Role of sex hormones in neuroinflammation in Alzheimer's disease
Kasumi Maekawa
Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
Department of Neuroscience and Pathobiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
Search for more papers by this authorCorresponding Author
Koji Yamanaka
Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
Department of Neuroscience and Pathobiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
Institute for Glyco-core Research (iGCORE), Nagoya University, Nagoya, Japan
Center for One Medicine Innovative Translational Research (COMIT), Gifu University Institute for Advanced Study, Gifu, Japan
Correspondence
Koji Yamanaka, Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Aichi 464-8601, Japan.
Email: [email protected]
Search for more papers by this authorKasumi Maekawa
Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
Department of Neuroscience and Pathobiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
Search for more papers by this authorCorresponding Author
Koji Yamanaka
Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
Department of Neuroscience and Pathobiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
Institute for Glyco-core Research (iGCORE), Nagoya University, Nagoya, Japan
Center for One Medicine Innovative Translational Research (COMIT), Gifu University Institute for Advanced Study, Gifu, Japan
Correspondence
Koji Yamanaka, Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Aichi 464-8601, Japan.
Email: [email protected]
Search for more papers by this authorAbstract
Neuroinflammation, which is mediated by microglia, astrocytes, and infiltrated immune cells and leads to the subsequent production of proinflammatory molecules, is associated with the pathomechanism of Alzheimer's disease (AD). As the incidence of AD is higher in females than males, multiple studies have focused on the relationship between sex hormones and AD pathology. Androgen and estrogen receptors are expressed throughout the brain, including the hippocampus; thus, both sex hormones may regulate brain function, including cognitive function. Endogenous sex hormone levels are depleted by aging and cancer therapies, including prostate cancer and breast cancer therapies. Previous cohort studies have revealed that these conditions may also increase the risk of developing AD. Here we review previous findings from epidemiologic and preclinical studies on AD and provide an overview of the roles of sex hormones as risk factors of AD and regulators of AD pathology, including neuroinflammation. Furthermore, we discuss the therapeutic potential of sex hormone supplementation as a preventive or therapeutic treatment for AD based on the results of randomized control trials.
CONFLICT OF INTEREST
The authors declare no conflicts of interest related to this article.
REFERENCES
- 1Bellenguez C, Kucukali F, Jansen IE, Kleineidam L, Moreno-Grau S, Amin N, et al. New insights into the genetic etiology of Alzheimer's disease and related dementias. Nat Genet. 2022; 54(4): 412–36.
- 2Heneka MT, Carson MJ, El Khoury J, Landreth GE, Brosseron F, Feinstein DL, et al. Neuroinflammation in Alzheimer's disease. Lancet Neurol. 2015; 14(4): 388–405.
- 3De Strooper B, Karran E. The cellular phase of Alzheimer's disease. Cell. 2016; 164(4): 603–15.
- 4Keren-Shaul H, Spinrad A, Weiner A, Matcovitch-Natan O, Dvir-Szternfeld R, Ulland TK, et al. A unique microglia type associated with restricting development of Alzheimer's disease. Cell. 2017; 169(7): 1276–90. e17.
- 5Jonsson T, Stefansson H, Steinberg S, Jonsdottir I, Jonsson PV, Snaedal J, et al. Variant of TREM2 associated with the risk of Alzheimer's disease. N Engl J Med. 2013; 368(2): 107–16.
- 6Guerreiro R, Wojtas A, Bras J, Carrasquillo M, Rogaeva E, Majounie E, et al. TREM2 variants in Alzheimer's disease. N Engl J Med. 2013; 368(2): 117–27.
- 7Saito T, Matsuba Y, Mihira N, Takano J, Nilsson P, Itohara S, et al. Single App knock-in mouse models of Alzheimer's disease. Nat Neurosci. 2014; 17(5): 661–3.
- 8Santacruz K, Lewis J, Spires T, Paulson J, Kotilinek L, Ingelsson M, et al. Tau suppression in a neurodegenerative mouse model improves memory function. Science. 2005; 309(5733): 476–81.
