World J Mens Health. 2022 Apr;40(2):243-256. English.
Published online Jan 02, 2022.
Copyright © 2022 Korean Society for Sexual Medicine and Andrology
Review

Impact of Testosterone on Alzheimer’s Disease

Vittorio Emanuele Bianchi
    • Department of Endocrinology and Metabolism, Clinical Research Center Stella Maris, Falciano, San Marino, Italy.
Received August 31, 2021; Revised September 16, 2021; Accepted September 23, 2021.

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

Alzheimer’s disease (AD) is a neurodegenerative disease responsible for almost half of all dementia cases in the world and progressively increasing. The etiopathology includes heritability, genetic factors, aging, nutrition, but sex hormones play a relevant role. Animal models demonstrated that testosterone (T) exerted a neuroprotective effect reducing the production of amyloid-beta (Aβ), improving synaptic signaling, and counteracting neuronal death. This study aims to evaluate the impact of T deprivation and T administration in humans on the onset of dementia and AD. A search was conducted on MEDLINE and Scopus for the “androgen deprivation therapy” and “testosterone therapy” with “dementia” and “Alzheimer’s.” Studies lasting twenty years with low risk of bias, randomized clinical trial, and case-controlled studies were considered. Twelve articles on the effect of androgen deprivation therapy (ADT) and AD and seventeen on T therapy and AD were retrieved. Men with prostate cancer under ADT showed a higher incidence of dementia and AD. The effect of T administration in hypogonadal men with AD and cognitive impairment has evidenced some positive results. The majority of studies showed the T administration improved memory and cognition in AD while others did not find any benefit. Although some biases in the studies are evident, T therapy for AD patients may represent an essential clinical therapy to reduce dementia incidence and AD progression. However, more specific case-controlled trials on the effect of androgens therapy in men and women to reducing the onset of AD are necessary.

Keywords
Alzheimer disease; Amyloid beta-peptides; Dementia; Estradiol; Neuroprotection; Testosterone

INTRODUCTION

Alzheimer’s disease (AD) is a devastating neurodegenerative disease responsible for almost half of dementia cases [1] and progressively increasing with upper of 50 million people affected. Etiopathology of AD is multifactorial, including genetic factors and heritability, nutritional disorders, mitochondrial dysfunction, oxidative stress, and aging [2]. AD is characterized by an abnormal Aβ deposition in neuron and extracellular plaque formation responsible for the pathologic events, causing neuronal degeneration [3] and synapsis dysfunction [4]. Aβ deposition and the regulation of amyloid-beta (Aβ) protein precursor is regulated mostly by testosterone (T) pathways and are described in another review [5].

Sex hormones play a relevant role in developing AD, as evidenced by the greater incidence in women than men [6]. In cellular [7] and animal models of AD [8, 9], it was demonstrated that T level was closely associated with the neuronal efficiency and reduced the Aβ deposition in the brain. By activating AR signaling pathway, T stimulates the microglia phagocytosis, removing the Aβ deposition and inhibiting the inflammatory response [10]. In the rat model of AD, it was shown that T prevented cognitive decline scavenging free radicals, thereby enhancing synaptic plasticity [9, 11], and regulates neuronal bioenergetic increasing mitochondrial function [12], increasing antioxidant activity preventing neurodegenerative disorders. Furthermore, T reduces insulin-resistance in obesity, improving cognitive function [13]. Furthermore, T prevented vascular and neuronal aging by increasing eNOS activity and stimulating SIRT1 expression [14]. The behavioral performance and learning was associated with an increased SYN expression levels [15]. Dihydrotestosterone (DHT) appeared to be more effective treatment to reduce the onset of dementia [16].

In men, low serum T levels have been implicated in the pathogenesis of AD [17]. In contrast, a higher serum level of free T in both sexes seems to be protective against AD incidence and development [18]. Lee et al [18], in older subjects, evaluated by B-positron and magnetic resonance imaging, and found that a high free T level in females and males was correlated with lower cerebral Aβ deposition and lower cognitive impairment, while free estradiol was not related to Aβ or neurodegeneration in both sexes. This study evidenced that T is active in the early stage of the pathological accumulation of Aβ. Other studies showed that men’s low T serum levels were associated with an increased Aβ deposition, causing AD development [18, 19] and synaptic dysfunction with a consequent cognitive decline [4]. Considering the high impact of T on maintaining brain health, this study aims to evaluate the effect of androgens deprivation and treatment on the evolution of AD.

EFFECT OF TESTOSTERONE ON ALZHEIMER’S DISEASE

The wide distribution of androgen receptors (ARs) in the brain suggests that androgen may exert a relevant role in neuronal function. AR is mainly expressed in the hypothalamus and amygdala, areas deputies to learning and memory, in the telencephalon, amygdala, and spinal cord [20]. The neurotrophic effect of T consists of activating AR and preventing Aβ deposition on neurons directly and by the action of its metabolite 17β-estradiol [21]. T improves energy metabolism and reduces oxidative stress in neurons [22] and down-regulates the beta-secretase (BACE1) enzyme activity, enzyme that reduces the Aβ deposition, suggesting that endogenous T, independently from estrogen, may protect against AD in males [23]. The effect of T on cellular bioenergetics is more efficient than other sex hormones, such as progesterone and estrogens [24]. The action of T on neurons is complex and regulated by its direct action associated with the effects of the various metabolites originated by T molecule. T and its related neurosteroids (structurally diverse neurosteroids such as progesterone, estradiol, estrone, T, 3alpha-androstanediol [3α-Diol], DHEA, and allopregnanolone) are involved in the regulation of neuron activity [25].

T can be aromatized in 17β-estradiol, in DHT after the 5α-reductase effect, and androstenedione after the partial reduction by 3α-HSOR converted to 3α-Diol, which has estrogenic effect, which activates the GABA receptors. 17β-estradiol activates estrogen receptors (ERs), potentiating some effects of T. Metabolites of T, such as DHT and Androstenediol show interesting differences in their relative biologic effects in the activation of the AR.

