Половые гормоны и когнитивные функции: современные данные

Автор: Булгакова Светлана Викторовна, Романчук Наталья Петровна

Журнал: Бюллетень науки и практики @bulletennauki

Рубрика: Медицинские науки

Статья в выпуске: 3 т.6, 2020 года.

Бесплатный доступ

Гормоны оси гипоталамус-гипофиз-гонады, регулирующие репродуктивную функцию, оказывают множественное влияние на развитие и функции головного мозга. Ряд исследований показали половые различия в когнитивных функциях как в норме, так и при различных заболеваниях головного мозга, что может быть частично связано с половыми гормонами. Целью данной статьи явился анализ литературы о влиянии половых гормонов на когнитивные функции на протяжении всей жизни. роли половых гормонов в снижении когнитивных способностей, особенно при болезни Альцгеймера, возможности экзогенного введения половых гормонов для улучшения когнитивных функций и/или снижения риска развития болезни Альцгеймера. Врач и нейрофизиолог: современное решение проблемы реабилитации «когнитивного мозга» Homo sapiens с применением с одной стороны, инструментов и технологий искусственного интеллекта, а с другой - мультидисциплинарное взаимодействие нейрофизиолога с клиническим «универсальным» специалистом в области неврологии, психиатрии, психотерапии, психоанализа, эндокринологии и гериатрии.

Еще

Когнитивные функции, половые гормоны, старение

Короткий адрес: https://sciup.org/14116040

IDR: 14116040   |   DOI: 10.33619/2414-2948/52/09

Список литературы Половые гормоны и когнитивные функции: современные данные

  • Романчук П. И. Возраст и микробиота: эпигенетическая и диетическая защита, эндотелиальная и сосудистая реабилитация, новая управляемая здоровая биомикробиота // Бюллетень науки и практики. 2020. Т. 6. №2. С. 67-110. DOI: 10.33619/2414-2948/51/07
  • Романчук П. И., Волобуев А. Н. Современные инструменты и методики эпигенетической защиты здорового старения и долголетия Homo sapiens // Бюллетень науки и практики. 2020. Т. 6. №1. С. 43-70. DOI: 10.33619/2414-2948/50/06
  • Романчук Н. П., Романчук П. И. Нейрофизиология и нейрореабилитация когнитивных нарушений и расстройств // Бюллетень науки и практики. 2019. Т. 5. №11. С. 176-196. DOI: 10.33619/2414-2948/48/19
  • Романчук Н. П., Пятин В. Ф. Мелатонин: нейрофизиологические и нейроэндокринные аспекты // Бюллетень науки и практики. 2019. Т. 5. №7. С. 71-85. DOI: 10.33619/2414-2948/44/08
  • Пятин В. Ф., Романчук Н. П., Романчук П. И., Волобуев А. Н, Мозг, глаза, свет: биоэлектромагнитизм света и нейрореабилитация когнитивных нарушений // Бюллетень науки и практики. 2019. Т. 5. №12. С. 129-155. DOI: 10.33619/2414-2948/49/14
  • Булгакова С. В., Романчук П. И., Волобуев А. Н. Нейросети: нейроэндокринология и болезнь Альцгеймера // Бюллетень науки и практики. 2019. Т. 5. №6. С. 112-128.
  • DOI: 10.33619/2414-2948/43/16
  • Булгакова С. В., Романчук П. И., Волобуев А. Н. Клинико-биофизические принципы лечения сосудистой деменции и болезни Альцгеймера // Бюллетень науки и практики. 2019. Т. 5. №5. С. 57-72.
  • DOI: 10.33619/2414-2948/42/08
  • Волобуев А. Н., Романчук П. И., Булгакова С. В. Нейросеть "мозг-микробиота": регуляция "висцерального" мозга и накопление когнитивной памяти // Бюллетень науки и практики. 2019. Т. 5. №2. С. 33-52.
  • DOI: 10.33619/2414-2948/39/05
  • Gurvich C. et al. Sex differences and the influence of sex hormones on cognition through adulthood and the aging process // Brain sciences. 2018. V. 8. №9. P. 163.
