-
1Academic Journal
Συγγραφείς: A. A. Policheva, E. A. Oganesyan, I. S. Yarushkina, A. S. Martynenko, E. E. Kormukhina, Ch. O. Taimova, A. R. Mustafina, V. V. Kim, A. A. Valitova, N. R. Suleimanov, K. A. Gaibaryan, M. E. Radzhabov, A. E. Baimukhambetova, A. E. Razumova, А. А. Поличева, Э. А. Оганесян, И. С. Ярушкина, А. С. Мартыненко, Е. Э. Кормухина, Ч. О. Таимова, А. Р. Мустафина, В. В. Ким, А. А. Валитова, Н. Р. Сулейманов, К. А. Гайбарян, М. Э. Раджабов, А. Е. Баймухамбетова, А. Э. Разумова
Πηγή: Obstetrics, Gynecology and Reproduction; Online First ; Акушерство, Гинекология и Репродукция; Online First ; 2500-3194 ; 2313-7347
Θεματικοί όροι: таргетная терапия, ceramides, CERs, sphingosine-1-phosphate, S1P, ovarian reserve, oocytes, folliculogenesis, ovarian cancer, polycystic ovary syndrome, endometriosis, obesity, premature ovarian failure, reproductive health, angiogenesis, apoptosis, biomarkers, targeted therapy, церамиды, сфингозин-1-фосфат, овариальный резерв, ооциты, фолликулогенез, рак яичников, синдром поликистозных яичников, эндометриоз, ожирение, преждевременная недостаточность яичников, репродуктивное здоровье, ангиогенез
Περιγραφή αρχείου: application/pdf
Relation: https://www.gynecology.su/jour/article/view/2609/1406; Петров И.А., Дмитриева М.Л., Тихоновская О.А. и др. Тканевые и молекулярные основы фолликулогенеза. Механизмы раннего фолликулярного роста. Проблемы репродукции. 2017;23(5):33–41. https://doi.org/10.17116/repro201723533-41.; Pors S.E., Harðardóttir L., Olesen H.Ø. et al. Effect of sphingosine-1-phosphate on activation of dormant follicles in murine and human ovarian tissue. Mol Hum Reprod. 2020;26(5):301–11. https://doi.org/10.1093/molehr/gaaa022.; Zhang Y., Yan Z., Qin Q. et al. Transcriptome landscape of human folliculogenesis reveals oocyte and granulosa cell interactions. Mol Cell. 2018;72(6):1021–1034.e4. https://doi.org/10.1016/j.molcel.2018.10.029.; Hernández-Coronado C.G., Guzmán A., Castillo-Juárez H. et al. Sphingosine-1-phosphate (S1P) in ovarian physiology and disease. Ann Endocrinol (Paris). 2019;80(5–6):263–72. https://doi.org/10.1016/j.ando.2019.06.003.; Pitman M., Oehler M.K., Pitson S.M. Sphingolipids as multifaceted mediators in ovarian cancer. Cell Signal. 2021;81:109949. https://doi.org/10.1016/j.cellsig.2021.109949.; Quinville B.M., Deschenes N.M., Ryckman A.E., Walia J.S. A comprehensive review: sphingolipid metabolism and implications of disruption in sphingolipid homeostasis. Int J Mol Sci. 2021;22(11):5793. https://doi.org/10.3390/ijms22115793.; Sukocheva O., Wadham C., Holmes A. et al. Estrogen transactivates EGFR via the sphingosine 1-phosphate receptor Edg-3: the role of sphingosine kinase-1. J Cell Biol. 2006;173(2):301–10. https://doi.org/10.1083/jcb.200506033.; Chou C.H., Chen M.J. The effect of steroid hormones on ovarian follicle development. Vitam Horm. 2018;107:155–75. https://doi.org/10.1016/bs.vh.2018.01.013.; Zeleznik O.A., Clish C.B., Kraft P. et al. Circulating lysophosphatidylcholines, phosphatidylcholines, ceramides, and sphingomyelins and ovarian cancer risk: a 23-year prospective study. J Natl Cancer Inst. 2020;112(6):628–36. https://doi.org/10.1093/jnci/djz195.; Janneh A.H., Ogretmen B. Targeting sphingolipid metabolism as a therapeutic strategy in cancer treatment. Cancers (Basel). 2022;14(9):2183. https://doi.org/10.3390/cancers14092183.; Gomez-Larrauri A., Das Adhikari U., Aramburu-Nuñez M. et al. Ceramide metabolism enzymes-therapeutic targets against cancer. Medicina (Kaunas). 2021;57(7):729. https://doi.org/10.3390/medicina57070729.; Companioni O., Mir C., Garcia-Mayea Y., LLeonart M.E. Targeting sphingolipids for cancer therapy. Front Oncol. 2021;11:745092. https://doi.org/10.3389/fonc.2021.745092.; Yuan Y., Jia G., Wu C. et al. Structures of signaling complexes of lipid receptors S1PR1 and S1PR5 reveal mechanisms of activation and drug recognition. Cell Res. 2021;31(12):1263–74. https://doi.org/10.1038/s41422-021-00566-x.; Lucki N.C., Sewer M.B. The interplay between bioactive sphingolipids and steroid hormones. Steroids. 2010;75(6):390–9. https://doi.org/10.1016/j.steroids.2010.01.020.; Roth Z. Symposium review: reduction in oocyte developmental competence by stress is associated with alterations in mitochondrial function. J Dairy Sci. 2018;101(4):3642–54. https://doi.org/10.3168/jds.2017-13389.; Протопопов В.А., Секунов А.В., Панов А.В., Брындина И.Г. Взаимосвязь сфинголипидных механизмов с окислительным стрессом и изменениями митохондрий при функциональной разгрузке постуральных мышц. Acta Biomedica Scientifica. 2024;9(2):228–42. https://doi.org/10.29413/ABS.2024-9.2.23.; Kujjo L.L., Perez G.I. Ceramide and mitochondrial function in aging oocytes: joggling a new hypothesis and old players. Reproduction. 2012;143(1):1–10. https://doi.org/10.1530/REP-11-0350.; Zigdon H., Kogot-Levin A., Park J.W. et al. Ablation of ceramide synthase 2 causes chronic oxidative stress due to disruption of the mitochondrial respiratory chain. J Biol Chem. 2013;288(7):4947–56. https://doi.org/10.1074/jbc.M112.402719.; Arora A.S., Jones B.J., Patel T.C. et al. Ceramide induces hepatocyte cell death through disruption of mitochondrial function in the rat. Hepatology. 1997;25(4):958–63. https://doi.org/10.1002/hep.510250428.; Malott K.F., Luderer U. Toxicant effects on mammalian oocyte mitochondria†. Biol Reprod. 2021;104(4):784–93. https://doi.org/10.1093/biolre/ioab002.; Kasapoğlu I., Seli E. Mitochondrial dysfunction and ovarian aging. Endocrinology. 2020;161(2):bqaa001. https://doi.org/10.1210/endocr/bqaa001.; Smits M.A.J., Schomakers B.V., van Weeghel M. et al. Human ovarian aging is characterized by oxidative damage and mitochondrial dysfunction. Hum Reprod. 2023;38(11):2208–20. https://doi.org/10.1093/humrep/dead177.; Lee S., Kang H.G., Jeong P.S. et al. Heat stress impairs oocyte maturation through ceramide-mediated apoptosis in pigs. Sci Total Environ. 2021;755(Pt 1):144144. https://doi.org/10.1016/j.scitotenv.2020.144144.; Hernández-Coronado C.G., Guzmán A., Espinosa-Cervantes R. et al. Sphingosine-1-phosphate and ceramide are associated with health and atresia of bovine ovarian antral follicles. Animal. 2015;9(2):308–12. https://doi.org/10.1017/S1751731114002341.; Kujjo L.L., Acton B.M., Perkins G.A. et al. Ceramide and its transport protein (CERT) contribute to deterioration of mitochondrial structure and function in aging oocytes. Mech Ageing Dev. 2013;134(1–2):43–52. https://doi.org/10.1016/j.mad.2012.12.001.; Morita Y., Tilly J.L. Oocyte apoptosis: like sand through an hourglass. Dev Biol. 1999;213(1):1–17. https://doi.org/10.1006/dbio.1999.9344.; Hernández-Coronado C.G., Guzmán A., Rodríguez A. et al. Sphingosine-1-phosphate, regulated by FSH and VEGF, stimulates granulosa cell proliferation. Gen Comp Endocrinol. 2016;236:1–8. https://doi.org/10.1016/j.ygcen.2016.06.029.; Hao X., Zhang M. Roles of sphingosine-1-phosphate in follicle development and oocyte maturation. Anim Res One Health. 2024;2(3):314–22. https://doi.org/10.1002/aro2.53.; Park J.Y., Su Y.Q., Ariga M. et al. EGF-like growth factors as mediators of LH action in the ovulatory follicle. Science. 2004;303(5658):682–4. https://doi.org/10.1126/science.1092463.; Yamanaka M., Shegogue D., Pei H. et al. Sphingosine kinase 1 (SPHK1) is induced by transforming growth factor-beta and mediates TIMP-1 up-regulation. J Biol Chem. 2004;279(52):53994–4001. https://doi.org/10.1074/jbc.M410144200.; Squecco R., Sassoli C., Nuti F. et al. Sphingosine 1-phosphate induces myoblast differentiation through Cx43 protein expression: a role for a gap junction-dependent and -independent function. Mol Biol Cell. 2006;17(11):4896–910. https://doi.org/10.1091/mbc.e06-03-0243.; Giepmans B.N., Verlaan I., Hengeveld T. et al. Gap junction protein connexin-43 interacts directly with microtubules. Curr Biol. 2001;11(17):1364–8. https://doi.org/10.1016/s0960-9822(01)00424-9.; Hao X., Wang Y., Kong N. et al. Growth factor-mobilized intracellular calcium of cumulus cells decreases natriuretic peptide receptor 2 affinity for natriuretic peptide type C and induces oocyte meiotic resumption in the mouse. Biol Reprod. 