- 9Sobue A, Komine O, Hara Y, Endo F, Mizoguchi H, Watanabe S, et al. Microglial gene signature reveals loss of homeostatic microglia associated with neurodegeneration of Alzheimer's disease. Acta Neuropathol Commun. 2021; 9(1): 1.
- 10Liddelow SA, Guttenplan KA, Clarke LE, Bennett FC, Bohlen CJ, Schirmer L, et al. Neurotoxic reactive astrocytes are induced by activated microglia. Nature. 2017; 541(7638): 481–7.
- 11Endo F, Kasai A, Soto JS, Yu X, Qu Z, Hashimoto H, et al. Molecular basis of astrocyte diversity and morphology across the CNS in health and disease. Science. 2022; 378(6619): eadc9020.
- 12Seshadri S, Wolf PA, Beiser A, Au R, McNulty K, White R, et al. Lifetime risk of dementia and Alzheimer's disease. The impact of mortality on risk estimates in the Framingham Study. Neurology. 1997; 49(6): 1498–504.
- 13Niu H, Alvarez-Alvarez I, Guillen-Grima F, Aguinaga-Ontoso I. Prevalence and incidence of Alzheimer's disease in Europe: a meta-analysis. Neurologia. 2017; 32(8): 523–32.
- 14Beam CR, Kaneshiro C, Jang JY, Reynolds CA, Pedersen NL, Gatz M. Differences between women and men in incidence rates of dementia and Alzheimer's disease. J Alzheimers Dis. 2018; 64(4): 1077–83.
- 15Yoshida D, Ohara T, Hata J, Shibata M, Hirakawa Y, Honda T, et al. Lifetime cumulative incidence of dementia in a community-dwelling elderly population in Japan. Neurology. 2020; 95(5): e508–e18.
- 16Dong X, Jiang H, Li S, Zhang D. Low serum testosterone concentrations are associated with poor cognitive performance in older men but not women. Front Aging Neurosci. 2021; 13:712237.
- 17Giannos P, Prokopidis K, Church DD, Kirk B, Morgan PT, Lochlainn MN, et al. Associations of bioavailable serum testosterone with cognitive function in older men: results from the National Health and Nutrition Examination Survey. J Gerontol A Biol Sci Med Sci. 2023; 78(1): 151–7.
- 18Rosario ER, Chang L, Head EH, Stanczyk FZ, Pike CJ. Brain levels of sex steroid hormones in men and women during normal aging and in Alzheimer's disease. Neurobiol Aging. 2011; 32(4): 604–13.
- 19Rosario ER, Chang L, Stanczyk FZ, Pike CJ. Age-related testosterone depletion and the development of Alzheimer disease. JAMA. 2004; 292(12): 1431–2.
- 20Wolf OT, Kirschbaum C. Endogenous estradiol and testosterone levels are associated with cognitive performance in older women and men. Horm Behav. 2002; 41(3): 259–66.
- 21Ford AH, Yeap BB, Flicker L, Hankey GJ, Chubb SAP, Golledge J, et al. Sex hormones and incident dementia in older men: the health in men study. Psychoneuroendocrinology. 2018; 98: 139–47.
- 22Marriott RJ, Murray K, Flicker L, Hankey GJ, Matsumoto AM, Dwivedi G, et al. Lower serum testosterone concentrations are associated with a higher incidence of dementia in men: The UK Biobank prospective cohort study. Alzheimers Dement. 2022; 18(10): 1907–18.
- 23Liu JM, Shen CY, Lau WCY, Shao SC, Man KKC, Hsu RJ, et al. Association between androgen deprivation therapy and risk of dementia in men with prostate cancer. Cancers (Basel). 2021; 13(15): 3861.
- 24Kim JW, Kim DK, Lee HS, Park JY, Ahn HK, Ha JS, et al. Androgen deprivation therapy in patients with prostate cancer is associated with the risk of subsequent Alzheimer's disease but not with vascular dementia. World J Mens Health. 2022; 40(3): 481–9.
- 25Cherrier MM, Aubin S, Higano CS. Cognitive and mood changes in men undergoing intermittent combined androgen blockade for non-metastatic prostate cancer. Psychooncology. 2009; 18(3): 237–47.