  • DOI: 10.3390/brainsci8090163
  • Meethal S. V., Atwood C. S. The role of hypothalamic-pituitary-gonadal hormones in the normal structure and functioning of the brain // Cell Mol Life Sci. 2005. V. 62. №3. P. 257-270.
  • DOI: 10.1007/s00018-004-4381-3
  • Marrocco J., McEwen B. S. Sex in the brain: hormones and sex differences // Dialogues in clinical neuroscience. 2016. V. 18. №4. P. 373.
  • Tuscher J. J. et al. Estradiol-mediated spine changes in the dorsal hippocampus and medial prefrontal cortex of ovariectomized female mice depend on ERK and mTOR activation in the dorsal hippocampus // Journal of Neuroscience. 2016. V. 36. №5. P. 1483-1489.
  • DOI: 10.1523/JNEUROSCI.3135-15.2016
  • Woolley C. S. et al. Naturally occurring fluctuation in dendritic spine density on adult hippocampal pyramidal neurons // Journal of neuroscience. 1990. V. 10. №12. P. 4035-4039.
  • DOI: 10.1523/JNEUROSCI.10-12-04035
  • Hara Y. et al. Estrogen effects on cognitive and synaptic health over the lifecourse // Physiological reviews. 2015. V. 95. №3. P. 785-807.
  • DOI: 10.1152/physrev.00036.2014
  • Milne M. R. et al. Estradiol modulation of neurotrophin receptor expression in female mouse basal forebrain cholinergic neurons in vivo // Endocrinology. 2015. V. 156. №2. P. 613-626.
  • DOI: 10.1210/en.2014-1669
  • Mennenga S. E. et al. Understanding the cognitive impact of the contraceptive estrogen Ethinyl Estradiol: tonic and cyclic administration impairs memory, and performance correlates with basal forebrain cholinergic system integrity // Psychoneuroendocrinology. 2015. V. 54. P. 1-13.
  • DOI: 10.1016/j.psyneuen.2015.01.002
  • Sinclair D. et al. Impacts of stress and sex hormones on dopamine neurotransmission in the adolescent brain // Psychopharmacology. 2014. V. 231. №8. P. 1581-1599.
  • DOI: 10.1007/s00213-013-3415-z
  • Luine V. Estradiol: mediator of memories, spine density and cognitive resilience to stress in female rodents // The Journal of steroid biochemistry and molecular biology. 2016. V. 160. P. 189-195.
  • DOI: 10.1016/j.jsbmb.2015.07.022
  • Engler-Chiurazzi E. B., Singh M., Simpkins J. W. From the 90' s to now: A brief historical perspective on more than two decades of estrogen neuroprotection // Brain research. 2016. V. 1633. P. 96-100.
  • DOI: 10.1016/j.brainres.2015.12.044
  • Luine V. N. Estradiol and cognitive function: past, present and future // Hormones and behavior. 2014. V. 66. №4. P. 602-618.
  • DOI: 10.1016/j.yhbeh.2014.08.011
  • Galea L. A. M. et al. Why estrogens matter for behavior and brain health // Neuroscience & Biobehavioral Reviews. 2017. V. 76. P. 363-379.
  • DOI: 10.1016/j.neubiorev.2016.03.024
  • Joffe H. et al. Estrogen therapy selectively enhances prefrontal cognitive processes: a randomized, double-blind, placebo-controlled study with functional magnetic resonance imaging in perimenopausal and recently postmenopausal women // Menopause. 2006. V. 13. №3. С. 411-422.
  • DOI: 10.1097/01.gme.0000189618.48774.7b
  • Duff S. J., Hampson E. A beneficial effect of estrogen on working memory in postmenopausal women taking hormone replacement therapy // Hormones and behavior. 2000. V. 38. №4. P. 262-276.