2016;95(2):45. https://doi.org/10.1095/biolreprod.116.140137.; Yuan F., Hao X., Cui Y. et al. SphK-produced S1P in somatic cells is indispensable for LH-EGFR signaling-induced mouse oocyte maturation. Cell Death Dis. 2022;13(11):963. https://doi.org/10.1038/s41419-022-05415-2.; Mostafa S., Nader N., Machaca K. Lipid signaling during gamete maturation. Front Cell Dev Biol. 2022;10:814876. https://doi.org/10.3389/fcell.2022.814876.; Birbes H., El Bawab S., Hannun Y.A., Obeid L.M. Selective hydrolysis of a mitochondrial pool of sphingomyelin induces apoptosis. FASEB J. 2001;15(14):2669–79. https://doi.org/10.1096/fj.01-0539com.; Hernández-Corbacho M.J., Salama M.F., Canals D. et al. Sphingolipids in mitochondria. Biochim Biophys Acta Mol Cell Biol Lipids. 2017;1862(1):56–68. https://doi.org/10.1016/j.bbalip.2016.09.019.; Ueda N. Ceramide-induced apoptosis in renal tubular cells: a role of mitochondria and sphingosine-1-phoshate. Int J Mol Sci. 2015;16(3):5076–124. https://doi.org/10.3390/ijms16035076.; Fisher-Wellman K.H., Hagen J.T., Neufer P.D. et al. On the nature of ceramide-mitochondria interactions – dissection using comprehensive mitochondrial phenotyping. Cell Signal. 2021;78:109838. https://doi.org/10.1016/j.cellsig.2020.109838.; Eliyahu E., Shtraizent N., Martinuzzi K. et al. Acid ceramidase improves the quality of oocytes and embryos and the outcome of in vitro fertilization. FASEB J. 2010;24(4):1229–38. https://doi.org/10.1096/fj.09-145508.; Santiquet N.W., Greene A.F, Becker J. et al. A pre-in vitro maturation medium containing cumulus oocyte complex ligand-receptor signaling molecules maintains meiotic arrest, supports the cumulus oocyte complex and improves oocyte developmental competence. Mol Hum Reprod. 2017;23(9):594–606. https://doi.org/10.1093/molehr/gax032.; Eliyahu E., Shtraizent N., Shalgi R., Schuchman E.H. Construction of conditional acid ceramidase knockout mice and in vivo effects on oocyte development and fertility. Cell Physiol Biochem. 2012;30(3):735–48. https://doi.org/10.1159/000341453.; Morita Y., Perez G.I., Paris F. et al. Oocyte apoptosis is suppressed by disruption of the acid sphingomyelinase gene or by sphingosine-1-phosphate therapy. Nat Med. 2000;6(10):1109–14. https://doi.org/10.1038/80442.; Coll O., Morales A., Fernández-Checa J.C., Garcia-Ruiz C. Neutral sphingomyelinase-induced ceramide triggers germinal vesicle breakdown and oxidant-dependent apoptosis in Xenopus laevis oocytes. J Lipid Res. 2007;48(9):1924–35. https://doi.org/10.1194/jlr.M700069-JLR200.; Yuan F., Wang Z., Sun Y. et al. Sgpl1 deletion elevates S1P levels, contributing to NPR2 inactivity and p21 expression that block germ cell development. Cell Death Dis. 2021;12(6):574. https://doi.org/10.1038/s41419-021-03848-9.; Morita Y., Tilly J.L. Sphingolipid regulation of female gonadal cell apoptosis. Ann N Y Acad Sci. 2000;905:209–20. https://doi.org/10.1111/j.1749-6632.2000.tb06551.x.; Knapp P., Chomicz K., Świderska M. et al. Unique roles of sphingolipids in selected malignant and nonmalignant lesions of female reproductive system. Biomed Res Int. 2019;2019:4376583. https://doi.org/10.1155/2019/4376583.; Kreitzburg K.M., van Waardenburg R.C.A.M., Yoon K.J. Sphingolipid metabolism and drug resistance in ovarian cancer. Cancer Drug Resist. 2018;1:181–97. https://doi.org/10.20517/cdr.2018.06.; Rutherford T., Brown W.D., Sapi E. et al. Absence of estrogen receptor-beta expression in metastatic ovarian cancer. Obstet Gynecol. 2000;96(3):417–21. https://doi.org/10.1016/s0029-7844(00)00917-0.; Jeon S.-Y., Hwang K.-A., Choi K.-C. Effect of steroid hormones, estrogen and progesterone, on epithelial mesenchymal transition in ovarian cancer development. J Steroid Biochem Mol Biol. 2016;158:1–8. https://doi.org/10.1016/j.jsbmb.2016.02.005.; Mungenast F., Thalhammer T. Estrogen biosynthesis and action in ovarian cancer. Front Endocrinol (Lausanne). 2014;5:192. https://doi.org/10.3389/fendo.2014.00192.; Giaccari C., Antonouli S., Anifandis G. et al. An update on physiopathological roles of Akt in the reprodAKTive mammalian ovary. Life (Basel). 2024;14(6):722. https://doi.org/10.3390/life14060722.; Yang Y., Lang P., Zhang X. et al. Molecular characterization of extracellular vesicles derived from follicular fluid of women with and without PCOS: integrating analysis of differential miRNAs and proteins reveals vital molecules involving in PCOS. J Assist Reprod Genet. 2023;40(3):537–52. https://doi.org/10.1007/s10815-023-02724-z.; Liu L., Yin T.L., Chen Y. et al. Follicular dynamics of glycerophospholipid and sphingolipid metabolisms in polycystic ovary syndrome patients. J Steroid Biochem Mol Biol. 2019;185:142–9. https://doi.org/10.1016/j.jsbmb.2018.08.008.; Shi Y., Zhao H., Shi Y. et al. Genome-wide association study identifies eight new risk loci for polycystic ovary syndrome. Nat Genet. 2012;44(9):1020–5. https://doi.org/10.1038/ng.2384.; Parasar P., Ozcan P., Terry K.L. Endometriosis: epidemiology, diagnosis and clinical management. Curr Obstet Gynecol Rep. 2017;6(1):34–41. https://doi.org/10.1007/s13669-017-0187-1.; Lee Y.H., Tan C.W., Venkatratnam A. et al. Dysregulated sphingolipid metabolism in endometriosis. J Clin Endocrinol Metab. 2014;99(10):E1913–21. https://doi.org/10.1210/jc.2014-1340.; Zhang Q., Duan J., Liu X., Guo S.W. Platelets drive smooth muscle metaplasia and fibrogenesis in endometriosis through epithelial-mesenchymal transition and fibroblast-to-myofibroblast transdifferentiation. Mol Cell Endocrinol. 2016;428:1–16. https://doi.org/10.1016/j.mce.2016.03.015.; Bernacchioni C., Capezzuoli T., Vannuzzi V. et al. Sphingosine 1-phosphate receptors are dysregulated in endometriosis: possible implication in transforming growth factor β-induced fibrosis. Fertil Steril. 2021;115(2):501–11. https://doi.org/10.1016/j.fertnstert.2020.08.012.; Turathum B., Gao E.M., Grataitong K. et al. Dysregulated sphingolipid metabolism and autophagy in granulosa cells of women with endometriosis. Front Endocrinol (Lausanne). 2022;13:906570. https://doi.org/10.3389/fendo.2022.906570.; Itami N., Shirasuna K., Kuwayama T., Iwata H. Palmitic acid induces ceramide accumulation, mitochondrial protein hyperacetylation, and mitochondrial dysfunction in porcine oocytes. Biol Reprod. 2018;98(5):644–53. https://doi.org/10.1093/biolre/ioy023.; Fucho R., Casals N., Serra D., Herrero L. Ceramides and mitochondrial fatty acid oxidation in obesity. FASEB J. 2017;31(4):1263–72. https://doi.org/10.1096/fj.201601156R.; Torretta E., Barbacini P., Al-Daghri N.M., Gelfi C. Sphingolipids in obesity and correlated co-morbidities: the contribution of gender, age and environment. Int J Mol Sci. 2019;20(23):5901. https://doi.org/10.3390/ijms20235901.; Samad F., Hester K.D., Yang G. et al. Altered adipose and plasma sphingolipid metabolism in obesity: a potential mechanism for cardiovascular and metabolic risk. Diabetes. 2006;55(9):2579–87. https://doi.org/10.2337/db06-0330.; Shibahara H., Ishiguro A., Inoue Y. et al. Mechanism of palmitic acid-induced deterioration of in vitro development of porcine oocytes and granulosa cells. Theriogenology. 2020;141:54–61. https://doi.org/10.1016/j.theriogenology.2019.09.006.; Levi A.J., Raynault M.F., Bergh P.A. et al. Reproductive outcome in patients with diminished ovarian reserve. Fertil Steril. 2001;76(4):666–9. https://doi.org/10.1016/s0015-0282(01)02017-9.; Timur B., Aldemir O., İnan N. et al. Clinical significance of serum and follicular fluid ceramide levels in women with low ovarian reserve. Turk J Obstet Gynecol. 2022;19(3):207–14. https://doi.org/10.4274/tjod.galenos.2022.05760.; Alizadeh J., da Silva Rosa S.C., Weng X. et al. Ceramides and ceramide synthases in cancer: Focus on apoptosis and autophagy. Eur J Cell Biol. 2023;102(3):151337. https://doi.org/10.1016/j.ejcb.2023.151337.; Nakahara T., Iwase A., Nakamura T. et al. Sphingosine-1-phosphate inhibits H2O2-induced granulosa cell apoptosis via the PI3K/Akt signaling pathway. Fertil Steril. 2012;98(4):1001–8.e1. https://doi.org/10.1016/j.fertnstert.2012.06.008.; Valtetsiotis K., Valsamakis G., Charmandari E., Vlahos N.F. Metabolic mechanisms and potential therapeutic targets for prevention of ovarian aging: data from up-to-date experimental studies. Int J Mol Sci. 2023;24(12):9828. https://doi.org/10.3390/ijms24129828.; Li F., Turan V., Lierman S. et al. Sphingosine-1-phosphate prevents chemotherapy-induced human primordial follicle death. Hum Reprod. 2014;29(1):107–13. https://doi.org/10.1093/humrep/det391.; Pascuali N., Scotti L., Di Pietro M. et al. Ceramide-1-phosphate has protective properties against cyclophosphamide-induced ovarian damage in a mice model of premature ovarian failure. Hum Reprod. 