlaoui A., Mougou-Zerelli S. et al. Relationship between sperm aneuploidy, sperm DNA integrity, chromatin packaging, traditional semen parameters, and recurrent pregnancy loss. Fertil Steril 2016;105(1):58–64. DOI:10.1016/j.fertnstert.2015.09.041; Ribas-Maynou J., Benet J. Single and double strand sperm DNA damage: different reproductive effects on male fertility. Genes (Basel) 2019;10(2):105. DOI:10.3390/genes10020105; Vandekerckhove F.W., De Croo I., Gerris J. et al. Sperm chromatin dispersion test before sperm preparation is predictive of clinical pregnancy in cases of unexplained infertility treated with intrauterine insemination and induction with clomiphene citrate. Front Med 2016;3:63. DOI:10.3389/fmed.2016.00063; Chen Q., Zhao J.Y., Xue X., Zhu G.X. The association between sperm DNA fragmentation and reproductive outcomes following intrauterine insemination, a meta-analysis. Reprod Toxicol 2019;86:50–5. DOI:10.1016/j.reprotox.2019.03.004; Боровец С.Ю., Аль-Шукри С.Х., Белоусов В.Я. Прогностическая значимость фрагментации ДНК сперматозоидов в отношении исходов вспомогательных репродуктивных технологий. Урологические ведомости 2015;5(1):39. DOI:10.17816/uroved5139-39; Sugihara A., Van Avermaete F., Roelant E. et al. The role of sperm DNA fragmentation testing in predicting intra-uterine insemination outcome: a systematic review and meta-analysis. Eur J Obstet Gynecol Reprod Biol 2020;244:8–15. DOI:10.1016/j.ejogrb.2019.10.005; Zhao J., Zhang Q., Wang Y., Li Y. Whether sperm deoxyribonucleic acid fragmentation has an effect on pregnancy and miscarriage after in vitro fertilization/intracytoplasmic sperm injection: а systematic review and meta-analysis. Fertil Steril 2014;102:998–1005. e1008 DOI:10.1016/j.fertnstert.2014.06.033; Xie P., Keating D., Parrella A. et al. Sperm genomic integrity by TUNEL varies throughout the male genital tract. J Urol 2020;203(4):802–8. DOI:10.1097/JU.0000000000000659; Gawecka J.E., Boaz S., Kasperson K. et al. Luminal fluid of epididymis and vas deferens contributes to sperm chromatin fragmentation. Hum Reprod 2015;30(12):2725–36. DOI:10.1093/humrep/dev245; Ambar R.F., Agarwal A., Majzoub A. et al. The use of testicular sperm for intracytoplasmic sperm injection in patients with high sperm DNA damage: a systematic review. World J Mens Health 2020;39(3):391–8. DOI:10.5534/wjmh.200084; Коршунов М.Н., Коршунова Е.С., Даренков С.П. Способ лечения мужского бесплодия при высоком показателе ДНК-фрагментации эякуляторных сперматозоидов. Патент RU 2685797C1 от 23.04.2019.; Aitken R.J. DNA damage in human spermatozoa; important contributor to mutagenesis in the offspring. Trans Androl Urol 2017;6(Suppl 4):S761–S4. DOI:10.21037/tau.2017.09.13; Bungum M., Bungum L., Lynch K.F. et al. Spermatozoa DNA damage measured by sperm chromatin structure assay (SCSA) and birth characteristics in children conceived by IVF and ICSI. Int J Androl 2012;35(4):485–90. DOI:10.1111/j.1365-2605.2011.01222.x; Koppen G., Azqueta A., Pourrut B. et al. The next three decades of the comet assay: a report of the 11th International Comet Assay Workshop. Mutagenesis 2017;32(3):397–408. DOI:10.1093/mutage/gex002; Sharma R.K., Sabanegh E., Mahfouz R. et al. TUNEL as a test for sperm DNA damage in the evaluation of male infertility. Urology 2010;76(6):1380–6. DOI:10.1016/j.urology.2010.04.036; Feijó C.M., Esteves S.C. Diagnostic accuracy of sperm chromatin dispersion test to evaluate sperm deoxyribonucleic acid damage in men with unexplained infertility. Fertil Steril 2014;101(1):58–63.e3. DOI:10.1016/j.fertnstert.2013.09.002; Pratap H., Hottigoudar S.Y., Nichanahalli K.S., Chand P. Assessment of sperm deoxyribose nucleic acid fragmentation using sperm chromatin dispersion assay. J Pharmacol Pharmacother 2017;8(2):45–9. DOI:10.4103/jpp.JPP_187_16; Evenson D.P. Sperm Chromatin Structure Assay (SCSA(®)) and other sperm DNA fragmentation tests for evaluation of sperm nuclear DNA integrity as related to fertility. Anim Reprod Sci 2016;169:56–75. DOI:10.1016/j.anireprosci.2016.01.017; Majzoub A., Agarwal A., Cho C.L., Esteves S.C. Sperm DNA fragmentation testing: a cross sectional survey on current practices of fertility specialists. Transl Androl Urol 2017;6(Suppl 4):S710–S9. DOI:10.21037/tau.2017.06.21; Dahan M.H., Mills G., Khoudja R. et al. Three hour abstinence as a treatment for high sperm DNA fragmentation: a prospective cohort study. J Assist Reprod Genet 2021;38(1):227–33. DOI:10.1007/s10815-020-01999-w; Esteves S.C. Interventions to prevent sperm DNA damage effects on reproduction. Adv Exp Med Biol 2019;1166:119–48. DOI:10.1007/978-3-030-21664-1_8; Pini T., Makloski R., Maruniak K. et al. Mitigating the effects of oxidative sperm DNA damage. Antioxidants (Basel) 2020;9(7):589. DOI:10.3390/antiox9070589; Kopa Z., Keszthelyi M., Sofikitis N. Administration of antioxidants in the infertile male: when it may have a beneficial effect? Curr Pharm Des 2020;27(23):2665–8. DOI:10.2174/1381612826666200303115552; Гамидов С.И., Овчинников Р.И., Попова А.Ю. и др. Адъювантная антиоксидантная терапия у больных бесплодием при варикоцеле. Урология 2017;2(Suppl):64–72. DOI:10.18565/urol.2017.2-supplement.64-72; Gual-Frau J., Abad C., Amengual M.J. et al. Oral antioxidant treatment partly improves integrity of human sperm DNA in infertile grade I varicocele patients. Hum Fertil (Camb) 2015;18(3):225–9. DOI:10.3109/14647273.2015.1050462; 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; https://agx.abvpress.ru/jour/article/view/633Test