- 26Alibhai SM, Breunis H, Timilshina N, Marzouk S, Stewart D, Tannock I, et al. Impact of androgen-deprivation therapy on cognitive function in men with nonmetastatic prostate cancer. J Clin Oncol. 2010; 28(34): 5030–7.
- 27Sari Motlagh R, Quhal F, Mori K, Miura N, Aydh A, Laukhtina E, et al. The risk of new onset dementia and/or Alzheimer disease among patients with prostate cancer treated with androgen deprivation therapy: a systematic review and meta-analysis. J Urol. 2021; 205(1): 60–7.
- 28Alibhai SM, Timilshina N, Duff-Canning S, Breunis H, Tannock IF, Naglie G, et al. Effects of long-term androgen deprivation therapy on cognitive function over 36 mo in men with prostate cancer. Cancer. 2017; 123(2): 237–44.
- 29Baik SH, Kury FSP, McDonald CJ. Risk of Alzheimer's disease among senior medicare beneficiaries treated with androgen deprivation therapy for prostate cancer. J Clin Oncol. 2017; 35(30): 3401–9.
- 30Simerly RB, Chang C, Muramatsu M, Swanson LW. Distribution of androgen and estrogen receptor mRNA-containing cells in the rat brain: an in situ hybridization study. J Comp Neurol. 1990; 294(1): 76–95.
- 31Sarkey S, Azcoitia I, Garcia-Segura LM, Garcia-Ovejero D, DonCarlos LL. Classical androgen receptors in non-classical sites in the brain. Horm Behav. 2008; 53(5): 753–64.
- 32Zhou L, Blaustein JD, De Vries GJ. Distribution of androgen receptor immunoreactivity in vasopressin- and oxytocin-immunoreactive neurons in the male rat brain. Endocrinology. 1994; 134(6): 2622–7.
- 33Rosario ER, Carroll JC, Oddo S, LaFerla FM, Pike CJ. Androgens regulate the development of neuropathology in a triple transgenic mouse model of Alzheimer's disease. J Neurosci. 2006; 26(51): 13384–9.
- 34Pan W, Han S, Kang L, Li S, Du J, Cui H. Effects of dihydrotestosterone on synaptic plasticity of the hippocampus in mild cognitive impairment male SAMP8 mice. Exp Ther Med. 2016; 12(3): 1455–63.
- 35Yan XS, Yang ZJ, Jia JX, Song W, Fang X, Cai ZP, et al. Protective mechanism of testosterone on cognitive impairment in a rat model of Alzheimer's disease. Neural Regen Res. 2019; 14(4): 649–57.
- 36Gouras GK, Xu H, Gross RS, Greenfield JP, Hai B, Wang R, et al. Testosterone reduces neuronal secretion of Alzheimer's beta-amyloid peptides. Proc Natl Acad Sci U S A. 2000; 97(3): 1202–5.
- 37McAllister C, Long J, Bowers A, Walker A, Cao P, Honda S, et al. Genetic targeting aromatase in male amyloid precursor protein transgenic mice down-regulates beta-secretase (BACE1) and prevents Alzheimer-like pathology and cognitive impairment. J Neurosci. 2010; 30(21): 7326–34.
- 38Atallah A, Mhaouty-Kodja S, Grange-Messent V. Chronic depletion of gonadal testosterone leads to blood-brain barrier dysfunction and inflammation in male mice. J Cereb Blood Flow Metab. 2017; 37(9): 3161–75.
- 39Yao PL, Zhuo S, Mei H, Chen XF, Li N, Zhu TF, et al. Androgen alleviates neurotoxicity of beta-amyloid peptide (Abeta) by promoting microglial clearance of Abeta and inhibiting microglial inflammatory response to Abeta. CNS Neurosci Ther. 2017; 23(11): 855–65.
- 40Sierra A, Gottfried-Blackmore A, Milner TA, McEwen BS, Bulloch K. Steroid hormone receptor expression and function in microglia. Glia. 2008; 56(6): 659–74.