2018;33(5):844–59. https://doi.org/10.1093/humrep/dey045.; Абусуева З.А., Мухтарова М.М., Хашаева Т.Х. и др. Компаративная оценка провоспалительных цитокинов у женщин с диагностированными наследственными тромбофилиями различного генеза и их ассоциация с ранними и поздними эмбриональными потерями. Проблемы репродукции. 2022;28(3):10–7. https://doi.org/10.17116/repro20222803110.; Cianci A., Calogero A.E., Palumbo M.A. et al. Relationship between tumour necrosis factor alpha and sex steroid concentrations in the follicular fluid of women with immunological infertility. Hum Reprod. 1996;11(2):265–8. https://doi.org/10.1093/humrep/11.2.265.; Banaras S., Paracha R.Z., Nisar M. et al. System level modeling and analysis of TNF-α mediated sphingolipid signaling pathway in neurological disorders for the prediction of therapeutic targets. Front Physiol. 2022;13:872421. https://doi.org/10.3389/fphys.2022.872421.; Sukocheva O.A., Neganova M.E., Aleksandrova Y/ et al. Signaling controversy and future therapeutical perspectives of targeting sphingolipid network in cancer immune editing and resistance to tumor necrosis factor-α immunotherapy. Cell Commun Signal. 2024;22(1):251. https://doi.org/10.1186/s12964-024-01626-6.; Kolesnick R. The therapeutic potential of modulating the ceramide/sphingomyelin pathway. J Clin Invest. 2002;110(1):3–8. https://doi.org/10.1172/JCI16127.; Di Paolo A., Vignini A., Alia S. et al. Pathogenic role of the sphingosine 1-phosphate (S1P) pathway in common gynecologic disorders (GDs): a possible novel therapeutic target. Int J Mol Sci. 2022;23(21):13538. https://doi.org/10.3390/ijms232113538.; Коваль О.М., Хачанова Н.В., Журавлева М.В. и др. Безопасность воспроизведенного финголимода. Безопасность и риск фармакотерапии. 2018;6(1):23–31. https://doi.org/10.30895/2312-7821-2018-6-1-23-31.; https://www.gynecology.su/jour/article/view/2609
-
2Academic Journal
Συγγραφείς: O. V. Yakushevskaya, О. В. Якушевская
Πηγή: Meditsinskiy sovet = Medical Council; № 4 (2024); 141-146 ; Медицинский Совет; № 4 (2024); 141-146 ; 2658-5790 ; 2079-701X
Θεματικοί όροι: преждевременная недостаточность яичников, menopausal syndrome, 17β-estradiol, levonorgestrel, abnormal uterine bleeding, premature ovarian failure, климактерический синдром, 17β-эстрадиол, левоноргестрел, аномальное маточное кровотечение
Περιγραφή αρχείου: application/pdf
Relation: https://www.med-sovet.pro/jour/article/view/8201/7235; The 2022 Hormone Therapy Position Statement of The North American Menopause Society Advisory Panel. The 2022 hormone therapy position statement of The North American Menopause Society. Menopause. 2022;29(7):767–794. https://doi.org/10.1097/GME.0000000000002028.; Tanbo TG, Fedorcsak PZ. Can time to menopause be predicted? Acta Obstet Gynecol Scand. 2021;100(11):1961–1968. https://doi.org/10.1111/aogs.14253.; Юренева СВ, Якушевская ОВ, Комедина ВИ. Перименопауза – золотое время для профилактики заболеваний, ассоциированных с возрастом. Эффективная фармакотерапия. 2021;17(13):36–44. Режим доступа: https://umedp.ru/articles/perimenopauza_zolotoe_vremya_dlya_profilaktiki_zabolevaniy_assotsiirovannykh_s_vozrastom.html.; Turner RJ, Kerber IJ. Eu-estrogenemia, WHI, timing and the “geripause”. Int Urogynecol J Pelvic Floor Dysfunct. 2008;19(11):1461–1463. https://doi.org/10.1007/s00192-008-0708-6.; Юренева СВ, Дубровина АВ. Эволюция целей менопаузальной гормональной терапии. От лечения приливов к новым горизонтам кардиометаболической протекции. Акушерство и гинекология. 2018;(6):18–24. https://doi.org/10.18565/aig.2018.6.18-24.; Сметник ВП. Медицина климактерия. М.: Литера; 2009. 124 с.; Wiegratz I, Kuhl H. Progestogen therapies: differences in clinical effects? Trends Endocrinol Metab. 2004;15(6):277–285. https://doi.org/10.1016/j.tem.2004.06.006.; Raymond EG, Coeytaux F, Gemzell-Danielsson K, Moore K, Trussell J, Winikoff B. Embracing post-fertilisation methods of family planning: a call to action. J Fam Plann Reprod Health Care. 2013;39(4):244–246. https://doi.org/10.1136/jfprhc-2013-100702.; Friend DR. Development of controlled release systems over the past 50 years in the area of contraception. J Control Release. 2016;240:235–241. https://doi.org/10.1016/j.jconrel.2015.12.043.; Kubíková D. Menopauzální symptomy a hormonální substituční terapie. Praktické lékárenství. 2014;10(2). Available at: https://www.solen.sk/sk/casopisy/prakticke-lekarnictvo/menopauzalni-symptomy-a-hormonalnisubstitucni-terapie.; Basaraba CN, Westhoff CL, Pike MC, Nandakumar R, Cremers S. Estimating systemic exposure to levonorgestrel from an oral contraceptive. Contraception. 2017;95(4):398–404. https://doi.org/10.1016/j.contraception.2016.12.007.; Reinecke I, Hofmann B, Mesic E, Drenth HJ, Garmann D. An Integrated Population Pharmacokinetic Analysis to Characterize Levonorgestrel Pharmacokinetics After Different Administration Routes. J Clin Pharmacol. 2018;58(12):1639–1654. https://doi.org/10.1002/jcph.1288.; Fotherby K. Levonorgestrel. Clinical pharmacokinetics. Clin Pharmacokinet. 1995;28(3):203–215 https://doi.org/10.2165/00003088-199528030-00003.; Schindler AE, Campagnoli C, Druckmann R, Huber J, Pasqualini JR, Schweppe KW, Thijssen JH. Classification and pharmacology of progestins. Maturitas. 2003;(Suppl. 1):S7-S16. https://doi.org/10.1016/j.maturitas.2003.09.014.; Knörr K, Knörr-Gärtner H, Beller FK, Lauritzen C. Geburtshilfe und Gynäkologie: Physiologie und Pathologie der Reproduktion. Springer-Verlag; 1989. Available at: https://link.springer.com/book/10.1007/978-3-642-95583-9.; Jetley S, Rana S, Jairajpuri ZS. Morphological spectrum of endometrial pathology in middle-aged women with atypical uterine bleeding: a study of 219 cases. J Midlife Health. 2013;4(4):216–220. https://doi.org/10.4103/0976-7800.122242.; Кузнецова ИВ., Тихоновская ИВ. Менопаузальная гормональная терапия у женщин в перименопаузе с аномальными маточными кровотечениями. Доктор.Ру. 2016;120(3):25–29. Режим доступа: https://journaldoctor.ru/catalog/endokrinologiya/menopauzalnayagormonalnaya-terapiya-u-zhenshchin-v-perimenopauze-s-anomalnymimatochnymi-krovoteche/?lang=ru.; Jayaraman A, Carroll JC, Morgan TE, Lin S, Zhao L, Arimoto JM et al. 17β-estradiol and progesterone regulate expression of β-amyloid clearance factors in primary neuron cultures and female rat brain. Endocrinology. 2012;153(11):5467–5479. https://doi.org/10.1210/en.2012-1464.; Walker DM, Gore AC. Epigenetic impacts of endocrine disruptors in the brain. Front Neuroendocrinol. 2017;44:1–26. https://doi.org/10.1016/j.yfrne.2016.09.002.; Jayaraman A, Pike CJ. Differential effects of synthetic progestagens on neuron survival and estrogen neuroprotection in cultured neurons. Mol Cell Endocrinol. 2014;384(1-2):52–60. https://doi.org/10.1016/j.mce.2014.01.003.; Darney PD. The androgenicity of progestins. Am J Med. 1995;98(1A):104S–110S. https://doi.org/10.1016/s0002-9343(99)80067-9.; Davis SR, Wahlin-Jacobsen S. Testosterone in women – the clinical significance. Lancet Diabetes Endocrinol. 2015;3(12):980–992. https://doi.org/10.1016/S2213-8587(15)00284-3.; Lello S, Cavani A. Ethynilestradiol 20 mcg plus Levonorgestrel 100 mcg: Clinical Pharmacology. Int J Endocrinol. 2014;2014:102184. https://doi.org/10.1155/2014/102184.; Skouby SO. Menopause weight gain: the influence of TSEC intervention. Menopause. 2016;23(4):357–358. https://doi.org/10.1097/GME.0000000000000623.; Vinogradova Y, Coupland C, Hippisley-Cox J. Use of combined oral contraceptives and risk of venous thromboembolism: nested case-control studies using the QResearch and CPRD databases. BMJ. 2015;350:h2135. https://doi.org/10.1136/bmj.h2135.; Terauchi M, Honjo H, Mizunuma H, Aso T. Effects of oral estradiol and levonorgestrel on cardiovascular risk markers in postmenopausal women. Arch Gynecol Obstet. 2012;285(6):1647–1656. https://doi.org/10.1007/s00404-012-2222-9.; Сметник ВП, Сухих ГТ, Андреева ЕН, Балан ВЕ, Гависова АА, Григорян ОР и др. Менопаузальная гормонотерапия и сохранение здоровья женщин зрелого возраста: клинические рекомендации. М.; 2015.; Аполихина ИА, Тарнаева ЛА, Кондратьева НЕ. Результаты исследований биоэквивалентности препаратов диеногеста, прогестерона, левоноргестрела + эстрадиол у здоровых добровольцев после однократного приема натощак. Акушерство и гинекология. 2023;(1):110–121. https://doi.org/10.18565/aig.2022.304.