  • DOI: 10.1006/hbeh.2000.1625
  • Krug R., Born J., Rasch B. A 3-day estrogen treatment improves prefrontal cortex-dependent cognitive function in postmenopausal women // Psychoneuroendocrinology. 2006. V. 31. №8. P. 965-975.
  • DOI: 10.1016/j.psyneuen.2006.05.007
  • Toffoletto S. et al. Emotional and cognitive functional imaging of estrogen and progesterone effects in the female human brain: a systematic review // Psychoneuroendocrinology. 2014. V. 50. P. 28-52.
  • DOI: 10.1016/j.psyneuen.2014.07.025
  • Rossetti M. F. et al. Oestrogens and progestagens: synthesis and action in the brain // Journal of neuroendocrinology. 2016. V. 28. №7.
  • DOI: 10.1111/jne.12402
  • Singh M., Su C. Progesterone and neuroprotection // Hormones and behavior. 2013. V. 63. №2. P. 284-290.
  • DOI: 10.1016/j.yhbeh.2012.06.003
  • Blair J. A. et al. Luteinizing hormone: evidence for direct action in the CNS // Hormones and behavior. 2015. V. 76. P. 57-62.
  • DOI: 10.1016/j.yhbeh.2015.06.020
  • Koebele S. V., Bimonte-Nelson H. A. The endocrine-brain-aging triad where many paths meet: female reproductive hormone changes at midlife and their influence on circuits important for learning and memory // Experimental gerontology. 2017. V. 94. P. 14-23.
  • DOI: 10.1016/j.exger.2016.12.011
  • Lei Z. M. et al. Novel expression of human chorionic gonadotropin/luteinizing hormone receptor gene in brain // Endocrinology. 1993. V. 132. №5. P. 2262-2270.
  • DOI: 10.1210/endo.132.5.8477671
  • Rodrigues M. A. et al. Gonadotropins and cognition in older women // Journal of Alzheimer's Disease. 2008. V. 13. №3. P. 267-274.
  • DOI: 10.3233/JAD-2008-13304
  • Hyde J. S. Gender similarities and differences // Annual review of psychology. 2014. V. 65. P. 373-398.
  • DOI: 10.1146/annurev-psych-010213-115057
  • Hyde J. S. Sex and cognition: gender and cognitive functions // Current opinion in neurobiology. 2016. V. 38. P. 53-56.
  • DOI: 10.1016/j.conb.2016.02.007
  • Miller D. I., Halpern D. F. The new science of cognitive sex differences // Trends in cognitive sciences. 2014. V. 18. №1. P. 37-45.
  • DOI: 10.1016/j.tics.2013.10.011
  • Li R., Singh M. Sex differences in cognitive impairment and Alzheimer's disease // Frontiers in neuroendocrinology. 2014. V. 35. №3. P. 385-403.
  • DOI: 10.1016/j.yfrne.2014.01.002
  • Auyeung B., Lombardo M. V., Baron-Cohen S. Prenatal and postnatal hormone effects on the human brain and cognition // Pflügers Archiv-European Journal of Physiology. 2013. V. 465. №5. P. 557-571.
  • DOI: 10.1007/s00424-013-1268-2
  • Beck-Peccoz P. et al. Maturation of hypothalamic-pituitary-gonadal function in normal human fetuses: circulating levels of gonadotropins, their common a-subunit and free testosterone, and discrepancy between immunological and biological activities of circulating follicle-stimulating hormone // The Journal of Clinical Endocrinology & Metabolism. 1991. V. 73. №3. P. 525-532.
  • DOI: 10.1210/jcem-73-3-525
  • Vogel J. J., Bowers C. A., Vogel D. S. Cerebral lateralization of spatial abilities: a meta-analysis // Brain and cognition. 2003. V. 52. №2. P. 197-204.
  • DOI: 10.1016/S0278-2626(03)00056-3
  • Pfannkuche K. A., Bouma A., Groothuis T. G. G. Does testosterone affect lateralization of brain and behaviour? A meta-analysis in humans and other animal species // Philosophical Transactions of the Royal Society B: Biological Sciences. 2009. V. 364. №1519. P. 929-942.