- 41Garcia-Ovejero D, Veiga S, Garcia-Segura LM, Doncarlos LL. Glial expression of estrogen and androgen receptors after rat brain injury. J Comp Neurol. 2002; 450(3): 256–71.
- 42Puy L, MacLusky NJ, Becker L, Karsan N, Trachtenberg J, Brown TJ. Immunocytochemical detection of androgen receptor in human temporal cortex characterization and application of polyclonal androgen receptor antibodies in frozen and paraffin-embedded tissues. J Steroid Biochem Mol Biol. 1995; 55(2): 197–209.
- 43Peng R, Dai W, Li D, Li Y. Gonadal hormone trigger the dynamic microglial alterations through Traf6/TAK1 axis that correlate with depressive behaviors. J Psychiatr Res. 2022; 152: 128–38.
- 44Yang L, Zhou R, Tong Y, Chen P, Shen Y, Miao S, et al. Neuroprotection by dihydrotestosterone in LPS-induced neuroinflammation. Neurobiol Dis. 2020; 140:104814.
- 45Barreto G, Veiga S, Azcoitia I, Garcia-Segura LM, Garcia-Ovejero D. Testosterone decreases reactive astroglia and reactive microglia after brain injury in male rats: role of its metabolites, oestradiol and dihydrotestosterone. Eur J Neurosci. 2007; 25(10): 3039–46.
- 46Melcangi RC, Riva MA, Fumagalli F, Magnaghi V, Racagni G, Martini L. Effect of progesterone, testosterone and their 5 alpha-reduced metabolites on GFAP gene expression in type 1 astrocytes. Brain Res. 1996; 711(1–2): 10–5.
- 47Gu F, Hata R, Toku K, Yang L, Ma YJ, Maeda N, et al. Testosterone up-regulates aquaporin-4 expression in cultured astrocytes. J Neurosci Res. 2003; 72(6): 709–15.
- 48Unger MS, Li E, Scharnagl L, Poupardin R, Altendorfer B, Mrowetz H, et al. CD8(+) T-cells infiltrate Alzheimer's disease brains and regulate neuronal- and synapse-related gene expression in APP-PS1 transgenic mice. Brain Behav Immun. 2020; 89: 67–86.
- 49Zheng B, Sun J, Luo H, Yang L, Li Q, Zhang L, et al. Testosterone protects mice against zika virus infection and suppresses the inflammatory response in the brain. iScience. 2022; 25(11):105300.
- 50Grandys M, Majerczak J, Zapart-Bukowska J, Duda K, Kulpa JK, Zoladz JA. Lowered serum testosterone concentration is associated with enhanced inflammation and worsened lipid profile in men. Front Endocrinol (Lausanne). 2021; 12:735638.
- 51Hussain R, Ghoumari AM, Bielecki B, Steibel J, Boehm N, Liere P, et al. The neural androgen receptor: a therapeutic target for myelin repair in chronic demyelination. Brain. 2013; 136(Pt 1): 132–46.
- 52Lara A, Esperante I, Meyer M, Liere P, Di Giorgio N, Schumacher M, et al. Neuroprotective effects of testosterone in male wobbler mouse, a model of amyotrophic lateral sclerosis. Mol Neurobiol. 2021; 58(5): 2088–106.
- 53Alves JV, da Costa RM, Pereira CA, Fedoce AG, Silva CAA, Carneiro FS, et al. Supraphysiological levels of testosterone induce vascular dysfunction via activation of the NLRP3 inflammasome. Front Immunol. 2020; 11: 1647.
- 54Mosconi L, Berti V, Quinn C, McHugh P, Petrongolo G, Varsavsky I, et al. Sex differences in Alzheimer risk: brain imaging of endocrine vs chronologic aging. Neurology. 2017; 89(13): 1382–90.
- 55Mosconi L, Berti V, Quinn C, McHugh P, Petrongolo G, Osorio RS, et al. Perimenopause and emergence of an Alzheimer's bioenergetic phenotype in brain and periphery. PLoS One. 2017; 12(10):e0185926.
- 56Gilsanz P, Lee C, Corrada MM, Kawas CH, Quesenberry CP Jr, Whitmer RA. Reproductive period and risk of dementia in a diverse cohort of health care members. Neurology. 2019; 92(17): e2005–e14.