-
3Academic Journal
Συγγραφείς: A. Sh. Abdulaeva, P. I. Kuzmina, K. R. Bakhtiyarov, А. Ш. Абдулаева, П. И. Кузьмина, К. Р. Бахтияров
Πηγή: Meditsinskiy sovet = Medical Council; № 4 (2024); 30-36 ; Медицинский Совет; № 4 (2024); 30-36 ; 2658-5790 ; 2079-701X
Θεματικοί όροι: мезенхимальные стволовые клетки пуповины человека, premature ovarian insufficiency, premature menopause, early menopause, hormone replacement therapy, IVF, ICSI, PRP, human umbilical cord-derived mesenchymal stem cells, преждевременная недостаточность яичников, ранняя менопауза, заместительная гормональная терапия, ЭКО, ИКСИ
Περιγραφή αρχείου: application/pdf
Relation: https://www.med-sovet.pro/jour/article/view/8172/7200; Research on the menopause in the 1990s. Report of a WHO Scientific Group. World Health Organ Tech Rep Ser. 1996;866:1–107. Available at: https://pubmed.ncbi.nlm.nih.gov/8942292.; Shifren JL, Gass ML; NAMS Recommendations for Clinical Care of Midlife Women Working Group. The North American Menopause Society recommendations for clinical care of midlife women. Menopause. 2014;21(10):1038–1062. https://doi.org/10.1097/GME.0000000000000319.; InterLACE Study Team. Variations in reproductive events across life: a pooled analysis of data from 505 147 women across 10 countries. Hum Reprod. 2019;34(5):881–893. https://doi.org/10.1093/humrep/dez015.; Coulam CB, Adamson SC, Annegers JF. Incidence of Premature Ovarian Failure. Obstetrical & Gynecological Survey. 1987;42:182–183. https://doi.org/10.1097/00006254-198742030-00020.; Rostami Dovom M, Bidhendi-Yarandi R, Mohammad K, Farahmand M, Azizi F, Ramezani Tehrani F. Prevalence of premature ovarian insufficiency and its determinants in Iranian populations: Tehran lipid and glucose study. BMC Womens Health. 2021;21(1):79. https://doi.org/10.1186/s12905-021-01228-1.; Golezar S, Ramezani Tehrani F, Khazaei S, Ebadi A, Keshavarz Z. The global prevalence of primary ovarian insufficiency and early menopause: a meta-analysis. Climacteric. 2019;22(4):403–411. https://doi.org/10.1080/13697137.2019.1574738.; Mishra GD, Chung HF, Cano A, Chedraui P, Goulis DG, Lopes P et al. EMAS position statement: Predictors of premature and early natural menopause. Maturitas. 2019;123:82–88. https://doi.org/10.1016/j.maturitas.2019.03.008.; Rossetti R, Di Pasquale E, Marozzi A, Bione S, Toniolo D, Grammatico P et al. BMP15 mutations associated with primary ovarian insufficiency cause a defective production of bioactive protein. Hum Mutat. 2009;30(5):804–810. https://doi.org/10.1002/humu.20961.; Bodin L, Di Pasquale E, Fabre S, Bontoux M, Monget P, Persani L, Mulsant P. A novel mutation in the bone morphogenetic protein 15 gene causing defective protein secretion is associated with both increased ovulation rate and sterility in Lacaune sheep. Endocrinology. 2007;148(1):393–400. https://doi.org/10.1210/en.2006-0764.; Di Pasquale E, Beck-Peccoz P, Persani L. Hypergonadotropic ovarian failure associated with an inherited mutation of human bone morphogenetic protein-15 (BMP15) gene. Am J Hum Genet. 2004;75(1):106–111. https://doi.org/10.1086/422103.; Mansouri MR, Schuster J, Badhai J, Stattin EL, Lösel R, Wehling M et al. et al. Alterations in the expression, structure and function of progesterone receptor membrane component-1 (PGRMC1) in premature ovarian failure. Hum Mol Genet. 2008;17(23):3776–3783. https://doi.org/10.1093/hmg/ddn274.; Barros F, Carvalho F, Barros A, Dória S. Premature ovarian insufficiency: clinical orientations for genetic testing and genetic counseling. Porto Biomed J. 2020;5(3):e62. https://doi.org/10.1097/j.pbj.0000000000000062.; Qin Y, Jiao X, Simpson JL, Chen ZJ. Genetics of primary ovarian insufficiency: new developments and opportunities. Hum Reprod Update. 2015;21(6):787–808. https://doi.org/10.1093/humupd/dmv036.; ACOG Committee Opinion No. 469: Carrier screening for fragile X syndrome. Obstet Gynecol. 2010;116(4):1008–1010. https://doi.org/10.1097/AOG.0b013e3181fae884.; Jiao X, Qin C, Li J, Qin Y, Gao X, Zhang B et al. Cytogenetic analysis of 531 Chinese women with premature ovarian failure. Hum Reprod. 2012;27(7):2201–2207. https://doi.org/10.1093/humrep/des104.; Ishizuka B. Current Understanding of the Etiology, Symptomatology, and Treatment Options in Premature Ovarian Insufficiency (POI). Front Endocrinol (Lausanne). 2021;12:626924. https://doi.org/10.3389/fendo.2021.626924.; European Society for Human Reproduction and Embryology (ESHRE) Guideline Group on POI, Webber L, Davies M, Anderson R, Bartlett J, Braat D, Cartwright B et al. ESHRE Guideline: management of women with premature ovarian insufficiency. Hum Reprod. 2016;31(5):926–937. https://doi.org/10.1093/humrep/dew027.; Muka T, Oliver-Williams C, Kunutsor S, Laven JS, Fauser BC, Chowdhury R et al. Association of Age at Onset of Menopause and Time Since Onset of Menopause With Cardiovascular Outcomes, Intermediate Vascular Traits, and All-Cause Mortality: A Systematic Review and Meta-analysis. JAMA Cardiol. 2016;1(7):767–776. https://doi.org/10.1001/jamacardio.2016.2415.; Archer DF. Premature menopause increases cardiovascular risk. Climacteric. 2009;12(Suppl. 1):26–31. https://doi.org/10.1080/13697130903013452.; Rocca WA, Bower JH, Maraganore DM, Ahlskog JE, Grossardt BR, de Andrade M, Melton LJ 3rd. Increased risk of cognitive impairment or dementia in women who underwent oophorectomy before menopause. Neurology. 2007;69(11):1074–1083. https://doi.org/10.1212/01.wnl.0000276984.19542.e6.; Stuenkel CA, Davis SR, Gompel A, Lumsden MA, Murad MH, Pinkerton JV, Santen RJ. Treatment of Symptoms of the Menopause: An Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab. 2015;100(11):3975–4011. https://doi.org/10.1210/jc.2015-2236.; Webber L, Anderson RA, Davies M, Janse F, Vermeulen N. HRT for women with premature ovarian insufficiency: a comprehensive review. Hum Reprod Open. 2017;2017(2):hox007. https://doi.org/10.1093/hropen/hox007.; Mueck AO. Postmenopausal hormone replacement therapy and cardiovascular disease: the value of transdermal estradiol and micronized progesterone. Climacteric. 2012;15(Suppl. 1):11–17. https://doi.org/10.3109/13697137.2012.669624.; Leite-Silva P, Bedone A, Pinto-Neto AM, Costa JV, Costa-Paiva L. Factors associated with bone density in young women with karyotypically normal spontaneous premature ovarian failure. Arch Gynecol Obstet. 2009;280(2):177–181. https://doi.org/10.1007/s00404-008-0881-3.; Popat VB, Calis KA, Vanderhoof VH, Cizza G, Reynolds JC, Sebring N et al. Bone mineral density in estrogen-deficient young women. J Clin Endocrinol Metab. 2009;94(7):2277–2283. https://doi.org/10.1210/jc.2008-1878.; Cartwright B, Robinson J, Seed PT, Fogelman I, Rymer J. Hormone Replacement Therapy Versus the Combined Oral Contraceptive Pill in Premature Ovarian Failure: A Randomized Controlled Trial of the Effects on Bone Mineral Density. J Clin Endocrinol Metab. 2016;101(9):3497–3505. https://doi.org/10.1210/jc.2015-4063.; Donnez J, Dolmans MM. Fertility Preservation in Women. N Engl J Med. 2017;377(17):1657–1665. https://doi.org/10.1056/NEJMra1614676.; Chae-Kim JJ, Gavrilova-Jordan L. Premature Ovarian Insufficiency: Procreative Management and Preventive Strategies. Biomedicines. 2018;7(1):2. https://doi.org/10.3390/biomedicines7010002.; Luyckx V, Dolmans MM, Vanacker J, Legat C, Fortuño Moya C, Donnez J, Amorim CA. A new step toward the artificial ovary: survival and proliferation of isolated murine follicles after autologous transplantation in a fibrin scaffold. Fertil Steril. 2014;101(4):1149–1156. https://doi.org/10.1016/j.fertnstert.2013.12.025.; Donnez J, Dolmans MM, Demylle D, Jadoul P, Pirard C, Squifflet J et al. Livebirth after orthotopic transplantation of cryopreserved ovarian tissue. Lancet. 2004;364(9443):1405–1410. https://doi.org/10.1016/S0140-6736(04)17222-X.; Meirow D, Levron J, Eldar-Geva T, Hardan I, Fridman E, Zalel Y et al. Pregnancy after transplantation of cryopreserved ovarian tissue in a patient with ovarian failure after chemotherapy. N Engl J Med. 2005;353(3):318–321. https://doi.org/10.1056/NEJMc055237.; Donnez J, Dolmans MM, Diaz C, Pellicer A. Ovarian cortex transplantation: time to move on from experimental studies to open clinical application. Fertil Steril. 2015;104(5):1097–1098. https://doi.org/10.1016/j.fertnstert.2015.08.005.; Practice Committee of American Society For Reproductive Medicine. Increased maternal cardiovascular mortality associated with pregnancy in women with Turner syndrome. Fertil Steril. 2012;97(2):282–284. https://doi.org/10.1016/j.fertnstert.2011.11.049.; Ding X, Liu G, Xu B, Wu C, Hui N, Ni X et al. Human GV oocytes generated by mitotically active germ cells obtained from follicular aspirates. Sci Rep. 2016;6:28218. https://doi.org/10.1038/srep28218.; Zhang C, Wu J. Production of offspring from a germline stem cell line derived from prepubertal ovaries of germline reporter mice. Mol Hum Reprod. 2016;22(7):457–464. https://doi.org/10.1093/molehr/gaw030.; Silvestris E, Cafforio P, D’Oronzo S, Felici C, Silvestris F, Loverro G. In vitro differentiation of human oocyte-like cells from oogonial stem cells: single-cell isolation and molecular characterization. Hum Reprod. 2018;33(3):464–473. https://doi.org/10.1093/humrep/dex377.; Guo K, Li CH, Wang XY, He DJ, Zheng P. Germ stem cells are active in postnatal mouse ovary under physiological conditions. Mol Hum Reprod. 2016;22(5):316–328. https://doi.org/10.1093/molehr/gaw015.; Martin JJ, Woods DC, Tilly JL. Implications and Current Limitations of Oogenesis from Female Germline or Oogonial Stem Cells in Adult Mammalian Ovaries. Cells. 2019;8(2):93. https://doi.org/10.3390/cells8020093.; Silvestris E, Minoia C, Guarini A, Opinto G, Negri A, Dellino M et al. Ovarian Stem Cells (OSCs) from the Cryopreserved Ovarian Cortex: A Potential for Neo-Oogenesis in Women with Cancer-Treatment Related Infertility: A Case Report and a Review of Literature. Curr Issues Mol Biol. 2022;44(5):2309–2320. https://doi.org/10.3390/cimb44050157.; Vo KCT, Kawamura K. Ovarian Fragmentation and AKT Stimulation for Expansion of Fertile Lifespan. Front Reprod Health. 2021;3:636771. https://doi.org/10.3389/frph.2021.636771.; Suzuki N, Yoshioka N, Takae S, Sugishita Y, Tamura M, Hashimoto S et al. Successful fertility preservation following ovarian tissue vitrification in patients with primary ovarian insufficiency. Hum Reprod. 2015;30(3):608–615. https://doi.org/10.1093/humrep/deu353.; Kawamura K, Cheng Y, Suzuki N, Deguchi M, Sato Y, Takae S et al. Hippo signaling disruption and Akt stimulation of ovarian follicles for infertility treatment. Proc Natl Acad Sci U S A. 2013;110(43):17474–17479. https://doi.org/10.1073/pnas.1312830110.; Zhai J, Yao G, Dong F, Bu Z, Cheng Y, Sato Y et al. In Vitro Activation of Follicles and Fresh Tissue Auto-transplantation in Primary Ovarian Insufficiency Patients. J Clin Endocrinol Metab. 2016;101(11):4405–4412. https://doi.org/10.1210/jc.2016-1589.; Ferreri J, Fàbregues F, Calafell JM, Solernou R, Borrás A, Saco A et al. Drug-free in-vitro activation of follicles and fresh tissue autotransplantation as a therapeutic option in patients with primary ovarian insufficiency. Reprod Biomed Online. 2020;40(2):254–260. https://doi.org/10.1016/j.rbmo.2019.11.009.; Gupta S, Lodha P, Karthick MS, Tandulwadkar SR. Role of Autologous Bone Marrow-Derived Stem Cell Therapy for Follicular Recruitment in Premature Ovarian Insufficiency: Review of Literature and a Case Report of World’s First Baby with Ovarian Autologous Stem Cell Therapy in a Perimenopausal Woman of Age 45 Year. J Hum Reprod Sci. 2018;11(2):125–130. https://doi.org/10.4103/jhrs.JHRS_57_18.; Panda SR, Sachan S, Hota S. A Systematic Review Evaluating the Efficacy of Intra-Ovarian Infusion of Autologous Platelet-Rich Plasma in Patients With Poor Ovarian Reserve or Ovarian Insufficiency. Cureus. 2020;12(12):e12037. https://doi.org/10.7759/cureus.12037.