  • DOI: 10.1098/rstb.2008.0282
  • Puts D. A. et al. Spatial ability and prenatal androgens: meta-analyses of congenital adrenal hyperplasia and digit ratio (2D: 4D) studies // Archives of sexual behavior. 2008. V. 37. №1. P. 100.
  • DOI: 10.1007/s10508-007-9271-3
  • Heil M. et al. Mental rotation in female fraternal twins: Evidence for intra-uterine hormone transfer? // Biological psychology. 2011. V. 86. №1. P. 90-93.
  • DOI: 10.1016/j.biopsycho.2010.11.002
  • Berenbaum S. A. Beyond pink and blue: The complexity of early androgen effects on gender development // Child development perspectives. 2018. V. 12. №1. P. 58-64.
  • DOI: 10.1111/cdep.12261
  • Berenbaum S. A., Beltz A. M. How early hormones shape gender development // Current opinion in behavioral sciences. 2016. V. 7. P. 53-60.
  • DOI: 10.1016/j.cobeha.2015.11.011
  • Schulz K. M., Molenda-Figueira H. A., Sisk C. L. Back to the future: the organizational-activational hypothesis adapted to puberty and adolescence // Hormones and behavior. 2009. V. 55. №5. P. 597-604.
  • DOI: 10.1016/j.yhbeh.2009.03.010
  • Schulz K. M., Sisk C. L. The organizing actions of adolescent gonadal steroid hormones on brain and behavioral development // Neuroscience & Biobehavioral Reviews. 2016. V. 70. P. 148-158.
  • DOI: 10.1016/j.neubiorev.2016.07.036
  • Herting M. M. et al. The role of testosterone and estradiol in brain volume changes across adolescence: a longitudinal structural MRI study // Human brain mapping. 2014. V. 35. №11. P. 5633-5645.
  • DOI: 10.1002/hbm.22575
  • Vijayakumar N. et al. Puberty and the human brain: Insights into adolescent development // Neuroscience & Biobehavioral Reviews. 2018. V. 92. P. 417-436.
  • DOI: 10.1016/j.neubiorev.2018.06.004
  • Berenbaum S. A., Beltz A. M. Sexual differentiation of human behavior: effects of prenatal and pubertal organizational hormones // Frontiers in neuroendocrinology. 2011. V. 32. №2. P. 183-200.
  • DOI: 10.1016/j.yfrne.2011.03.001
  • Beltz A. M., Berenbaum S. A. Cognitive effects of variations in pubertal timing: is puberty a period of brain organization for human sex-typed cognition? // Hormones and behavior. 2013. V. 63. №5. P. 823-828.
  • DOI: 10.1016/j.yhbeh.2013.04.002
  • Lenton E. A. et al. Individual variation in gonadotrophin and steroid concentrations and in the lengths of the follicular and luteal phases in women with regular menstrual cycles // Clinical reproduction and fertility. 1983. V. 2. №2. P. 143-150. PMID:
  • ISBN: 6423258
  • Sundström Poromaa I., Gingnell M. Menstrual cycle influence on cognitive function and emotion processing - from a reproductive perspective // Frontiers in neuroscience. 2014. V. 8. P. 380.
  • DOI: 10.3389/fnins.2014.00380
  • Courvoisier D. S. et al. Sex hormones and mental rotation: an intensive longitudinal investigation // Hormones and Behavior. 2013. V. 63. №2. P. 345-351.
  • DOI: 10.1016/j.yhbeh.2012.12.007
  • Barry J. A., Parekh H. S. K., Hardiman P. J. Visual-spatial cognition in women with polycystic ovarian syndrome: the role of androgens // Human Reproduction. 2013. V. 28. №10. P. 2832-2837.
  • DOI: 10.1093/humrep/det335
  • Warren A. M. et al. A systematic review of the impact of oral contraceptives on cognition // Contraception. 2014. V. 90. №2. P. 111-116.
  • DOI: 10.1016/j.contraception.2014.03.015
  • Gogos A. Natural and synthetic sex hormones: effects on higher-order cognitive function and prepulse inhibition // Biological psychology. 2013. V. 93. №1. P. 17-23.