- 57Fu C, Hao W, Shrestha N, Virani SS, Mishra SR, Zhu D. Association of reproductive factors with dementia: a systematic review and dose-response meta-analyses of observational studies. EClinicalMedicine. 2022; 43:101236.
- 58Sawada S, Sato K, Kusuhara M, Ayaori M, Yonemura A, Tamaki K, et al. Effect of anastrozole and tamoxifen on lipid metabolism in Japanese postmenopausal women with early breast cancer. Acta Oncol. 2005; 44(2): 134–41.
- 59Lum SS, Woltering EA, Fletcher WS, Pommier RF. Changes in serum estrogen levels in women during tamoxifen therapy. Am J Surg. 1997; 173(5): 399–402.
- 60Bender CM, Merriman JD, Gentry AL, Ahrendt GM, Berga SL, Brufsky AM, et al. Patterns of change in cognitive function with anastrozole therapy. Cancer. 2015; 121(15): 2627–36.
- 61Branigan GL, Soto M, Neumayer L, Rodgers K, Brinton RD. Association between hormone-modulating breast cancer therapies and incidence of neurodegenerative outcomes for women with breast cancer. JAMA Netw Open. 2020; 3(3):e201541.
- 62Thompson MR, Niu J, Lei X, Nowakowska M, Wehner MR, Giordano SH, et al. Association of endocrine therapy and dementia in women with breast cancer. Breast Cancer (Dove Med Press). 2021; 13: 219–24.
- 63Blanchette PS, Lam M, Le B, Richard L, Shariff SZ, Pritchard KI, et al. The association between endocrine therapy use and dementia among post-menopausal women treated for early-stage breast cancer in Ontario, Canada. J Geriatr Oncol. 2020; 11(7): 1132–7.
- 64Liao KF, Lin CL, Lai SW. Nationwide case-control study examining the association between tamoxifen use and Alzheimer's disease in aged women with breast cancer in Taiwan. Front Pharmacol. 2017; 8: 612.
- 65Ording AG, Jensen AB, Cronin-Fenton D, Pedersen L, Sorensen HT, Lash TL. Null association between tamoxifen use and dementia in Danish breast cancer patients. Cancer Epidemiol Biomarkers Prev. 2013; 22(5): 993–6.
- 66Bromley SE, Matthews A, Smeeth L, Stanway S, Bhaskaran K. Risk of dementia among postmenopausal breast cancer survivors treated with aromatase inhibitors versus tamoxifen: a cohort study using primary care data from the UK. J Cancer Surviv. 2019; 13(4): 632–40.
- 67Manly JJ, Merchant CA, Jacobs DM, Small SA, Bell K, Ferin M, et al. Endogenous estrogen levels and Alzheimer's disease among postmenopausal women. Neurology. 2000; 54(4): 833–7.
- 68Yue X, Lu M, Lancaster T, Cao P, Honda S, Staufenbiel M, et al. Brain estrogen deficiency accelerates Abeta plaque formation in an Alzheimer's disease animal model. Proc Natl Acad Sci U S A. 2005; 102(52): 19198–203.
- 69Montague D, Weickert CS, Tomaskovic-Crook E, Rothmond DA, Kleinman JE, Rubinow DR. Oestrogen receptor alpha localisation in the prefrontal cortex of three mammalian species. J Neuroendocrinol. 2008; 20(7): 893–903.
- 70Shughrue PJ, Lane MV, Merchenthaler I. Comparative distribution of estrogen receptor-alpha and -beta mRNA in the rat central nervous system. J Comp Neurol. 1997; 388(4): 507–25.
10.1002/(SICI)1096-9861(19971201)388:4<507::AID-CNE1>3.0.CO;2-6 CAS PubMed Web of Science® Google Scholar
- 71Almey A, Milner TA, Brake WG. Estrogen receptors in the central nervous system and their implication for dopamine-dependent cognition in females. Horm Behav. 2015; 74: 125–38.