-
4Academic Journal
Συγγραφείς: Елена Николаевна Кравченко, Мария Сергеевна Воронцова
Πηγή: Мать и дитя в Кузбассе, Vol 23, Iss 1, Pp 4-10 (2022)
Θεματικοί όροι: преждевременная недостаточность яичников, бесплодие, аменорея, остеопороз, Pediatrics, RJ1-570, Gynecology and obstetrics, RG1-991
Περιγραφή αρχείου: electronic resource
Relation: https://mednauki.ru/index.php/MD/article/view/570; https://doaj.org/toc/1991-010X; https://doaj.org/toc/2542-0968
Σύνδεσμος πρόσβασης: https://doaj.org/article/5e5e1af11472473b9dfefb97ccfc42ae
-
5Report
Θεματικοί όροι: внутриматочные синехии, reduced ovarian reserve, premature ovarian insufficiency, platelet-rich plasma, плохой овариальный ответ, экстракорпоральное оплодотворение, регенерация эндометрия, intrauterine synechiae, Asherman syndrome, обогащённая тромбоцитами плазма, endometrial regeneration, poor ovarian response, репродуктология, growth factors, синдром Ашермана, факторы роста, in vitro fertilization, преждевременная недостаточность яичников, сниженный овариальный резерв, reproductive medicine
-
6Academic Journal
Συγγραφείς: R. G. Goloeva, L. P. Ananyeva, Z. S. Alekberova, S. I. Glukhova, M. V. Cherkasova, L. A. Garzanova, O. A. Koneva, T. M. Reshetnyak, Р. Г. Голоева, Л. П. Ананьева, З. С. Алекберова, С. И. Глухова, М. В. Черкасова, Л. А. Гарзанова, О. А. Конева, Т. М. Решетняк
Συνεισφορές: Работа выполнена за счет средств бюджетного финансирования на выполнение государственного задания по фундаментальной научной теме № 12204004000-7 «FURS-2022-003».
Πηγή: Rheumatology Science and Practice; Vol 61, No 3 (2023); 349–355 ; Научно-практическая ревматология; Vol 61, No 3 (2023); 349–355 ; 1995-4492 ; 1995-4484
Θεματικοί όροι: интерстициальное поражение легких, ovarian reserve, anti-Müllerian hormone, premature ovarian insufficiency, interstitial lung damage, овариальный резерв, антимюллеров гормон, преждевременная недостаточность яичников
Περιγραφή αρχείου: application/pdf
Relation: https://rsp.mediar-press.net/rsp/article/view/3362/2290; Denton C, Khanna D. Systemic sclerosis. Lancet. 2017;390 (10103):1685-1699. doi:10.1016/S0140-6736(17)30933-9; Овсянникова ОБ, Ананьева ЛП, Конева ОА, Гарзанова ЛА, Десинова ОВ, Шаяхметова РУ, и др. Трансплантация аутологичных гемопоэтических стволовых клеток в лечении системной склеродермии. Часть 2. Безопасность и осложнения. Современная ревматология. 2021;15(1):66-72. doi:10.14412/1996-7012-2021-1-66-72; Jutiviboonsuk A, Salang L, Eamudomkarn N, Mahakkanukrauh A, Suwannaroj S, Foocharoen C. Prevalence and clinical associations with premature ovarian insufficiency, early menopause, and low ovarian reserve in systemic sclerosis. Clin Rheumatol. 2021;40(6):2267-2275. doi:10.1007/s10067-020-05522-5; Steen V, Medsger T, Fertility and pregnancy outcome in women with systemic sclerosis. Arthritis Rheum. 1999;42(4):763-768. doi:10.1002/1529-0131(199904)42:43.0.CO;2-V; Bhadauria S, Moser DK, Clements PJ, Singh RR, Lachenbruch PA, Pitkin RM, et al. Genital tract abnormalities and female sexual function impairment in systemic sclerosis. Am J Obstet Gynecol. 1995;172(2 Pt 1):580-587. doi:10.1016/0002-9378(95)90576-6; Nigam A, Prakash A, Sharma S, Kumar N. Premature ovarian failure – an unusual manifestation of systemic sclerosis. J Hum Reprod Sci. 2017;10(1):58-60. doi:10.4103/jhrs.JHRS_77_16; Kauffman R, Castracane V. Premature ovarian failure associated with autoimmune polyglandular syndrome: Pathophysiological mechanisms and future fertility. J Womens Health (Larchmt). 2003;12(5):513-520. doi:10.1089/154099903766651649; Kalantaridou S, Davis S, Nelson L. Premature ovarian failure. Endocrin Metab Clin. 1998;27:989-1006. doi:10.1016/s0889-8529(05)70051-7; Anasti J. Premature ovarian failure: an update. Fertil Steril. 1998;70:1-15. doi:10.1016/s0015-0282(98)00099-5; Coulam C, Ryan R. Prevalence of circulating antibodies directed toward ovaries among women with premature ovarian failure. Am J Reprod Immunol Microbiol. 1985;9:23-24. doi:10.1111/j.1600-0897.1985.tb00336.x; Forges T, Monnier-Barbarino P, Faure G, Bene M. Autoimmunity and antigenic targets in ovarian pathology. Hum Reprod Update. 2004;10:163-175. doi:10.1093/humupd/dmh014; Gasparin AA, Chakr RM, Brenol CV, Palominos PE, Xavier RM, Souza L, et al. Hormônio anti-Mülleriano como preditor de reserva ovariana em pacientes lúpicas: Uma revisão [Anti-Müllerian hormone levels as a predictor of ovarian reserve in systemic lupus erythematosus patients: A review]. Rev Bras Reumatol. 2015;55(4): 363-367 (In Portuguese). doi:10.1016/j.rbr.2014.05.008; Lee MM, Donahoe PK. Müllerian inhibiting substance: A gonadal hormone with multiple functions. Endocr Rev. 1993;14(2):152-164. doi:10.1210/edrv-14-2-152; La Marca A, Stabile G, Artenisio A, Volpe A. Serum anti-Müllerian hormone throughout the human menstrual cycle. Hum Reprod. 2006;21(12):3103-3107. doi:10.1093/humrep/del291; Visser J, De Jong F, Laven J, Themmen A. Anti-Müllerian hormone: A new marker for ovarian function. Reproduction. 2006; 131(1):1-9. doi:10.1530/rep.1.00529; Fréour T, Mirallié S, Bach-Ngohou K, Denis M, Barrière P, Masson D. Measurement of serum anti-Müllerian hormone by Beckman Coulter ELISA and DSL ELISA: Comparison and relevance in assisted reproduction technology (ART). Clin Chim Acta. 2007;375(1-2):162-164. doi:10.1016/j.cca.2006.06.013; Broekmans F, Kwee J, Hendriks D. A systematic review of tests predicting ovarian reserve and IVF outcome. Hum Reprod Update. 2006;12(6):685-718. doi:10.1093/humupd/dml034; van den Hoogen F, Khanna D, Fransen J, Johnson SR, Baron M, Tyndall A, et al. 2013 classification criteria for systemic sclerosis: An American College of Rheumato logy/European League against Rheumatism collaborative initiative. Arthritis Rheum. 2013;65(11):2737-2747. doi:10.1002/art.38098; Henes M, Froeschlin J, Taran FA, Brucker S, Rall KK, Xenitidis T, et al. Ovarian reserve alterations in premenopausal women with chronic inflammatory rheumatic diseases: Impact of rheumatoid arthritis, Behçet’s disease and spondyloarthritis on anti-Müllerian hormone levels. Rheumatology (Oxford). 2015;54(9):1709-1712. doi:10.1093/rheumatology/kev124; Власова ГА, Перминова СГ, Кошелева НМ. Подходы к реализации репродуктивной функции у женщин с системной красной волчанкой и антифосфолипидным синдромом. Гинекология. 2021;23(2):167-172. doi:10.26442/20795696.2021.2.200739; Yalçın Bahat P, Kadiroğulları P, Topbas Selcuki NF, Yücel B, Çakmak K, Üreyen Özdemir E. Ovarian reserve in patients with ankylosing spondylitis. Arch Gynecol Obstet. 2021;303(1):189-193. doi:10.1007/s00404-020-05824-8; Brouwer J, Laven J, Hazes J, Schipper I, Dolhain R. Levels of serum anti-Mullerian hormone, a marker for ovarian reserve, in women with rheumatoid arthritis. Arthritis Care Res (Hoboken). 2013;65:1534-1538. doi:10.1002/acr.22013; Алекберова ЗС, Егорова ОН, Голоева РГ, Черкасова МВ. Антиммюллеров гормон при системной красной волчанке. Современная ревматология. 2021;15(1):105-110. doi:10.14412/1996-7012-2021-1-105-110; Luo W, Mao P, Zhang L, Chen X, Yang Z. Assessment of ovarian reserve by serum anti-Müllerian hormone in patients with systemic lupus erythematosus: A meta-analysis. Ann Palliat Med. 2020;9(2):207-215. doi:10.21037/apm.2020.02.11; Hoek A, Schoemaker J, Drexhage HA. Premature ovarian failure and ovarian autoimmunity. Endocr Rev. 1997;18(1):107-134. doi:10.1210/edrv.18.1.0291; Vural B, Caliskan E, Doger E, Ercin C. Uterine prolapse in a young nulligravida with scleroderma and premature ovarian failure. Int Urogynecol J Pelvic Floor Dysfunct. 