However, T has a direct neuroprotective effect, independently by its conversion into estradiol [23], potentiating the anti-Aβ effect and reducing the neuronal death of 80% to 90% [26]. The metabolite 3α-Diol is of relevant interest because it is potent GABA(A) receptor-modulating neurosteroids with anticonvulsant properties, and 3α-diol production, but not T, restores cognitive and affective performance [27]. Androstenediol is active on GABA and N-methyl-d-aspartate (NMDA) receptors responsible for memory, learning impairments, and psychosis. Notably, the 5α-androstane, 3β,17β-diol (3β-Diol) activate ER and not AR. The NMDA receptor RNA is also influenced by the GH and insulin-like growth factor-1 (IGF-1) levels that increase the expression and are more prominently affected by chronological age than by the sex hormones (Fig. 1) [28].

Fig. 1
Testosterone, the effect of α-reductase, is reduced to DHT, the strongest non-aromatizable androgen. DHT is then in Androstenediol from which metabolites 3α- and 3β-diol have a weak effect on AR while are more active on Erα and Erβ. 3α-diol activates GABA-receptors which regulate anxiety, depression and seizure. Testosterone is also aromatized in 17β-estradiol which, activating Erα and β, stimulates mitochondrial function, neurotransmission, and anti-inflammatory effect with consequent improved cognition. DHT and DHEAS activate the NMDA receptors which regulate memory, learning impairment, and psychosis. DHT: dihydrotestosterone, NMDA: N-methyl-d-aspartate, AR: androgen receptor.

T influences cognition, enhancing synaptic plasticity [11], increasing the number of intact cells and the dendritic spine density in the hippocampal region [8]. Castration reduced the hippocampal dendritic spine density, which is restored by androgen administration. In the presence of low serum T levels, many biochemical and metabolic functions in the brain are compromised (Fig. 1).

A meta-analysis showed that low plasma T level was significantly associated with increased risk of AD, and it should be considered a risk factor in worsening cognitive function in elderly men [17].

THE ROLE OF ESTRADIOL

The mouse model demonstrated that estradiol exerted an essential role in regulating endogenous neurogenesis, synaptic plasticity, and cognitive function in the early stage of AD [29] and protected the young APP/PS1 mice from cognitive decline [30]. In women, estrogens exert a protective role against neurodegeneration, and with the onset of menopause, the drop-in plasma level is considered a determinant factor in AD development. In menopausal women, plasma serum androgens and SHBG decline progressively with advancing age [31, 32]. Consequently, estradiol derives primarily from the aromatization of T in extra-gonadal tissue and is regulated by the aromatase expression in the tissues [33]. The medium plasma level of androgens in women significantly decreases with advancing age. The plasma level of total and free T in the range of age 65 to 74 compared to the range of 18 to 24 years varies from 1.8 to 0.66 nmoL/L, and 23.61 to 10.81 pmoL/L respectively. DHEAS and Androstenedione also decreases of one-third [31].

However, whether estrogens administration in menopausal women to prevent AD was effective remains questionable. Although some observational studies found a reduced incidence of AD and dementia between women taking estrogen therapy [34, 35, 36], others did not find any positive effect [37, 38]. A recent study on a large population of 84,739 postmenopausal women showed that the systematic administration of estrogens was associated with an overall increased incidence of AD [39]. Although possibly beneficial if taken during a critical window near menopause, estrogens therapy (especially opposed compounds) initiated in later life may be associated with increased risk in A.D. Placebo-controlled trials reported an increased risk of the incidence of dementia in women who received a conjugated equine estrogen independently from the association of medroxyprogesterone acetate [40]. Tolppanen et al [41] did not find any differences in systemic estrogen use among Finnish women with AD than those without AD. The relation of AD risk to timing and type of hormone replacement deserves further study [42].

The higher incidence of AD in women is related not only to estrogen activity but also to the plasma level of androgens and this may explain why women are at increased risk of cognitive decline and AD compared with men.

Estrogens exert a different, when not opposite, effect on the brain in males and females [43]. The estrogen action is not related only to the plasma levels but also to those synthesized in non-reproductive tissues, particularly in the brain, and has cell-specific estrogen synthesis and ER signaling [44]. Tibolone, a synthetic hormone with estrogenic, androgenic, and progestogenic activity, showed a neuroprotective effect [45]. Although few studies on the impact of tibolone on Central Nervous System, it improved memory and learning [46, 47, 48]. These studies suggest that the association of T with estrogen can probably be relevant in the treatment of AD, considering that T can be aromatized at the cellular level. Furthermore, the neuroprotective actions induced by estrogens are interlinked with the IGF-1 signaling pathway [49] that should be considered in the evolution of AD. In conclusion, the effects of 17β-estradiol on the brain per se is complex because is not only evaluable by its serum level, but the formation at cellular level seems more effective, and they would be evaluated concerning the plasma level of androgens and IGF-1.

METHODS

The search was conducted, finding out on MEDLINE and Scopus from the year 2000 until now, clinical studies with the following keywords: “androgen deprivation therapy” with “Alzheimer’s disease,” and “dementia” and “Alzheimer’s disease” and “testosterone therapy” with AD.

RESULTS

For androgen deprivation therapy (ADT) and dementia, twenty articles (Table 1) and seventeen for T therapy and AD (Table 2) were retrieved. Inclusion criteria were absence of history of cancer before the diagnosis of prostate cancer or those who received both orchiectomy and GnRH agonist.

Table 1
Effect of ADT on cognitive impairment and AD development

Table 2
Effect of testosterone therapy on AD and cognitive impairment

ANDROGEN DEPRIVATION THERAPY AND DEMENTIA

Twenty studies conducted on large cohorts of patients have investigated the effect of ADT on the risk of developing AD or dementia have been selected [50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69]. The studies are summarized in Table 1. The majority (13 studies) found a significant association between ADT with cognitive function and dementia [50, 51, 52, 53, 54, 56, 57, 60, 63, 64, 66, 68, 69], while others did not [55, 58, 59, 60, 61, 62, 65, 67]. The type of therapy plays a despaired effect on AD [51]. Longitudinal studies evidenced that in men with prostate cancer (PC) treated with antiandrogen therapy, the plasma T level decreased while the Aβ level increased [19, 70]. Systematic reviews demonstrated that men with PC under ADT have a higher risk of cognitive impairment and dementia [71] and worsening depression [72] confirmed by a recent meta-analysis [73].