  • DOI: 10.1016/j.biopsycho.2013.02.001
  • Mordecai K. L., Rubin L. H., Maki P. M. Effects of menstrual cycle phase and oral contraceptive use on verbal memory // Hormones and Behavior. 2008. V. 54. №2. P. 286-293.
  • DOI: 10.1016/j.yhbeh.2008.03.006
  • Wharton W. et al. Oral contraceptives and androgenicity: Influences on visuospatial task performance in younger individuals // Experimental and clinical psychopharmacology. 2008. V. 16. №2. P. 156. https://psycnet.apa.org/doi/
  • DOI: 10.1037/1064-1297.16.2.156
  • Griksiene R., Ruksenas O. Effects of hormonal contraceptives on mental rotation and verbal fluency // Psychoneuroendocrinology. 2011. V. 36. №8. P. 1239-1248.
  • DOI: 10.1016/j.psyneuen.2011.03.001
  • Davis S. R., Wahlin-Jacobsen S. Testosterone in women - the clinical significance // The Lancet Diabetes & Endocrinology. 2015. V. 3. №12. P. 980-992.
  • DOI: 10.1016/S2213-8587(15)00284-3
  • Aleman A. et al. A single administration of testosterone improves visuospatial ability in young women // Psychoneuroendocrinology. 2004. V. 29. №5. P. 612-617.
  • DOI: 10.1016/S0306-4530(03)00089-1
  • Postma A. et al. Effects of testosterone administration on selective aspects of object-location memory in healthy young women // Psychoneuroendocrinology. 2000. V. 25. №6. P. 563-575.
  • DOI: 10.1016/S0306-4530(00)00010-X
  • Epperson C. N., Sammel M. D., Freeman E. W. Menopause effects on verbal memory: findings from a longitudinal community cohort // The Journal of clinical endocrinology & metabolism. 2013. V. 98. №9. P. 3829-3838.
  • DOI: 10.1210/jc.2013-1808
  • Schaafsma M., Homewood J., Taylor A. Subjective cognitive complaints at menopause associated with declines in performance of verbal memory and attentional processes // Climacteric. 2010. V. 13. №1. P. 84-98.
  • DOI: 10.3109/13697130903009187
  • Weber M. et al. Reconciling subjective memory complaints with objective memory performance in the menopausal transition // Menopause (New York, NY). 2012. V. 19. №7. P. 735.
  • DOI: 10.1097/gme.0b013e318241fd22
  • Weber M. T., Maki P. M., McDermott M. P. Cognition and mood in perimenopause: a systematic review and meta-analysis // The Journal of steroid biochemistry and molecular biology. 2014. V. 142. P. 90-98.
  • DOI: 10.1016/j.jsbmb.2013.06.001
  • Berent-Spillson A. et al. Hormonal environment affects cognition independent of age during the menopause transition // The Journal of Clinical Endocrinology & Metabolism. 2012. V. 97. №9. P. E1686-E1694.
  • DOI: 10.1210/jc.2012-1365
  • LeBlanc E. S. et al. Hot flashes and estrogen therapy do not influence cognition in early menopausal women // Menopause. 2007. V. 14. №2. P. 191-202.
  • DOI: 10.1097/01.gme.0000230347.28616.1c
  • Rocca W. A. et al. Increased risk of cognitive impairment or dementia in women who underwent oophorectomy before menopause // Neurology. 2007. V. 69. №11. P. 1074-1083.
  • DOI: 10.1212/01.wnl.0000276984.19542.e6
  • Phung T. K. T. et al. Hysterectomy, oophorectomy and risk of dementia: a nationwide historical cohort study // Dementia and geriatric cognitive disorders. 2010. V. 30. №1. P. 43-50.
  • DOI: 10.1159/000314681
  • Bove R. et al. Age at surgical menopause influences cognitive decline and Alzheimer pathology in older women // Neurology. 2014. V. 82. №3. P. 222-229.