2003;16:415-417. doi:10.1007/s00192-004-1269-y; Esteves SC, Roque M, Bedoschi GM, Conforti A, Humaidan P, Alviggi C. Defining low prognosis patients undergoing assisted reproductive technology: POSEIDON criteria-the why. Front Endocrinol (Lausanne). 2018;17(9):461. doi:10.3389/fendo.2018.00461; Сигидин ЯА, Гусева НГ, Иванова ММ. Диффузные болезни соединительной ткани. Руководство для врачей. М.:Медицина;2004.; Mok C, Lau C, Wong R. Risk factors for ovarian failure in patients with systemic lupus erythematosus receiving cyclophosphamide therapy. Arthritis Rheum. 1998;41:831-837. doi:10.1002/1529-0131 (199805)41:53.0.CO;2-1; Laskari K, Zintzaras E, Tzioufas AG. Ovarian function is preserved in women with severe systemic lupus erythematosus after a 6-month course of cyclophosphamide followed by mycophenolate mofetil. Clin Exp Rheumatol. 2010;28(1):83-86.; Ioannidis J, Katsifis G, Tzioufas A, Moutsopoulos H. Predictors of sustained amenorrhea from pulsed intravenous cyclophosphamide in premenopausal women with systemic lupus erythematosus. J Rheumatol. 2002;29(10):2129-2135.; D’Angelo S, Cuomo G, Paone C, Colutta E, La Montagna G, Valentini G. Low-dose intravenous cyclophosphamide in systemic sclerosis: A preliminary safety study. Clin Rheumatol. 2003;22(6):393-396. doi:10.1007/s10067-003-0756-8; Vinet É, Bernatsky S, Hudson M, Pineau CA, Baron M; Canadian Scleroderma Research Group. Effect of menopause on the modified Rodnan skin score in systemic sclerosis. Arthritis Res Ther. 2014;16(3):R130. doi:10.1186/ar4587
-
7Academic Journal
Συγγραφείς: L. V. Tkachenko, I. A. Gritsenko, K. Yu. Tikhaeva, N. I. Sviridova, I. S. Gavrilova, V. A. Dolgova, Л. В. Ткаченко, И. А. Гриценко, К. Ю. Тихаева, Н. И. Свиридова, И. С. Гаврилова, В. А. Долгова
Συνεισφορές: The work was supported financially by internal grants from Volgograd State Medical University (Order “On approval of the results of the competition for financing projects in the field of research activities” dated of December 7, 2020 No. 1444-KO)., Работа выполнена при финансовой поддержке внутренних грантов ФГБОУ ВО ВолгГМУ Минздрава России (Приказ «Об утверждении результатов конкурса на финансирование проектов в области научно-исследовательской деятельности» от 7 декабря 2020 г. №1444-КО).
Πηγή: Obstetrics, Gynecology and Reproduction; Vol 17, No 2 (2023); 244-251 ; Акушерство, Гинекология и Репродукция; Vol 17, No 2 (2023); 244-251 ; 2500-3194 ; 2313-7347
Θεματικοί όροι: ОР, premature ovarian failure, POF, pre-abortion counseling, ovarian reserve, OR, преждевременная недостаточность яичников, ПНЯ, доабортное консультирование, овариальный резерв
Περιγραφή αρχείου: application/pdf
Relation: https://www.gynecology.su/jour/article/view/1630/1102; Abortion care guideline. World Health Organization, 2022. Режим доступа: https://www.who.int/publications/i/item/9789240039483. [Дата обращения: 05.11.2022].; Bearak J.M., Popinchalk A., Beavin C. et al. Country-specific estimates of unintended pregnancy and abortion incidence: a global comparative analysis of levels in 2015–2019. BMJ Glob Health. 2022;7(3):e007151. https://doi:10.1136/bmjgh-2021-007151.; Здравоохранение в России. 2021. Статистический сборник. М., Росстат, 2021. 171 с. Режим доступа: http://www.demoscope.ru/weekly/2022/0931/biblio01.php. [Дата обращения: 05.11.2022].; Niinimäki M., Suhonen S., Mentula M. et al. Comparison of rates of adverse events in adolescent and adult women undergoing medical abortion: population register based study. BMJ. 2011;342:d2111. https://doi.org/10.1136/bmj.d2111.; Петров А.Ю., Узбекова Л.Д., Середа Е.В. Ближайшие и отдаленные последствия артифициального прерывания беременности. Международный научно-исследовательский журнал. 2022;(6–2):131–4. https://doi.org/10.23670/IRJ.2022.120.6.055.; Panay N., Anderson R.A., Nappi R.E. et al. Premature ovarian insufficiency: an International Menopause Society White Paper. Climacteric. 2020;23(5):426–46. https://doi.org/10.1080/13697137.2020.1804547.; European Society for Human Reproduction and Embryology (ESHRE) Guideline Group on POI; Webber L., Davies M., Anderson R. et al. ESHRE Guideline: management of women with premature ovarian insufficiency. Hum Reprod. 2016;31(5):926–37.; Wright K.J, Pace L.E., Cuneo C.N., Bartz D. Reproductive injustice at the southern border and beyond: An analysis of current events and hope for the future. Women's Health Issues. 2021;31(4):306–9. https://doi.org/10.1016/j.whi.2021.03.007.; Федеральный закон от 21.11.2011 № 323-ФЗ (ред. от 02.07.2021) «Об основах охраны здоровья граждан в Российской Федерации». M.: Министерствo здравоохранения Российской Федерации, 2021. 133 c. Режим доступа: https://edu.rosminzdrav.ru/fileadmin/user_upload/documents/zakoni/Federalnyi_zakon_ot_21_11_2011_N_323_FZ_red_ot_02_07_2021_1.pdf. [Дата обращения: 05.11.2022].; Приказ Министерства здравоохранения Российской Федерации от 20 октября 2020 г. № 1130н «Об утверждении Порядка оказания медицинской помощи по профилю "акушерство и гинекология"». M.: Министерствo здравоохранения Российской Федерации, 2020. 688 p. Режим доступа: http://perinatcentr.ru/files/N_1130.pdf. [Дата обращения: 05.11.2022].; Информационно-методическое письмо Министерства здравоохранения Российской Федерации от 17 июля 2017 года N 15-4/10/2-4792 «Психологическое консультирование женщин, планирующих искусственное прерывание беременности». M.: Министерствo здравоохранения Российской Федерации, 2017. 68 с. Режим доступа: http://old.fap.ru/upload/iblock/f7a/Metodicheskoe_pismo_po_doabortnomu_konsultirovaniyu.pdf. [Дата обращения: 05.11.2022].; Гриценко И.А., Ткаченко Л.В., Тихаева К.Ю. и др. Проблемы доабортного консультирования в РФ. VII Общероссийская конференция с международным участием «Перинатальная медицина: от прегравидарной подготовки к здоровому материнству и детству»: тезисы докладов. (18–20 февраля 2021 г., Санкт-Петербург). М.: Изд-во журнала StatusPraesens, 2021. 111.; Ткаченко Л.В., Гриценко И.А., Тихаева К.Ю. и др. Аборт: репродуктивный выбор или потеря репродукции? Медицинский алфавит. 2021;(8):44–8. https://doi.org/10.33667/2078-5631-2021-8-44-48.; Bentzen J.G., Forman J.L., Larsen E.C. et al. Maternal menopause as a predictor of anti-Müllerian hormone level and antral follicle count in daughters during reproductive age. Hum Reprod. 2013;28(1):247–55. https://doi.org/10.1093/humrep/des356.; Iwase A., Nakamura T., Nakahara T. et al. Assessment of ovarian reserve using anti-Müllerian hormone levels in benign gynecologic conditions and surgical interventions: a systematic narrative review. Reprod Biol Endocrinol. 2014;12(1):1–8. https://doi.org/10.1186/1477-7827-12-125.; Köninger A., Kauth A., Schmidt B. et al. Anti-Mullerian-hormone levels during pregnancy and postpartum. Reprod Biol Endocrinol. 2013;11(1):1–9. https://doi.org/10.1186/1477-7827-11-60.; https://www.gynecology.su/jour/article/view/1630
-
8Academic Journal
Συγγραφείς: O. S. Zahorodnia, I. B. Ventskivska, A. V. Kazak
Πηγή: Репродуктивная эндокринология, Vol 0, Iss 50, Pp 12-16 (2019)
Reproductive Endocrinology; № 50 (2019); 12-16
Репродуктивная эндокринология; № 50 (2019); 12-16
Репродуктивна ендокринологія; № 50 (2019); 12-16Θεματικοί όροι: преждевременная недостаточность яичников, заместительная гормональная терапия, эстрогены, передчасна недостатність яєчників, замісна гормональна терапія, естрогени, 03 medical and health sciences, premature ovarian insufficiency, 0302 clinical medicine, RG1-991, replace hormone therapy, Gynecology and obstetrics, estrogens, 3. Good health
Περιγραφή αρχείου: application/pdf
-
9Academic Journal
Συγγραφείς: Dolgova, V. A., Gritsenko, I. A., Tikhaeva, K. Yu., Долгова, В. А., Гриценко, И. А., Тихаева, К. Ю.