The controversial results that emerged from the studies show the complexity of the investigation on ADT’s effect on brain efficiency and dementia. Baik et al [60] investigated a population of 1,238,879 patients, of which 35% underwent either chemical or surgical ADT in a follow-up of an average of 5.5 years, and they did not find any correlation between ADT with AD. However, the study lacked of essential information; there was no account for the use of antiandrogens, family history of AD, smoking habits, and PC staging information and biomarkers. Notably, the routine therapy used by the patients was not taken into account. Furthermore, a specific test to evaluate cognitive state was not performed. Chung et al [67] demonstrated in a large population that ADT was not correlated with increased incidence of AD or Parkinson’s disease. However, data were retrieved from an extensive insurance database (5,340 subjects) and patients were tracked for 5 years and received the diagnosis from their index database. No specific information about the patients was given, familiarity, correction for confounding factors such as body weight, diabetes. We individually tracked each patient (n=5,340) for a 5-year period (from the years 2001 to 2013) to discriminate those who subsequently received a diagnosis of AD, starting from their index date.

These data suggest the complexity of drawing significant conclusions from a large population study. The study of Nead et al [73] conducted on a population of 16,888 individuals with PC supported a statistically significant association between ADT use and AD and with the duration of ADT. The methodology of the study was accurate. Men who received chemotherapy were excluded because chemotherapy was associated with cognitive dysfunction and an expected high correlation between receipt of chemotherapy and ADT use. Patients with a history of dementia were considered, and only those who started the follow-up after initiation of ADT were included. The selection of the patient’s group of investigation is essential to reduce to risk of bias. Large cohorts of patients suffering from PC analyzed heterogeneous populations, including patients with different cancer progression stages, and palliative treatment can include various confounder factors, like pain, chemotherapy, and psychosocial and emotional stress. Memory declines under emotional conditions, such as depression and anxiety, and chronic psychosocial stress [74] that can trigger AD [75].

Furthermore, tests to investigate mental disorders were not regularly applied with the same methodology. Finally, yet significantly, nutritional needs and hormones such as estradiol and IGF-1, which are generally not considered in the studies, influence significantly the memory decline. Different modality of treatment is associated with a high risk of bias in the research and false results. No significant cognitive decline changes were found in men with PC who received ADT treatment by radiotherapy [59].

ADT consists of different methodologies, including bilateral orchiectomy or drug treatment using gonadotropin-releasing hormone (GnRH) agonists, antiandrogens, or combination therapy [50]. The various forms of ADT have a different effect on the hypothalamic-pituitary-gonadal axis than can affect dementia development.

Kao et al [62] in the Chinese population find no correlation between ADT and incidence of dementia, particularly ADT with GnRH agonists and without GnRH agonists. This may contribute to the variability of the effects in studies evaluating dementia. Hong et al [50] found that the cognitive decline was higher in patients receiving antiandrogens therapy than those who underwent combined androgen blockade, bilateral orchiectomy, GnRH agonist, and non-ADT treatment. In men, the androgen blocked therapy due to PC, a significant rise in the plasma levels of Aβ, and increased depression and anxiety scores were found [19].

Factors affecting cognitive decline that interact with ADT are not only physiologic but also include mood and fatigue, particularly in patients who face a high risk of death from disease, and neurocognitive decline is reasonable [76]. The cognitive decline in the elderly is a tricky clinical aspect to evaluate, and many emotional and psychological factors can be conditioning. Particularly in the studies conducted on a large population, the risk of bias should be considered. Cognitive symptoms detected with the neurophysiological test can be easily confused with psychological symptoms [66].

EFFECT OF TESTOSTERONE THERAPY ON COGNITION IN PATIENTS WITH ALZHEIMER’S DISEASE

The effect of T administration to improve cognition and reduce the progression of AD have been investigated in seventeen studies selected (Table 2) [77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93]. Some studies showed positive effects of T therapy on certain cognitive domains in normal and hypogonadal older men [78, 81, 82, 83, 88, 91, 92, 93], while others had no conclusive results [77, 79, 84, 85, 87, 90.].

Most studies that did not improve cognition and memory were conducted on a relatively healthy population (60–65 y) with sexual impairment, but not cognitive impairment. T administration was transdermal gel varying from 90 days until 4 years. Resnick et al [77], on a large population of 788 men, 65 years old, with sexual impairment, did not find any effect of T therapy on memory and cognitive function. Treatment consisted of T gel for 90 days to restore the physiological plasma T levels. Huang et al [79] T gel treatment in men 60 years old with low plasma T level and did not improve memory. Asih et al [80] found similar results in 61-year-old men with transdermal T administration. Emmelot-Vonk et al [84] investigated healthy men per 6 and 36 weeks administrating 80 mg of T undecenoate orally did not evidence any cognitive improvement. Cherrier et al [81] evaluated a small group of hypogonadal men with mild cognitive impairment, found only a modest improvement in verbal memory.

T therapy includes a wide range of doses varying from transdermal (gel 7.5 g of 1%), oral (80 mg/day), and intramuscular (200 mg/weekly), and this contributes to substantially different clinical outcomes.

Maki et al [86] found that T enanthate (200 mg i.m. every other week in ordinary men decreased verbal memory. However, the number of patients was very restricted; only 15 subjects and the study has some risk of bias. Various systematic reviews showed that low plasma T level may be associted with a reduced cognitive ability and T therapy exerted positive effects on cognitive function in normal and hypogonadal elderly men [94, 95, 96].

Verdile et al [97], in 427 men with cognitive impairment, found that LH and plasma free T inversely correlated with plasma Aβ level and brain amyloid deposition through the highly imaging, biomarkers and lifestyle study. The histopathology analysis of postmortem brain tissue in postmenopausal women showed no changes in androgen and estrogen levels. In contrast, women with AD androgen and estrogen levels were low, indifferently from the age of patients. In the male brain, aging was correlated with low androgen and estrogen levels. In those with advanced AD and brain dysfunction, the brain T levels, but not estrogen, were significantly reduced [98]. Noteworthy, in patients with memory loss, the Aβ level was correlated with total and free T levels [99]. The problem of a standardized approach to assessment is determinant.

DISCUSSION

Although animal models have demonstrated the effect of T in the reduction of βA deposition and the development of AD in the brain, in human subjects remains not homogeneous.

Majority of studies showed that ADT in patients with PC compromises cognition and increases the risk of PD. Not all ADT treatments have the same effect. Chemotherapy and androgen depressant drugs had the most deleterious effects, while LHRH inhibitors seem less involved in the cognition worsening process. Urologists when starting the therapy program should consider this clinical aspect.