  • DOI: 10.1212/WNL.0000000000000033
  • Rocca W. A., Grossardt B. R., Shuster L. T. Oophorectomy, estrogen, and dementia: a 2014 update // Molecular and cellular endocrinology. 2014. V. 389. №1-2. P. 7-12.
  • DOI: 10.1016/j.mce.2014.01.020
  • Park D. C. et al. Models of visuospatial and verbal memory across the adult life span // Psychology and aging. 2002. V. 17. №2. P. 299. https://psycnet.apa.org/doi/
  • DOI: 10.1037/0882-7974.17.2.299
  • Finkel D. et al. Surprising lack of sex differences in normal cognitive aging in twins // The International Journal of Aging and Human Development. 2006. V. 62. №4. P. 335-357. https://doi.org/10.2190%2FC39X-9QHY-49DM-X9GJ
  • De Frias C. M., Nilsson L. G., Herlitz A. Sex differences in cognition are stable over a 10-year period in adulthood and old age // Aging, Neuropsychology, and Cognition. 2006. V. 13. №3-4. P. 574-587.
  • DOI: 10.1080/13825580600678418
  • Ferreira L. et al. Rate of cognitive decline in relation to sex after 60 years-of-age: A systematic review // Geriatrics & gerontology international. 2014. V. 14. №1. P. 23-31.
  • DOI: 10.1111/ggi.12093
  • McCarrey A. C. et al. Sex differences in cognitive trajectories in clinically normal older adults // Psychology and aging. 2016. Т. 31. №2. С. 166. https://psycnet.apa.org/doi/
  • DOI: 10.1037/pag0000070
  • Zárate S., Stevnsner T., Gredilla R. Role of estrogen and other sex hormones in brain aging. Neuroprotection and DNA repair // Frontiers in aging neuroscience. 2017. V. 9. P. 430.
  • DOI: 10.3389/fnagi.2017.00430
  • Barha C. K., Galea L. A. M. Influence of different estrogens on neuroplasticity and cognition in the hippocampus // Biochimica et Biophysica Acta (BBA)-General Subjects. 2010. V. 1800. №10. P. 1056-1067.
  • DOI: 10.1016/j.bbagen.2010.01.006
  • Rannevik G. et al. A longitudinal study of the perimenopausal transition: altered profiles of steroid and pituitary hormones, SHBG and bone mineral density // Maturitas. 1995. V. 21. №2. P. 103-113.
  • DOI: 10.1016/0378-5122(94)00869-9
  • Henderson V. W. Gonadal hormones and cognitive aging: a midlife perspective // Women's Health. 2011. V. 7. №1. P. 81-93. https://doi.org/10.2217%2FWHE.10.87
  • Araujo A. B., Wittert G. A. Endocrinology of the aging male // Best practice & research Clinical endocrinology & metabolism. 2011. Т. 25. №2. P. 303-319.
  • DOI: 10.1016/j.beem.2010.11.004
  • Barron A. M., Pike C. J. Sex hormones, aging, and Alzheimer's disease // Frontiers in bioscience (Elite edition). 2012. V. 4. P. 976.
  • Laws K. R., Irvine K., Gale T. M. Sex differences in Alzheimer's disease // Current opinion in psychiatry. 2018. V. 31. №2. P. 133-139.
  • DOI: 10.1097/YCO.0000000000000401
  • Gao S. et al. The relationships between age, sex, and the incidence of dementia and Alzheimer disease: a meta-analysis // Archives of general psychiatry. 1998. V. 55. №9. P. 809-815.
  • DOI: 10.1001/archpsyc.55.9.809
  • Georgakis M. K. et al. Age at menopause and duration of reproductive period in association with dementia and cognitive function: a systematic review and meta-analysis // Psychoneuroendocrinology. 2016. V. 73. P. 224-243.
  • DOI: 10.1016/j.psyneuen.2016.08.003
  • Gregory C. W. et al. Antigonadotropins: a novel strategy to halt Alzheimer's disease progression // Current pharmaceutical design. 2006. V. 12. №6. P. 685-690.