- 72Yun J, Yeo IJ, Hwang CJ, Choi DY, Im HS, Kim JY, et al. Estrogen deficiency exacerbates Abeta-induced memory impairment through enhancement of neuroinflammation, amyloidogenesis and NF-kB activation in ovariectomized mice. Brain Behav Immun. 2018; 73: 282–93.
- 73Luo M, Zeng Q, Jiang K, Zhao Y, Long Z, Du Y, et al. Estrogen deficiency exacerbates learning and memory deficits associated with glucose metabolism disorder in APP/PS1 double transgenic female mice. Genes Dis. 2022; 9(5): 1315–31.
- 74Li R, He P, Cui J, Staufenbiel M, Harada N, Shen Y. Brain endogenous estrogen levels determine responses to estrogen replacement therapy via regulation of BACE1 and NEP in female Alzheimer's transgenic mice. Mol Neurobiol. 2013; 47(3): 857–67.
- 75Liang K, Yang L, Yin C, Xiao Z, Zhang J, Liu Y, et al. Estrogen stimulates degradation of beta-amyloid peptide by up-regulating neprilysin. J Biol Chem. 2010; 285(2): 935–42.
- 76Vegeto E, Belcredito S, Ghisletti S, Meda C, Etteri S, Maggi A. The endogenous estrogen status regulates microglia reactivity in animal models of neuroinflammation. Endocrinology. 2006; 147(5): 2263–72.
- 77Brown CM, Mulcahey TA, Filipek NC, Wise PM. Production of proinflammatory cytokines and chemokines during neuroinflammation: novel roles for estrogen receptors alpha and beta. Endocrinology. 2010; 151(10): 4916–25.
- 78Dimayuga FO, Reed JL, Carnero GA, Wang C, Dimayuga ER, Dimayuga VM, et al. Estrogen and brain inflammation: effects on microglial expression of MHC, costimulatory molecules and cytokines. J Neuroimmunol. 2005; 161(1–2): 123–36.
- 79Vegeto E, Bonincontro C, Pollio G, Sala A, Viappiani S, Nardi F, et al. Estrogen prevents the lipopolysaccharide-induced inflammatory response in microglia. J Neurosci. 2001; 21(6): 1809–18.
- 80Bruce-Keller AJ, Keeling JL, Keller JN, Huang FF, Camondola S, Mattson MP. Antiinflammatory effects of estrogen on microglial activation. Endocrinology. 2000; 141(10): 3646–56.
- 81Acosta-Martinez M. Shaping microglial phenotypes through estrogen receptors: relevance to sex-specific neuroinflammatory responses to brain injury and disease. J Pharmacol Exp Ther. 2020; 375(1): 223–36.
- 82Ghisletti S, Meda C, Maggi A, Vegeto E. 17beta-estradiol inhibits inflammatory gene expression by controlling NF-kappaB intracellular localization. Mol Cell Biol. 2005; 25(8): 2957–68.
- 83Lu YP, Zeng M, Hu XY, Xu H, Swaab DF, Ravid R, et al. Estrogen receptor alpha-immunoreactive astrocytes are increased in the hippocampus in Alzheimer's disease. Exp Neurol. 2003; 183(2): 482–8.
- 84Spence RD, Hamby ME, Umeda E, Itoh N, Du S, Wisdom AJ, et al. Neuroprotection mediated through estrogen receptor-alpha in astrocytes. Proc Natl Acad Sci U S A. 2011; 108(21): 8867–72.
- 85Acaz-Fonseca E, Avila-Rodriguez M, Garcia-Segura LM, Barreto GE. Regulation of astroglia by gonadal steroid hormones under physiological and pathological conditions. Prog Neurobiol. 2016; 144: 5–26.
- 86Liang Z, Valla J, Sefidvash-Hockley S, Rogers J, Li R. Effects of estrogen treatment on glutamate uptake in cultured human astrocytes derived from cortex of Alzheimer's disease patients. J Neurochem. 2002; 80(5): 807–14.
- 87Cherrier MM, Matsumoto AM, Amory JK, Asthana S, Bremner W, Peskind ER, et al. Testosterone improves spatial memory in men with Alzheimer disease and mild cognitive impairment. Neurology. 2005; 64(12): 2063–8.