Many clinical evidences showed that subjects with a low androgens level are at higher risk of cognitive decline [100], memory loss, attention deficit, and motor function in multiple sclerosis [100, 101, 102] and AD [88]. A progressive reduction of serum level of LH and T in the early preclinical stage may be considered prognostic of AD risk [97]. The prevalence of studies clearly showed that physiological plasma T levels are necessary to maintain brain function, and reduced plasma T levels predispose to dementia and AD, remarkably free T level, may predispose to cognitive decline and increased risk of AD [103]. These neurologic dysfunctions were observed before the diagnosis [104]. The most significant cohort studies found that ADT in men with PC were correlated with a higher incidence of AD [50, 51, 52, 53, 57]. ADT should be specified in which methodology it was done: antiandrogens, chemotherapy, GnRH, because each treatment exerts a different clinical effect. It can be assumed that androgens play a protective role in maintaining neuron integrity and functional integrity. T’s neuroprotective effect is expressed at cellular lev by increasing mitochondrial efficiency, improving the cellular bioenergetics more efficiently than other sex hormones, such as progesterone and estrogens [24]. The expression of AR, ERα, and aromatase is markedly reduced in hypogonadal men and men with type 2 diabetes, concerning eugonadal, but T replacement can reverse these deficits [105], contributing to significantly reduced cellular responses to sex hormones. The aromatase reduced activity is a critical factor for the 17β-estradiol production at cellular level.

However, the effects of the T administration in patients with AD evidenced controversial results. With the discrepancy in the hazard ratio, a better understanding of the methodological differences among studies should be uniformed. One of the most critical aspects in the methodology is represented by the dose of administration of T and adherence to therapy. The adherence to therapy is essential to maintain a regular plasma level of hormone. However, only 38.7% of the patient using T therapy met the criteria and the discontinuation time differed significantly among formulations that was longest among recipients of oral [106] and topical treatment [107]. T gel that can provide a low serum level and then be ineffective because a dose-dependent improvement on memory has been demonstrated [108]. T injections may be more efficacious than topical administration, as reported by Skinner et al [109]. Long-term follow-up, especially in men with mild cognitive impairment, may only be achievable with long-acting preparations (injections of T undecanoate or T pellet implants) administered by the investigator or treating physician.

Furthermore, the plasma level of free T and 17β-estradiol is necessary to evaluate the effect of the therapy. After T administration, it is essential to determine its metabolites because they help maintain neuronal efficiency, such as 17β-estradiol, DHT. Although patients receive the same T treatment, they may have a different clinical effect due to T’s different absorption and metabolism (Fig. 1).

Secondly, T administration’s effects have been evaluated in healthy men, hypogonadal men, and only a few studies assessed AD patients’ impact [88, 110]. Both found an improvement in clinical outcomes.

A recent review evidenced that T administration positively affected some cognitive domains in normal and hypogonadal older adults [94], and the clinical effect was small [111].

Another critical aspect of these studies examines the relationship between global cognition evaluation with the Mini-Mental State Examination (MMSE) and T levels. Although the test is widely used, the determinations are not sensitive to slight/subtle changes in cognition, particularly in healthy subjects and community-dwelling people [112]. Furthermore, only high levels of free T were associated with global cognition evaluated by MMSE, and a non-linear relationship between MMSE score and total T level was observed. Another critical aspect is depression, which is a clinical condition detrimental to the hippocampus and may alter a mental test.

Of relevance, T level regulates AR and ER expression in the tissues, and aromatase activity is significantly reduced in men with low T levels [105].

Besides T, other androgens can be used as neuroregeneration therapy in men and women, such as synthetic androgens (oxandrolone, stanozolol, nandrolone, etc.) and the selective androgen receptors modulators (SARM), which have a relevant neuroprotective effect in AD [113] and this potential therapeutic applications are still being explored.

However, T treatment may need to be long-term and require monitorization to maintain T serum level at physiological levels and its metabolites. It is possible that a combination of T therapy together with a healthy lifestyle approach, including improved diet and exercise, may significantly reduce AD risk [114].

Essential confounding factors, generally not considered, are nutrition, body composition [115], and physical exercise that may significantly reduce cognition decline in older adults with AD [116, 117] and the disease progression [118].

Some studies’ weak association between T and AD may reflect inverse etiological mechanisms, risk of bias, or insufficient or inappropriate control for potential confounding factors.

FUTURE PERSPECTIVE

Studies conducted on a large population, with a more specific approach to assessment adequately cognitive performance and cause-effect of T administration in AD, correcting for the confounding factors and including the evaluation of plasma levels of the total and free T, 17β-estradiol, and IGF-1 are required. Another relevant problem in medical research is related to the lacking of a dedicated population in clinical trials.

CONCLUSIONS

Although some clinical discrepancy exists between the studies, androgens significantly impact brain function and are beneficial in patients with AD. Low circulating androgen levels should be considered a substantial risk factor for AD development and memory loss. T administration in men with low plasma T levels enhances global cognitive performance, memory, and executive function and the treatment should be start at the early phase of the disease. In men and women with AD or mental impairment, androgens may improve the mental condition and reduce the progression of AD, exerting a protective effect.

Notes

Conflict of Interest:The authors have nothing to disclose.

Funding:None.