  • DOI: 10.2174/138161206775474288
  • Al-Hader A. A. et al. Fetal rat brains contain luteinizing hormone/human chorionic gonadotropin receptors // Early pregnancy: biology and medicine: the official journal of the Society for the Investigation of Early Pregnancy. 1997. V. 3. №4. P. 323-329. PMID:
  • ISBN: 10086084
  • Al-Hader A. A., Lei Z. M., Rao C. V. Neurons from fetal rat brains contain functional luteinizing hormone/chorionic gonadotropin receptors // Biology of reproduction. 1997. V. 56. №5. P. 1071-1076.
  • DOI: 10.1095/biolreprod56.5.1071
  • Hyde Z. et al. Higher luteinizing hormone is associated with poor memory recall: the health in men study // Journal of Alzheimer's Disease. 2010. V. 19. №3. P. 943-951.
  • DOI: 10.3233/JAD-2010-1342
  • Verdile G. et al. Luteinizing hormone levels are positively correlated with plasma amyloid-β protein levels in elderly men // Journal of Alzheimer's Disease. 2008. V. 14. №2. P. 201-208.
  • DOI: 10.3233/JAD-2008-14208
  • Mielke M. M., Vemuri P., Rocca W. A. Clinical epidemiology of Alzheimer's disease: assessing sex and gender differences // Clinical epidemiology. 2014. V. 6. P. 37.
  • DOI: 10.2147/CLEP.S37929
  • Laws K. R., Irvine K., Gale T. M. Sex differences in cognitive impairment in Alzheimer's disease // World journal of psychiatry. 2016. V. 6. №1. P. 54.
  • DOI: 10.5498/wjp.v6.i1.54
  • Irvine K. et al. Greater cognitive deterioration in women than men with Alzheimer's disease: a meta analysis // Journal of clinical and experimental neuropsychology. 2012. V. 34. №9. P. 989-998.
  • DOI: 10.1080/13803395.2012.712676
  • Zhao L. et al. Sex differences in metabolic aging of the brain: insights into female susceptibility to Alzheimer's disease // Neurobiology of aging. 2016. V. 42. P. 69-79.
  • DOI: 10.1016/j.neurobiolaging.2016.02.011
  • Richetin K. et al. Differential alteration of hippocampal function and plasticity in females and males of the APPxPS1 mouse model of Alzheimer's disease // Neurobiology of aging. 2017. V. 57. P. 220-231.
  • DOI: 10.1016/j.neurobiolaging.2017.05.025
  • Jiao S. S. et al. Sex dimorphism profile of Alzheimer's disease-type pathologies in an APP/PS1 mouse model // Neurotoxicity research. 2016. V. 29. №2. P. 256-266.
  • DOI: 10.1007/s12640-015-9589-x
  • Pike C. J. Sex and the development of Alzheimer's disease // Journal of neuroscience research. 2017. V. 95. №1-2. P. 671-680.
  • DOI: 10.1002/jnr.23827
  • Altmann A. et al. Sex modifies the APOE-related risk of developing Alzheimer disease // Annals of neurology. 2014. V. 75. №4. P. 563-573.
  • DOI: 10.1002/ana.24135
  • Hohman T. J. et al. Sex-specific association of apolipoprotein E with cerebrospinal fluid levels of tau // JAMA neurology. 2018. V. 75. №8. P. 989-998.
  • DOI: 10.1001/jamaneurol.2018.0821
  • Neu S. C. et al. Apolipoprotein E genotype and sex risk factors for Alzheimer disease: a meta-analysis // JAMA neurology. 2017. V. 74. №10. P. 1178-1189.
  • DOI: 10.1001/jamaneurol.2017.2188
  • Fukumoto N. et al. Sexually dimorphic effect of the Val66Met polymorphism of BDNF on susceptibility to Alzheimer's disease: New data and meta-analysis // American Journal of Medical Genetics Part B: Neuropsychiatric Genetics. 2010. V. 153. №1. P. 235-242.