- 88Cherrier MM, Anderson K, Shofer J, Millard S, Matsumoto AM. Testosterone treatment of men with mild cognitive impairment and low testosterone levels. Am J Alzheimers Dis Other Demen. 2015; 30(4): 421–30.
- 89Lu PH, Masterman DA, Mulnard R, Cotman C, Miller B, Yaffe K, et al. Effects of testosterone on cognition and mood in male patients with mild Alzheimer disease and healthy elderly men. Arch Neurol. 2006; 63(2): 177–85.
- 90Asih PR, Wahjoepramono EJ, Aniwiyanti V, Wijaya LK, de Ruyck K, Taddei K, et al. Testosterone replacement therapy in older male subjective memory complainers: double-blind randomized crossover placebo-controlled clinical trial of physiological assessment and safety. CNS Neurol Disord Drug Targets. 2015; 14(5): 576–86.
- 91Wahjoepramono EJ, Asih PR, Aniwiyanti V, Taddei K, Dhaliwal SS, Fuller SJ, et al. The effects of testosterone supplementation on cognitive functioning in older men. CNS Neurol Disord Drug Targets. 2016; 15(3): 337–43.
- 92Huang G, Wharton W, Bhasin S, Harman SM, Pencina KM, Tsitouras P, et al. Effects of long-term testosterone administration on cognition in older men with low or low-to-normal testosterone concentrations: a prespecified secondary analysis of data from the randomised, double-blind, placebo-controlled TEAAM trial. Lancet Diabetes Endocrinol. 2016; 4(8): 657–65.
- 93Tan S, Sohrabi HR, Weinborn M, Tegg M, Bucks RS, Taddei K, et al. Effects of testosterone supplementation on separate cognitive domains in cognitively healthy older men: a meta-analysis of current randomized clinical trials. Am J Geriatr Psychiatry. 2019; 27(11): 1232–46.
- 94Kantarci K, Lowe VJ, Lesnick TG, Tosakulwong N, Bailey KR, Fields JA, et al. Early postmenopausal transdermal 17beta-estradiol therapy and amyloid-beta deposition. J Alzheimers Dis. 2016; 53(2): 547–56.
- 95Schelbaum E, Loughlin L, Jett S, Zhang C, Jang G, Malviya N, et al. Association of reproductive history with brain MRI biomarkers of dementia risk in midlife. Neurology. 2021; 97(23): e2328–e39.
- 96Wang Y, Hernandez G, Mack WJ, Schneider LS, Yin F, Brinton RD. Retrospective analysis of phytoSERM for management of menopause-associated vasomotor symptoms and cognitive decline: a pilot study on pharmacogenomic effects of mitochondrial haplogroup and APOE genotype on therapeutic efficacy. Menopause. 2020; 27(1): 57–65.
- 97Kling JM, Dowling NM, Bimonte-Nelson H, Gleason CE, Kantarci K, Stonnington CM, et al. Associations between pituitary-ovarian hormones and cognition in recently menopausal women independent of type of hormone therapy. Maturitas. 2023; 167: 113–22.
- 98Reis de Assis D, Szabo A, Requena Osete J, Puppo F, O'Connell KS, Akkouh IA, et al. Using iPSC models to understand the role of estrogen in neuron-glia interactions in schizophrenia and bipolar disorder. Cell. 2021; 10(2): 209.
- 99Supakul S, Okano H, Maeda S. Utilization of human induced pluripotent stem cells-derived in vitro models for the future study of sex differences in Alzheimer's disease. Front Aging Neurosci. 2021; 13:768948.
- 100Shum C, Macedo SC, Warre-Cornish K, Cocks G, Price J, Srivastava DP. Utilizing induced pluripotent stem cells (iPSCs) to understand the actions of estrogens in human neurons. Horm Behav. 2015; 74: 228–42.
- 101Adhya D, Annuario E, Lancaster MA, Price J, Baron-Cohen S, Srivastava DP. Understanding the role of steroids in typical and atypical brain development: advantages of using a “brain in a dish” approach. J Neuroendocrinol. 2018; 30(2):e12547.