References

    1. Querfurth HW, LaFerla FM. Alzheimer's disease. N Engl J Med 2010;362:329–344.
    1. Tobore TO. On the etiopathogenesis and pathophysiology of Alzheimer's disease: a comprehensive theoretical review. J Alzheimers Dis 2019;68:417–437.
    1. Selkoe DJ, Hardy J. The amyloid hypothesis of Alzheimer's disease at 25 years. EMBO Mol Med 2016;8:595–608.
    1. Spires-Jones TL, Hyman BT. The intersection of amyloid beta and tau at synapses in Alzheimer's disease. Neuron 2014;82:756–771.
    1. Bianchi VE, Rizzi L, Bresciani E, Omeljaniuk RJ, Torsello A. Androgen therapy in neurodegenerative diseases. J Endocr Soc 2020;4:bvaa120
    1. Baum LW. Sex, hormones, and Alzheimer's disease. J Gerontol A Biol Sci Med Sci 2005;60:736–743.
    1. Yao PL, Zhuo S, Mei H, Chen XF, Li N, Zhu TF, et al. Androgen alleviates neurotoxicity of β-amyloid peptide (Aβ) by promoting microglial clearance of Aβ and inhibiting microglial inflammatory response to Aβ. CNS Neurosci Ther 2017;23:855–865.
    1. Huo DS, Sun JF, Zhang B, Yan XS, Wang H, Jia JX, et al. Protective effects of testosterone on cognitive dysfunction in Alzheimer's disease model rats induced by oligomeric beta amyloid peptide 1-42. J Toxicol Environ Health A 2016;79:856–863.
    1. Yan 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;14:649–657.
    1. Lau CF, Ho YS, Hung CH, Wuwongse S, Poon CH, Chiu K, et al. Protective effects of testosterone on presynaptic terminals against oligomeric β-amyloid peptide in primary culture of hippocampal neurons. Biomed Res Int 2014;2014:103906
    1. Jia JX, Cui CL, Yan XS, Zhang BF, Song W, Huo DS, et al. Effects of testosterone on synaptic plasticity mediated by androgen receptors in male SAMP8 mice. J Toxicol Environ Health A 2016;79:849–855.
    1. Grimm A, Schmitt K, Lang UE, Mensah-Nyagan AG, Eckert A. Improvement of neuronal bioenergetics by neurosteroids: implications for age-related neurodegenerative disorders. Biochim Biophys Acta 2014;1842(12 Pt A):2427–2438.
    1. Pintana H, Chattipakorn N, Chattipakorn S. Testosterone deficiency, insulin-resistant obesity and cognitive function. Metab Brain Dis 2015;30:853–876.
    1. Ota H, Akishita M, Akiyoshi T, Kahyo T, Setou M, Ogawa S, et al. Testosterone deficiency accelerates neuronal and vascular aging of SAMP8 mice: protective role of eNOS and SIRT1. PLoS One 2012;7:e29598
    1. Jian-xin J, Cheng-li C, Song W, Yan XS, Huo DS, Wang H, et al. Effects of testosterone treatment on synaptic plasticity and behavior in senescence accelerated mice. J Toxicol Environ Health A 2015;78:1311–1320.
    1. Kang L, Li S, Xing Z, Li J, Su Y, Fan P, et al. Dihydrotestosterone treatment delays the conversion from mild cognitive impairment to Alzheimer's disease in SAMP8 mice. Horm Behav 2014;65:505–515.
    1. Lv W, Du N, Liu Y, Fan X, Wang Y, Jia X, et al. Low testosterone level and risk of Alzheimer's disease in the elderly men: a systematic review and meta-analysis. Mol Neurobiol 2016;53:2679–2684.
    1. Lee JH, Byun MS, Yi D, Choe YM, Choi HJ, Baek H, et al. KBASE Research Group. Sex-specific association of sex hormones and gonadotropins, with brain amyloid and hippocampal neurodegeneration. Neurobiol Aging 2017;58:34–40.
    1. Almeida OP, Waterreus A, Spry N, Flicker L, Martins RN. One year follow-up study of the association between chemical castration, sex hormones, beta-amyloid, memory and depression in men. Psychoneuroendocrinology 2004;29:1071–1081.
    1. Simerly 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;294:76–95.
    1. Goodenough S, Engert S, Behl C. Testosterone stimulates rapid secretory amyloid precursor protein release from rat hypothalamic cells via the activation of the mitogen-activated protein kinase pathway. Neurosci Lett 2000;296:49–52.
    1. Wang L, Pei JH, Jia JX, Wang J, Song W, Fang X, et al. Inhibition of oxidative stress by testosterone improves synaptic plasticity in senescence accelerated mice. J Toxicol Environ Health A 2019;82:1061–1068.
    1. McAllister 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;30:7326–7334.
    1. Grimm A, Biliouris EE, Lang UE, Götz J, Mensah-Nyagan AG, Eckert A. Sex hormone-related neurosteroids differentially rescue bioenergetic deficits induced by amyloid-β or hyperphosphorylated tau protein. Cell Mol Life Sci 2016;73:201–215.
    1. di Michele F, Luchetti S, Bernardi G, Romeo E, Longone P. Neurosteroid and neurotransmitter alterations in Parkinson's disease. Front Neuroendocrinol 2013;34:132–142.
    1. Zhang Y, Champagne N, Beitel LK, Goodyer CG, Trifiro M, LeBlanc A. Estrogen and androgen protection of human neurons against intracellular amyloid beta1-42 toxicity through heat shock protein 70. J Neurosci 2004;24:5315–5321.
    1. Frye CA, Edinger KL, Lephart ED, Walf AA. 3alpha-androstanediol, but not testosterone, attenuates age-related decrements in cognitive, anxiety, and depressive behavior of male rats. Front Aging Neurosci 2010;2:15
    1. Adams MM, Morrison JH, Gore AC. N-methyl-D-aspartate receptor mRNA levels change during reproductive senescence in the hippocampus of female rats. Exp Neurol 2001;170:171–179.
    1. Sahab-Negah S, Hajali V, Moradi HR, Gorji A. The impact of estradiol on neurogenesis and cognitive functions in Alzheimer's disease. Cell Mol Neurobiol 2020;40:283–299.
    1. Qin Y, An D, Xu W, Qi X, Wang X, Chen L, et al. Estradiol replacement at the critical period protects hippocampal neural stem cells to improve cognition in APP/PS1 mice. Front Aging Neurosci 2020;12:240
    1. Davison SL, Bell R, Donath S, Montalto JG, Davis SR. Androgen levels in adult females: changes with age, menopause, and oophorectomy. J Clin Endocrinol Metab 2005;90:3847–3853.