  • DOI: 10.1002/ajmg.b.30986
  • Kane R. L. et al. Interventions to prevent age-related cognitive decline, mild cognitive impairment, and clinical Alzheimer's-type dementia. 2017. https://www.ncbi.nlm.nih.gov/books/NBK442425
  • Rapp S. R. et al. Baseline experience with modified mini mental state exam: the Women's Health Initiative Memory Study (WHIMS) // Aging & mental health. 2003. V. 7. №3. P. 217-223.
  • DOI: 10.1080/1360786031000101201
  • Harman S. M. Estrogen replacement in menopausal women: recent and current prospective studies, the WHI and the KEEPS // Gender medicine. 2006. V. 3. №4. P. 254-269.
  • DOI: 10.1016/S1550-8579(06)80214-7
  • Hogervorst E. Effects of gonadal hormones on cognitive behaviour in elderly men and women // Journal of neuroendocrinology. 2013. V. 25. №11. P. 1182-1195.
  • DOI: 10.1111/jne.12080
  • Sherwin B. B. Estrogen and memory in women: how can we reconcile the findings? // Hormones and behavior. 2005. V. 47. №3. P. 371-375.
  • DOI: 10.1016/j.yhbeh.2004.12.002
  • Maki P. M. The critical window hypothesis of hormone therapy and cognition: a scientific update on clinical studies // Menopause (New York, NY). 2013. V. 20. №6. P. 695.
  • DOI: 10.1097/GME.0b013e3182960cf8
  • Gleason C. E. et al. Effects of hormone therapy on cognition and mood in recently postmenopausal women: findings from the randomized, controlled KEEPS-cognitive and affective study // PLoS medicine. 2015. V. 12. №6.
  • DOI: 10.1371/journal.pmed.1001833
  • Hodis H. N. et al. Methods and baseline cardiovascular data from the Early versus Late Intervention Trial with Estradiol testing the menopausal hormone timing hypothesis // Menopause (New York, NY). 2015. V. 22. №4. P. 391.
  • DOI: 10.1097/GME.0000000000000343
  • Henderson V. W. et al. Cognitive effects of estradiol after menopause: a randomized trial of the timing hypothesis // Neurology. 2016. V. 87. №7. P. 699-708.
  • DOI: 10.1212/WNL.0000000000002980
  • Berent-Spillson A. et al. Distinct cognitive effects of estrogen and progesterone in menopausal women // Psychoneuroendocrinology. 2015. V. 59. P. 25-36.
  • DOI: 10.1016/j.psyneuen.2015.04.020
  • Maki P. M. Minireview: effects of different HT formulations on cognition // Endocrinology. 2012. V. 153. №8. P. 3564-3570.
  • DOI: 10.1210/en.2012-1175
  • Resnick S. M. et al. Testosterone treatment and cognitive function in older men with low testosterone and age-associated memory impairment // Jama. 2017. V. 317. №7. P. 717-727.
  • DOI: 10.1001/jama.2016.21044
  • Beral V. et al. Ovarian cancer and hormone replacement therapy in the Million Women Study // The Lancet. 2007. V. 369. №9574. P. 1703-1710.
  • DOI: 10.1016/S0140-6736(07)60534-0
  • Shang Y., Brown M. Molecular determinants for the tissue specificity of SERMs // Science. 2002. V. 295. №5564. P. 2465-2468.
  • DOI: 10.1126/science.1068537
  • Yang Z. D., Yu J., Zhang Q. Effects of raloxifene on cognition, mental health, sleep and sexual function in menopausal women: a systematic review of randomized controlled trials // Maturitas. 2013. V. 75. №4. P. 341-348.
  • DOI: 10.1016/j.maturitas.2013.05.010
  • Snyder H. M. et al. Sex biology contributions to vulnerability to Alzheimer's disease: A think tank convened by the Women's Alzheimer's Research Initiative // Alzheimer's & Dementia. 2016. V. 12. №11. P. 1186-1196.
  • DOI: 10.1016/j.jalz.2016.08.004
Еще
Статья обзорная