Πηγή: Сборник статей
Θεματικοί όροι: PREMATURE OVARIAN FAILURE, OVARIAN RESERVE, ABORTION, ПРЕЖДЕВРЕМЕННАЯ НЕДОСТАТОЧНОСТЬ ЯИЧНИКОВ, ОВАРИАЛЬНЫЙ РЕЗЕРВ, ИСКУССТВЕННОЕ ПРЕРЫВАНИЕ БЕРЕМЕННОСТИ
Περιγραφή αρχείου: application/pdf
Relation: Актуальные вопросы современной медицинской науки и здравоохранения: материалы VII Международной научно-практической конференции молодых учёных и студентов, Екатеринбург, 17-18 мая 2022 г.; http://elib.usma.ru/handle/usma/7369
Διαθεσιμότητα: http://elib.usma.ru/handle/usma/7369
-
10Academic Journal
Συγγραφείς: Кравченко, Елена Николаевна, Воронцова, Мария Сергеевна
Πηγή: Mother and Baby in Kuzbass; № 1 (2022): март; 4-10 ; Мать и Дитя в Кузбассе; № 1 (2022): март; 4-10 ; 2542-0968 ; 1991-010X
Θεματικοί όροι: premature ovarian insufficiency, infertility, amenorrhea, osteoporosis, преждевременная недостаточность яичников, бесплодие, аменорея, остеопороз
Περιγραφή αρχείου: text/html; application/pdf
Relation: http://mednauki.ru/index.php/MD/article/view/570/1193; http://mednauki.ru/index.php/MD/article/view/570/1212; http://mednauki.ru/index.php/MD/article/view/570
Διαθεσιμότητα: http://mednauki.ru/index.php/MD/article/view/570
-
11Academic Journal
Συγγραφείς: Мария Сергеевна Воронцова
Πηγή: Мать и дитя в Кузбассе, Vol 23, Iss 1, Pp 4-10 (2022)
Θεματικοί όροι: преждевременная недостаточность яичников, бесплодие, аменорея, остеопороз, Pediatrics, RJ1-570, Gynecology and obstetrics, RG1-991
Relation: https://mednauki.ru/index.php/MD/article/view/570; https://doaj.org/toc/1991-010X; https://doaj.org/toc/2542-0968; https://doaj.org/article/5e5e1af11472473b9dfefb97ccfc42ae
Διαθεσιμότητα: https://doaj.org/article/5e5e1af11472473b9dfefb97ccfc42ae
-
12Academic Journal
Πηγή: Мать и дитя в Кузбассе, Vol 23, Iss 1, Pp 4-10 (2022)
Θεματικοί όροι: аменорея, RG1-991, бесплодие, Gynecology and obstetrics, остеопороз, Pediatrics, преждевременная недостаточность яичников, RJ1-570
Σύνδεσμος πρόσβασης: https://doaj.org/article/5e5e1af11472473b9dfefb97ccfc42ae
-
13
-
14Academic Journal
Συγγραφείς: M. D. Salimova, Ya. G. Nadelyaeva, I. N. Danusevich, М. Д. Салимова, Я. Г. Наделяева, И. Н. Данусевич
Πηγή: Acta Biomedica Scientifica; Том 5, № 6 (2020); 42-50 ; 2587-9596 ; 2541-9420
Θεματικοί όροι: преждевременная недостаточность яичников, овариальный резерв, диагностика преждевременной недостаточности яичников, бесплодие, аменорея, антимюллеров гормон, фолликулостимулирующий гормон, premature ovarian insufficiency, ovarian reserve, diagnosis of POI, infertility, amenorrhea, anti-Müllerian hormone, follicle-stimulating hormone
Περιγραφή αρχείου: application/pdf
Relation: https://www.actabiomedica.ru/jour/article/view/2494/2079; Webber L, Davies M, Anderson R, Bartlett J, Braat D, Cartwright B, et al. ESHRE guideline: Management of women with premature ovarian insufficiency. Hum Reprod 2016; 31(5): 926-937. doi:10.1093/humrep/dew027; Табеева Г.И., Шамилова Н.Н., Жахур Н.А., Позднякова А.А., Марченко Л.А. Преждевременная недостаточность яичников – загадка XXI века. Акушерство и гинекология. 2013; (12): 16-21.; Fraison E, Crawford G, Casper G, Harris V, Ledger W. Pregnancy following diagnosis of premature ovarian insufficiency: A systematic review. Reproductive Biomed Online. 2019; 39(3): 467-476. doi:10.1016/j.rbmo.2019.04.019; Царегородцева М.В., Новикова Я.С., Подолян О.Ф. Преждевременная недостаточность яичников: новые возможности терапии. Климактерий. 2016; (3): 26-31.; Torrealday S, Kodaman P, Pal L. Premature ovarian insufficiency – an update on recent advances in understanding and management. F1000Res. 2017;6:2069 doi:10.12688/f1000research.11948.1; Baber RJ, Panay N, Fenton A, IMS Writing Group. 2016 IMS Recommendations on women’s midlife health and menopause hormone therapy. Climacteric. 2016; 19(2): 109-150. doi:10.3109/13697137.2015.1129166; Коваленко И.И., Данусевич И.Н., Наделяева Я.Г., Лазарева Л.М., Аталян А.В., Сутурина Л.В. Характеристика пациенток с преждевременной овариальной недостаточности по данным госпитального регистра. Международный журнал прикладных и фундаментальных исследований. 2017; (11-1): 53-56.; Cox L, Liu JH. Primary ovarian insufficiency: An update. Int J Womens Health. 2014; (6): 235-243. doi:10.2147/IJWH.S37636; Komorowska B. Autoimmune premature ovarian failure. Prz Menopauzalny. 2016; 15(4): 210-214. doi:10.5114/pm.2016.65666; Kruszyńska A, Słowińska-Srzednicka J. Anti-Müllerian hormone (AMH) as a good predictor of time of menopause. Prz Menopauzalny. 2017; 16(2): 47-50. doi:10.5114/pm.2017.68591; Vincent A, Farrell E. Premature menopause. In: Dvornyk V (ed.). Current topics in menopause. Sharjah: Bentham Science; 2013: 414-441. doi:10.2174/97816080545341130101; Bilgin EM, Kovanci E. Genetics of premature ovarian failure. Curr Opin Obstet Gynecol. 2015; 27(3): 167-174. doi:10.1097/GCO.0000000000000177; Pastore LM, Johnson J. The FMR1 gene, infertility, and reproductive decision-making: A review. Front Genet. 2014; (5): 195. doi:10.3389/fgene.2014.00195; Maclaran K, Nick P. Current concepts in premature ovarian insufficiency. Womens Health (Lond). 2015; 11(2): 169-182. doi:10.2217/whe.14.82; Hudson MM. Reproductive outcomes for survivors of childhood cancer. Obstet Gynecol. 2010; 116(5): 1171-1183. doi:10.1097/AOG.0b013e3181f87c4b; Nguyen HH, Milat F, Vincent A. Premature ovarian insufficiency in general practice: Meeting the needs of women. Aust Fam Physician.2017; 46(6): 360-366.; Петров И.А., Тихоновская О.А., Куприянова И.И., Окороков А.О., Логвинов С.В., Петрова М.С. и др. Механизмы вторичной недостаточности яичников при операциях на органах малого таза (экспериментальное исследование). Акушерство, гинекология и репродукция. 2015; 9(4): 6-17. doi:10.17749/2070-4968.2015.9.4.006-017; Соснова Е.А. Эмболизация маточных артерий при миоме матки у пациенток репродуктивного возраста и её роль в формировании аутоиммунного оофорита. Архив акушерства и гинекологии им. В.Ф. Снегирева. 2016; 3(2): 81-87. doi:10.18821/2313-8726-2016-3-2-81-87; Atabekoglu C, Taskin S, Kahraman K, Gemici A, Taskin EA, Ozmen B, et al. The effect of total abdominal hysterectomy on serum anti-Müllerian hormone levels: A pilot study. Climacteric. 2012; 15(4): 393-397. doi:10.3109/13697137.2011.642426; Fenton A, Panay N. Does routine gynecological surgery contribute to an early menopause? Climacteric. 2015; 15(1): 1-2. doi:10.3109/13697137.2012.647623; Muzii L, Di Tucci C, Di Feliciantonio M, Galati G, Di Donato V, Mussela A, et al. Antimüllerian hormone is reduced in the presence of ovarian endometriomas: A systematic review and meta-analysis. Fertil Steril. 2018; 110(5): 932- 940.e1. doi:10.1016/j.fertnstert.2018.06.025; Amer SA, El Shamy TT, James C, Yosef AH, Mohamed AA. The impact of laparoscopic ovarian drilling on AMH and ovarian reserve: A meta-analysis. Reproduction. 2017; 154(1): R13-R21. doi:10.1530/REP-17-0063; Dillon KE, Sammel MD, Prewitt M, Ginsberg JP, Walker D, Mersereau JE, et al. Pretreatment antimüllerian hormone levels determine rate of post therapy ovarian reserve recovery: Acute changes in ovarian reserve during and after chemotherapy. Fertil Steril. 2013; 99(2): 477-483. doi:10.1016/j.fertnstert.2012.09.039; Fenton AJ. Premature ovarian insufficiency: Pathogenesis and management. J Midlife Health. 2015; 6(4): 147-153. doi:10.4103/0976-7800.172292; Gersak K, Gersak ZM, Turcin A. Reproductive aging: Perimenopause and psychopathological symptoms. In: Drevensek G (ed.). Sex Hormones in Neurodegenerative Processes and Diseases. IntechOpen; 2018: 95-126. doi:10.5772/intechopen.74159; Committee on Gynecologic Practice. Committee Opinion No. 698: Hormone Therapy in Primary Ovarian Insufficiency. Obstet Gynecol. 2017; 129(5): e134-e141. doi:10.1097/AOG.0000000000002044; Наделяева Я.Г., Сутурина Л.В. Синдром поликистоза яичников и менопауза. Доктор.Ру. 2018; 10(154): 47-50. doi:10.31550/1727-2378-2018-154-10-47-50; Pluchino N, Carmignani A, Cubeddu A, Santoro A, Cela V, Errasti T. Androgen therapy in women: For whom and when. Arch Gynecol Obstet. 2013; 288(4): 731-737. doi:10.1007/s00404-013-2969-7; Madaeva I, Semenova N, Berdina O, Kolesnikova L. OSA syndrome and sleep structure in climacteric women in East Siberia: Ethnic aspect. Chest. 2019; 155(4): 307a. doi:10.1016/j.chest.2019.02.298; Semenova N, Madaeva I, Kolesnikova L. Insomnia in menopausal women: Racial differences. Maturitas. 2019; 124: 165. doi:10.1016/j.maturitas.2019.04.149; Мадаева И.