    1. Burger HG, Dudley EC, Cui J, Dennerstein L, Hopper JL. A prospective longitudinal study of serum testosterone, dehydroepiandrosterone sulfate, and sex hormone-binding globulin levels through the menopause transition. J Clin Endocrinol Metab 2000;85:2832–2838.
    1. Simpson ER, Davis SR. Minireview: aromatase and the regulation of estrogen biosynthesis--some new perspectives. Endocrinology 2001;142:4589–4594.
    1. Tang MX, Jacobs D, Stern Y, Marder K, Schofield P, Gurland B, et al. Effect of oestrogen during menopause on risk and age at onset of Alzheimer's disease. Lancet 1996;348:429–432.
    1. Zandi PP, Carlson MC, Plassman BL, Welsh-Bohmer KA, Mayer LS, Steffens DC, et al. Hormone replacement therapy and incidence of Alzheimer disease in older women: the Cache County Study. JAMA 2002;288:2123–2129.
    1. Song YJ, Li SR, Li XW, Chen X, Wei ZX, Liu QS, et al. The effect of estrogen replacement therapy on Alzheimer's disease and Parkinson's disease in postmenopausal women: a meta-analysis. Front Neurosci 2020;14:157
    1. Imtiaz B, Tuppurainen M, Rikkonen T, Kivipelto M, Soininen H, Kröger H, et al. Postmenopausal hormone therapy and Alzheimer disease: a prospective cohort study. Neurology 2017;88:1062–1068.
    1. Imtiaz B, Taipale H, Tanskanen A, Tiihonen M, Kivipelto M, Heikkinen AM, et al. Risk of Alzheimer's disease among users of postmenopausal hormone therapy: a nationwide case-control study. Maturitas 2017;98:7–13.
    1. Savolainen-Peltonen H, Rahkola-Soisalo P, Hoti F, Vattulainen P, Gissler M, Ylikorkala O, et al. Use of postmenopausal hormone therapy and risk of Alzheimer's disease in Finland: nationwide case-control study. BMJ 2019;364:l665
    1. Shumaker SA, Legault C, Rapp SR, Thal L, Wallace RB, Ockene JK, et al. Estrogen plus progestin and the incidence of dementia and mild cognitive impairment in postmenopausal women: the Women's Health Initiative Memory Study: a randomized controlled trial. JAMA 2003;289:2651–2662.
    1. Tolppanen AM, Tiihonen M, Taipale H, Koponen M, Tanskanen A, Lavikainen P, et al. Systemic estrogen use and discontinuation after Alzheimer's disease diagnosis in Finland 2005-2012: a nationwide exposure-matched cohort study. Drugs Aging 2018;35:985–992.
    1. Shao H, Breitner JC, Whitmer RA, Wang J, Hayden K, Wengreen H, et al. Hormone therapy and Alzheimer disease dementia: new findings from the Cache County Study. Neurology 2012;79:1846–1852.
    1. Gillies GE, McArthur S. Estrogen actions in the brain and the basis for differential action in men and women: a case for sex-specific medicines. Pharmacol Rev 2010;62:155–198.
    1. Cui J, Shen Y, Li R. Estrogen synthesis and signaling pathways during aging: from periphery to brain. Trends Mol Med 2013;19:197–209.
    1. Cummings SR, Ettinger B, Delmas PD, Kenemans P, Stathopoulos V, Verweij P, et al. The effects of tibolone in older postmenopausal women. N Engl J Med 2008;359:697–708.
    1. Albertazzi P, Natale V, Barbolini C, Teglio L, Di Micco R. The effect of tibolone versus continuous combined norethisterone acetate and oestradiol on memory, libido and mood of postmenopausal women: a pilot study. Maturitas 2000;36:223–229.
    1. Genazzani AR, Pluchino N, Bernardi F, Centofanti M, Luisi M. Beneficial effect of tibolone on mood, cognition, well-being, and sexuality in menopausal women. Neuropsychiatr Dis Treat 2006;2:299–307.
    1. Palomba S, Orio F Jr, Falbo A, Oppedisano R, Tolino A, Zullo F. Tibolone reverses the cognitive effects caused by leuprolide acetate administration, improving mood and quality of life in patients with symptomatic uterine leiomyomas. Fertil Steril 2008;90:165–173.
    1. Correia SC, Santos RX, Cardoso S, Carvalho C, Santos MS, Oliveira CR, et al. Effects of estrogen in the brain: is it a neuroprotective agent in Alzheimer's disease? Curr Aging Sci 2010;3:113–126.
    1. Hong JH, Huang CY, Chang CH, Muo CH, Jaw FS, Lu YC, et al. Different androgen deprivation therapies might have a differential impact on cognition - an analysis from a population-based study using time-dependent exposure model. Cancer Epidemiol 2020;64:101657
    1. Huang WK, Liu CH, Pang ST, Liu JR, Chang JW, Liaw CC, et al. Type of androgen deprivation therapy and risk of dementia among patients with prostate cancer in Taiwan. JAMA Netw Open 2020;3:e2015189
    1. Jayadevappa R, Chhatre S, Malkowicz SB, Parikh RB, Guzzo T, Wein AJ. Association between androgen deprivation therapy use and diagnosis of dementia in men with prostate cancer. JAMA Netw Open 2019;2:e196562
    1. Krasnova A, Epstein M, Marchese M, Dickerman BA, Cole AP, Lipsitz SR, et al. Risk of dementia following androgen deprivation therapy for treatment of prostate cancer. Prostate Cancer Prostatic Dis 2020;23:410–418.
    1. Jarzemski P, Brzoszczyk B, Popiołek A, Stachowicz-Karpińska A, Gołota S, Bieliński M, et al. Cognitive function, depression, and anxiety in patients undergoing radical prostatectomy with and without adjuvant treatment. Neuropsychiatr Dis Treat 2019;15:819–829.
    1. Robinson D, Garmo H, Van Hemelrijck M, Damber JE, Bratt O, Holmberg L, et al. Androgen deprivation therapy for prostate cancer and risk of dementia. BJU Int 2019;124:87–92.
    1. Tae BS, Jeon BJ, Shin SH, Choi H, Bae JH, Park JY. Correlation of androgen deprivation therapy with cognitive dysfunction in patients with prostate cancer: a nationwide population-based study using the National Health Insurance Service database. Cancer Res Treat 2019;51:593–602.
    1. Nguyen C, Lairson DR, Swartz MD, Du XL. Risks of major long-term side effects associated with androgen-deprivation therapy in men with prostate cancer. Pharmacotherapy 2018;38:999–1009.
    1. Marzouk S, Naglie G, Tomlinson G, Duff Canning S, Breunis H, Timilshina N, et al. Impact of androgen deprivation therapy on self-reported cognitive function in men with prostate cancer. J Urol 2018;200:327–334.
    1. Deka R, Simpson DR, Bryant AK, Nalawade V, McKay R, Murphy JD, et al. Association of androgen deprivation therapy with dementia in men with prostate cancer who receive definitive radiation therapy. JAMA Oncol 2018;4:1616–1617.
    1. Baik 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;35:3401–3409.
    1. Alibhai 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 months in men with prostate cancer. Cancer 2017;123:237–244.
    1. Kao LT, Lin HC, Chung SD, Huang CY. No increased risk of dementia in patients receiving androgen deprivation therapy for prostate cancer: a 5-year follow-up study. Asian J Androl 2017;19:414–417.
    1. Gunlusoy B, Ceylan Y, Koskderelioglu A, Gedizlioglu M, Degirmenci T, Ortan P, et al. Cognitive effects of androgen deprivation therapy in men with advanced prostate cancer. Urology 2017;103:167–172.
    1. Nead KT, Gaskin G, Chester C, Swisher-McClure S, Leeper NJ, Shah NH. Association between androgen deprivation therapy and risk of dementia. JAMA Oncol 2017;3:49–55.
    1. Khosrow-Khavar F, Rej S, Yin H, Aprikian A, Azoulay L. Androgen deprivation therapy and the risk of dementia in patients with prostate cancer. J Clin Oncol 2017;35:201–207.
    1. Wu LM, Tanenbaum ML, Dijkers MP, Amidi A, Hall SJ, Penedo FJ, et al. Cognitive and neurobehavioral symptoms in patients with non-metastatic prostate cancer treated with androgen deprivation therapy or observation: a mixed methods study. Soc Sci Med 2016;156:80–89.
    1. Chung SD, Lin HC, Tsai MC, Kao LT, Huang CY, Chen KC. Androgen deprivation therapy did not increase the risk of Alzheimer's and Parkinson's disease in patients with prostate cancer. Andrology 2016;4:481–485.
    1. Nead KT, Gaskin G, Chester C, Swisher-McClure S, Dudley JT, Leeper NJ, et al. Androgen deprivation therapy and future Alzheimer's disease risk. J Clin Oncol 2016;34:566–571.
    1. Gonzalez BD, Jim HS, Booth-Jones M, Small BJ, Sutton SK, Lin HY, et al. Course and predictors of cognitive function in patients with prostate cancer receiving androgen-deprivation therapy: a controlled comparison. J Clin Oncol 2015;33:2021–2027.
    1. Gandy S, Almeida OP, Fonte J, Lim D, Waterrus A, Spry N, et al. Chemical andropause and amyloid-beta peptide. JAMA 2001;285:2195–2196.
    1. Sun M, Cole AP, Hanna N, Mucci LA, Berry DL, Basaria S, et al. Cognitive impairment in men with prostate cancer treated with androgen deprivation therapy: a systematic review and meta-analysis. J Urol 2018;199:1417–1425.
    1. Siebert AL, Lapping-Carr L, Morgans AK. Neuropsychiatric impact of androgen deprivation therapy in patients with prostate cancer: current evidence and recommendations for the clinician. Eur Urol Focus 2020;6:1170–1179.
    1. Nead KT, Sinha S, Nguyen PL. Androgen deprivation therapy for prostate cancer and dementia risk: a systematic review and meta-analysis. Prostate Cancer Prostatic Dis 2017;20:259–264.
    1. Caruso A, Nicoletti F, Mango D, Saidi A, Orlando R, Scaccianoce S. Stress as risk factor for Alzheimer's disease. Pharmacol Res 2018;132:130–134.
    1. Tran TT, Srivareerat M, Alkadhi KA. Chronic psychosocial stress triggers cognitive impairment in a novel at-risk model of Alzheimer's disease. Neurobiol Dis 2010;37:756–763.
    1. McHugh DJ, Root JC, Nelson CJ, Morris MJ. Androgen-deprivation therapy, dementia, and cognitive dysfunction in men with prostate cancer: how much smoke and how much fire? Cancer 2018;124:1326–1334.
    1. Resnick SM, Matsumoto AM, Stephens-Shields AJ, Ellenberg SS, Gill TM, Shumaker SA, et al. Testosterone treatment and cognitive function in older men with low testosterone and age-associated memory impairment. JAMA 2017;317:717–727.
    1. Wahjoepramono 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;15:337–343.
    1. Huang 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;4:657–665.
    1. Asih 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;14:576–586.
    1. Cherrier 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;30:421–430.
    1. Borst SE, Yarrow JF, Fernandez C, Conover CF, Ye F, Meuleman JR, et al. Cognitive effects of testosterone and finasteride administration in older hypogonadal men. Clin Interv Aging 2014;9:1327–1333.
    1. Young LA, Neiss MB, Samuels MH, Roselli CE, Janowsky JS. Cognition is not modified by large but temporary changes in sex hormones in men. J Clin Endocrinol Metab 2010;95:280–288.
    1. Emmelot-Vonk MH, Verhaar HJ, Nakhai Pour HR, Aleman A, Lock TM, Bosch JL, et al. Effect of testosterone supplementation on functional mobility, cognition, and other parameters in older men: a randomized controlled trial. JAMA 2008;299:39–52.
    1. Vaughan C, Goldstein FC, Tenover JL. Exogenous testosterone alone or with finasteride does not improve measurements of cognition in healthy older men with low serum testosterone. J Androl 2007;28:875–882.
    1. Maki PM, Ernst M, London ED, Mordecai KL, Perschler P, Durso SC, et al. Intramuscular testosterone treatment in elderly men: evidence of memory decline and altered brain function. J Clin Endocrinol Metab 2007;92:4107–4114.
    1. Cherrier MM, Matsumoto AM, Amory JK, Johnson M, Craft S, Peskind ER, et al. Characterization of verbal and spatial memory changes from moderate to supraphysiological increases in serum testosterone in healthy older men. Psychoneuroendocrinology 2007;32:72–79.
    1. Lu 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;63:177–185.
    1. Haren MT, Wittert GA, Chapman IM, Coates P, Morley JE. Effect of oral testosterone undecanoate on visuospatial cognition, mood and quality of life in elderly men with low-normal gonadal status. Maturitas 2005;50:124–133.