М., Семенова Н.В., Колесникова Л.И. Этнические особенности нарушений сна у женщин климактерического периода. Журнал неврологии и психиатрии им. C.C. Корсакова. 2019; 119(4-2): 44-49. doi:10.17116/jnevro201911904244; Татарчук Т.Ф., Косей Н.В., Тутченко Т.Н. Преждевременная недостаточность яичников: синдром или диагноз. Клиническая лекция. Репродуктивная эндокринология. 2017; (2): 16-22. doi:10.18370/2309-4117.2017.34.16-22; Lewis JE, Hilditch JR, Wong CJ. Further psychometric property development of the Menopause-Specific Quality of Life questionnaire and development of a modified version, MENQOLIntervention questionnaire. Maturitas. 2005; 50(3): 209-221. doi:10.1016/j.maturitas.2004.06.015; Cramer DW, Xu H, Harlow BL. Family history as a predictor of early menopause. Fertil Steril. 1995; 64(4): 740-745. doi:10.1016/s0015-0282(16)57849-2; Luisi S, Orlandini C, Regini C, Pizzo A, Vellucci F, Petraglia F. Premature ovarian insufficiency: from pathogenesis to clinical management. J Endocrinol Invest. 2015; 38(6): 597-603. doi:10.1007/s40618-014-0231-1; Abir R, Fisch B, Nahum R, Orvieto R, Nitke S, Rafael ZB. Turner’s syndrome and fertility: current status and possible putative prospects. Hum Reprod Update. 2001; 7(6): 603- 610. doi:10.1093/humupd/7.6.603; Bukman A, Heineman MJ. Ovarian reserve testing and the use of prognostic models in patients with subfertility. Hum Reprod Update. 2001; 7(6): 581-590. doi:10.1093/humupd/7.6.581; Panay N, Kalu E. Management of premature ovarian failure. Best Pract Res Clin Obstet Gynaecol. 2008; 23(1): 129-140. doi:10.1016/j.bpobgyn.2008.10.008; Karkanaki A, Vosnakis C, Panidis D. The clinical significance of anti-mullerian hormone evaluation in gynecological endocrinology. Hormones (Athens). 2011; 10(2): 95-103. doi:10.14310/horm.2002.1299; Кузнецова И.В., Драпкина Ю.С. Роль антимюллерова гормона в женской репродукции. Медицинский алфавит. 2017; 2(10): 9-16.; Jeppesen JV, Anderson RA, Kelsey TW, Christiansen SL, Kristensen SG, Jayaprakasan K, et al. Which follicles make the most anti-Mullerian hormone in humans? Evidence for an abrupt decline in AMH production at the time of follicle selection. Mol Hum Reprod. 2013; 19(8): 519-527. doi:10.1093/molehr/gat024; Dewailly D, Andersen CY, Balen A, Broekmans F, Dilaver N, Fanchin R, et al. The physiology and clinical utility of anti-Müllerian hormone in women. Hum Reprod Update. 2014; 20(3): 370-385. doi:10.1093/humupd/dmt062; Fong SL, Visser JA, Welt CK, de Rijke YB, Eijkemans MJC, Broekmans FJ, et al. Serum anti-müllerian hormone levels in healthy females: A nomogram ranging from infancy to adulthood. J Clin Endocrinol Metab. 2012; 97(12): 4650- 4655. doi:10.1210/jc.2012-1440; Kelsey TW, Wright P, Nelson SM, Anderson RA, Wallace WHB. A validated model of serum Anti-Mullerian hormone; from conception to menopause. PLoS ONE. 2011; 6(7): e22024. doi:10.1371/journal.pone.0022024; Sahmay S, Usta TA, Erel T, Atakul N, Aydogan B. Elevated LH levels draw a stronger distinction than AMH in premature ovarian insufficiency. Climacteric. 2014; 17(2): 197-203. doi:10.3109/13697137.2013.870149; van Disseldorp J, Lambalk CB, Kwee J, Looman CWN, Eijkemans MJC, Fauser BC, et al. Comparison of inter- and intra-cycle variability of anti-Mullerian hormone and antral follicle counts. Hum Reprod. 2010; 25(1): 221-227. doi:10.1093/humrep/dep366; Randolph Jr JF, Harlow SD, Helmuth ME, Zheng H, McConnell DS. Updated assays for inhibin B and AMH provide evidence for regular episodic secretion of inhibin B but not AMH in the follicular phase of the normal menstrual cycle. Hum Reprod. 2014; 29(3): 592-600. doi:10.1093/humrep/det447; Tal R, Seifer DB. Potential mechanisms for racial and ethnic differences in antimüllerian hormone and ovarian reserve. Int J Endocrinol. 2013; 2013: 818912. doi:10.1155/2013/818912; van Rooij IAJ, Broekmans FJM, Scheffer GJ, Looman CWN, Habbema JDF, de Jong FH, et al. Serum antimullerian hormone levels best reflect the reproductive decline with age in normal women with proven fertility: A longitudinal study. Fertil Steril. 2005; 83(4): 979-987. doi:10.1016/j.fertnstert.2004.11.029; Decanter C, Morschhauser F, Pigny P, Lefebvre C, Gallo C, Dewailly D. Anti-Müllerian hormone follow-up in young women treated by chemotherapy for lymphoma: Preliminary results. Reprod Biomed Online. 2010; 20(2): 280-285. doi:10.1016/j.rbmo.2009.11.010; Rosendahl M, Andersen CY, la Cour Freiesleben N, Juul A, Løssl K, Andersen AN. Dynamics and mechanisms of chemotherapy-induced ovarian follicular depletion in women of fertile age. Fertil Steril. 2010; 94(1): 156-166. doi:10.1016/j.fertnstert.2009.02.043; Perloe M, Levy DP, Sills ES. Strategies for ascertaining ovarian reserve among women suspected of subfertility. Int J Fertil Womens Med. 2000; 45(3): 215-224.; Broer SL, Eijkemans MJC, Scheffer GJ, van Rooij IAJ, de Vet A, Themmen APN, et al. Anti-Mullerian hormone predicts menopause: A long-term follow-up study in normoovulatory women. J Clin Endocrinol Metab. 2011; 96(8): 2532-2539. doi:10.1210/jc.2010-2776; Willis SA, Kuehl TJ, Spiekerman AM, Sulak PJ. Greater inhibition of the pituitary – ovarian axis in oral contraceptive regimens with a shortened hormone-free interval. Contraception. 2006; 74(2): 100-103. doi:10.1016/j.contraception.2006.02.006; Nelson SM. Biomarkers of ovarian response: current and future applications. Fertil Steril. 2013; 99(4): 963-969. doi:10.1016/j.fertnstert.2012.11.051; https://www.actabiomedica.ru/jour/article/view/2494
-
15Academic Journal
Πηγή: Reproductive Endocrinology; № 53 (2020); 35-39
Репродуктивная эндокринология; № 53 (2020); 35-39
Репродуктивна ендокринологія; № 53 (2020); 35-39Θεματικοί όροι: 03 medical and health sciences, 0302 clinical medicine, експериментальна модель, передчасна недостатність яєчників, кріоекстракт плаценти, experimental model, premature ovarian failure, placenta cryoextract, экспериментальная модель, преждевременная недостаточность яичников, криоэкстракт плаценты, 3. Good health
Περιγραφή αρχείου: application/pdf
Σύνδεσμος πρόσβασης: http://reproduct-endo.com/article/view/207737
-
16Academic Journal
Πηγή: Reproductive Endocrinology; № 53 (2020); 87-93
Репродуктивная эндокринология; № 53 (2020); 87-93
Репродуктивна ендокринологія; № 53 (2020); 87-93Θεματικοί όροι: 03 medical and health sciences, 0302 clinical medicine, hypergonadotropic hypogonadism, premature ovarian failure, amenorrhea, infertility, azoospermia, karyotyping, mosaicism, fluorescence in situ hybridization, FISH, гипергонадотропный гипогонадизм, преждевременная недостаточность яичников, аменорея, бесплодие, азооспермия, кариотипирование, мозаицизм, флуоресцентная гибридизация in situ, гіпергонадотропний гіпогонадизм, передчасна недостатність яєчників, безпліддя, азооспермія, каріотипування, мозаїцизм, флуоресцентна гібридизація іn situ, 3. Good health
Περιγραφή αρχείου: application/pdf
Σύνδεσμος πρόσβασης: http://reproduct-endo.com/article/view/207736
-
17Academic Journal
Πηγή: Scientific digest of association of obstetricians and gynecologists of Ukraine; № 1(41) (2018); 140-145
СБОРНИК НАУЧНЫХ ТРУДОВ Ассоциации акушеров-гинекологов Украины; № 1(41) (2018); 140-145
Збірник наукових праць Асоціації акушерів-гінекологів України; № 1(41) (2018); 140-145Θεματικοί όροι: Осложнения после химиотерапии, преждевременная недостаточность яичников, криоэкстракт плаценты, мезенхимальные стволовые клетки из жировой ткани, Complications after chemotherapy, premature ovarian failure, cryoextract of placenta, mesenchymal stem cells of adipose tissue, Ускладнення після хіміотерапії, передчасна недостатність яєчників, кріоекстракт плаценти, мезенхімальні стовбурові клітини з жирової тканини, [618.11-008.64-02:616-006.6-085.28]-092.9-035-085.361.013.85:602.9, 3. Good health
Περιγραφή αρχείου: application/pdf
Σύνδεσμος πρόσβασης: http://zbirnyk.aagu.com.ua/article/view/172562
-
18Academic Journal
Συγγραφείς: ПОЗДНЯКОВА АННА АЛЕКСЕЕВНА, ВОЛОДИНА МАРИЯ АЛЕКСАНДРОВНА, РШТУНИ САНДРА ДЖОНИЕВНА, МАРЧЕНКО ЛАРИСА АНДРЕЕВНА, ВЫСОКИХ МИХАИЛ ЮРЬЕВИЧ
Περιγραφή αρχείου: text/html
-
19
-
20Academic Journal
Πηγή: Акушерство, гинекология и репродукция.
Θεματικοί όροι: ПРЕЖДЕВРЕМЕННАЯ НЕДОСТАТОЧНОСТЬ ЯИЧНИКОВ,PREMATURE OVARIAN INSUFFICIENСY,МИТОХОНДРИЯ,MITOCHONDRIA,МИТОХОНДРИАЛЬНАЯ ДИСФУНКЦИЯ,MITOCHONDRIAL DISFUNCTION,ОКИСЛИТЕЛЬНЫЙ СТРЕСС,OXIDATIVE STRESS,АПОПТОЗ,APOPTOSIS,TSPO,P66SHC, 3. Good health
Περιγραφή αρχείου: text/html