    1. Kenny AM, Fabregas G, Song C, Biskup B, Bellantonio S. Effects of testosterone on behavior, depression, and cognitive function in older men with mild cognitive loss. J Gerontol A Biol Sci Med Sci 2004;59:75–78.
    1. Tan RS, Pu SJ. A pilot study on the effects of testosterone in hypogonadal aging male patients with Alzheimer's disease. Aging Male 2003;6:13–17.
    1. O’Connor DB, Archer J, Hair WM, Wu FC. Activational effects of testosterone on cognitive function in men. Neuropsychologia 2001;39:1385–1394.
    1. Cherrier MM, Asthana S, Plymate S, Baker L, Matsumoto AM, Peskind E, et al. Testosterone supplementation improves spatial and verbal memory in healthy older men. Neurology 2001;57:80–88.
    1. Mohamad NV, Ima-Nirwana S, Chin KY. A review on the effects of testosterone supplementation in hypogonadal men with cognitive impairment. Curr Drug Targets 2018;19:898–906.
    1. Beauchet O. Testosterone and cognitive function: current clinical evidence of a relationship. Eur J Endocrinol 2006;155:773–781.
    1. Hua JT, Hildreth KL, Pelak VS. Effects of testosterone therapy on cognitive function in aging: a systematic review. Cogn Behav Neurol 2016;29:122–138.
    1. Verdile G, Laws SM, Henley D, Ames D, Bush AI, Ellis KA, et al. AIBL Research Group. Associations between gonadotropins, testosterone and β amyloid in men at risk of Alzheimer's disease. Mol Psychiatry 2014;19:69–75.
    1. Rosario 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;32:604–613.
    1. Gillett MJ, Martins RN, Clarnette RM, Chubb SA, Bruce DG, Yeap BB. Relationship between testosterone, sex hormone binding globulin and plasma amyloid beta peptide 40 in older men with subjective memory loss or dementia. J Alzheimers Dis 2003;5:267–269.
    1. Cai Z, Li H. An updated review: androgens and cognitive impairment in older men. Front Endocrinol (Lausanne) 2020;11:586909
    1. Bove R, Musallam A, Healy BC, Raghavan K, Glanz BI, Bakshi R, et al. Low testosterone is associated with disability in men with multiple sclerosis. Mult Scler 2014;20:1584–1592.
    1. Cheung YT, Chemaitilly W, Mulrooney DA, Brinkman TM, Liu W, Banerjee P, et al. Association between dehydroepiandrosterone-sulfate and attention in long-term survivors of childhood acute lymphoblastic leukemia treated with only chemotherapy. Psychoneuroendocrinology 2017;76:114–118.
    1. Hogervorst E, Bandelow S, Combrinck M, Smith AD. Low free testosterone is an independent risk factor for Alzheimer's disease. Exp Gerontol 2004;39:1633–1639.
    1. Moffat SD, Zonderman AB, Metter EJ, Kawas C, Blackman MR, Harman SM, et al. Free testosterone and risk for Alzheimer disease in older men. Neurology 2004;62:188–193.
    1. Ghanim H, Dhindsa S, Abuaysheh S, Batra M, Kuhadiya ND, Makdissi A, et al. Diminished androgen and estrogen receptors and aromatase levels in hypogonadal diabetic men: reversal with testosterone. Eur J Endocrinol 2018;178:277–283.
    1. Martins D, Yao Z, Tadrous M, Shah BR, Juurlink DN, Mamdani MM, et al. Ontario Drug Policy Research Network. The appropriateness and persistence of testosterone replacement therapy in Ontario. Pharmacoepidemiol Drug Saf 2017;26:119–126.
    1. Grabner M, Hepp Z, Raval A, Tian F, Khera M. Topical testosterone therapy adherence and outcomes among men with primary or secondary hypogonadism. J Sex Med 2018;15:148–158.
    1. Jaeger ECB, Miller LE, Goins EC, Super CE, Chyr CU, Lower JW, et al. Testosterone replacement causes dose-dependent improvements in spatial memory among aged male rats. Psychoneuroendocrinology 2020;113:104550
    1. Skinner JW, Otzel DM, Bowser A, Nargi D, Agarwal S, Peterson MD, et al. Muscular responses to testosterone replacement vary by administration route: a systematic review and meta-analysis. J Cachexia Sarcopenia Muscle 2018;9:465–481.
    1. Preece P, Virley DJ, Costandi M, Coombes R, Moss SJ, Mudge AW, et al. Amyloid precursor protein mRNA levels in Alzheimer's disease brain. Brain Res Mol Brain Res 2004;122:1–9.
    1. Tan 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;27:1232–1246.
    1. Gluhm S, Goldstein J, Loc K, Colt A, Liew CV, Corey-Bloom J. Cognitive performance on the mini-mental state examination and the montreal cognitive assessment across the healthy adult lifespan. Cogn Behav Neurol 2013;26:1–5.
    1. Jayaraman A, Christensen A, Moser VA, Vest RS, Miller CP, Hattersley G, et al. Selective androgen receptor modulator RAD140 is neuroprotective in cultured neurons and kainate-lesioned male rats. Endocrinology 2014;155:1398–1406.
    1. Asih PR, Tegg ML, Sohrabi H, Carruthers M, Gandy SE, Saad F, et al. Multiple mechanisms linking type 2 diabetes and Alzheimer's disease: testosterone as a modifier. J Alzheimers Dis 2017;59:445–466.
    1. Coulbault L, Ritz L, Vabret F, Lannuzel C, Boudehent C, Nowoczyn M, et al. Thiamine and phosphate esters concentrations in whole blood and serum of patients with alcohol use disorder: a relation with cognitive deficits. Nutr Neurosci 2021;24:530–541.
    1. Jia RX, Liang JH, Xu Y, Wang YQ. Effects of physical activity and exercise on the cognitive function of patients with Alzheimer disease: a meta-analysis. BMC Geriatr 2019;19:181
    1. Yu F, Vock DM, Zhang L, Salisbury D, Nelson NW, Chow LS, et al. Cognitive effects of aerobic exercise in Alzheimer's disease: a pilot randomized controlled trial. J Alzheimers Dis 2021;80:233–244.
    1. Valenzuela PL, Castillo-García A, Morales JS, de la Villa P, Hampel H, Emanuele E, et al. Exercise benefits on Alzheimer's disease: state-of-the-science. Ageing Res Rev 2020;62:101108

Metrics
Share
Figures

1 / 1

Tables

1 / 2

PERMALINK