Open access peer-reviewed chapter

Neuroprotection in Perimenopausal Women

Written By

Manuela Cristina Russu and Alexandra Cristina Antonescu

Submitted: 07 August 2017 Reviewed: 24 January 2018 Published: 21 February 2018

DOI: 10.5772/intechopen.74330

From the Edited Volume

Sex Hormones in Neurodegenerative Processes and Diseases

Edited by Gorazd Drevenšek

Chapter metrics overview

1,408 Chapter Downloads

View Full Metrics

Abstract

Endocrine and neural senescence overlap in time, by intertwined complex feedback loops. Womens’ brain is genetically more prone to suffer during life, and perimenopause is a “critical period” in neuroaging, when the degenerative processes begin. Many hypotheses on the multifactorial nature of women’s brain aging are elaborated, and tested in high-tech research centers. The most analyzed Alzheimer’s disease (AD) is characterized not only by Aβ oligomers and fibrils accumulation, but also by metabolic and inflammatory changes, with the onset during menopausal transition and early years of menopause. Deep analysis of endocrine, neural, and metabolic pathways are giving new insights to the sequential view of Aβ-centric in AD pathogenesis, prevention, and treatment from perimenopause, for maintaining women’s neurological health.

Keywords

  • neuroaging
  • perimenopause
  • critical period

1. Introduction: sex differences in contemporary neurodegenerative disorders

Ovarian aging is very well-known in contemporary women’s life, and the jeopardizing menopausal effects of sex steroid hormones deficiency are clinically evident in late-life mental disorders. Endocrine and neural senescence overlap in time, and are mechanistically intertwined in complex feedback loops.

In the past century, both life expectancy and the average age of onset of menopause for women in many countries from Western Europe and North America were slightly over 50 years, whereas currently, women can expect to live until the age of 80 years, although the average age of menopause remains in the early 50s. Given the importance of the brain as a target organ for sex steroids, it is not surprising that many of the complaints that prompt women to seek treatment related to menopause are neurological in origin.

Dementia with its most severe entities, such as the Alzheimer’s disease (AD), Parkinson’s disease (PD), multiple sclerosis (MS), and amyotrophic lateral sclerosis (ALS), are the most frequent contemporary neurodegenerative disorders, connected in majority by neural cell loss and neuroinflammation.

There are sex/gender differences in cognitive trajectories in clinically normal older adults [1], and women are known to have a higher propensity to develop AD versus men [2, 3], a higher risk of mild cognitive impairment and a lower risk, but poorer outcomes after stroke [4]. Women’s risk for AD is considered to be through their organizational effects during developmental sexual differentiation of the fetal brain [5].

The AD is the most prevalent form of old-life mental failure in worldwide humans, being a progressive neurodegenerative disorder, for which a number of genetic, environmental, and lifestyle risk factors have been identified. The estimated prevalence of all-cause of dementia varies from 4.7% in Central Europe to 8.7% in North Africa/Middle East, with North America falling at 6.4%. Currently, over 46 million individuals live with dementia worldwide and the number is projected to increase to 131.5 million by 2050 [6]. The AD is familial with early-onset and sporadic with late-onset, and the present chapter will discuss the sporadic AD with late-onset in women, the most common form of AD representing more than 95% of the current human AD population [7].

In this very moment, the geroscience research is imperative with aims to forward a better and full understanding of neurodegeneration/neuroprotection of “the sexome” [8], and to prevent or delay by every tool the deleterious effects of brain aging [9]. Estrogen deficiency or estrogen disrupters are associated from menopause transition with episodic memory troubles, a cognitive domain in which impairments are associated with the increased risk of AD, being less known the onset of the other neurodegenerative disorders [10].

AD has an insidious onset and a gradual progression over several years—from 1.5 to 8 or up to 10 years, or a preclinical stage with a subtle loss of cognitive functioning—as verbal memory on new information, that precede several years the AD diagnostic, period considered as a transition period from normal aging brain to AD [11]. During this period, there are discovered several subtypes of mild cognitive impairment (MCI), 10–15% with the risk to future evolution to AD per year [12], or a 12% conversion rate from MIC to dementia yearly [13]. The characteristic deposits of β-amyloid and tau proteins depicted by neuroimaging or at autopsy located in the hippocampus, medial temporal regions, parietal, and frontal cortical regions [14] may be prevented from extension, as other structural degenerative diseases. It is imperious to prevent the intracellular appearance of the amyloid peptide, which induces by its toxicity neuronal apoptosis and cell death, events that can be prevented by sex steroid hormones [15].

The clinical symptoms/signs of MCI are considered by neuroscientists as prodrome to AD [16], and their algorithms for diagnosis may permit to initiate the hormone/estrogen therapy (HT/ET), because their onset moment is coincidental to perimenopause, as considering the North American clinicians [17]. The “timing” theory regarding the reproductive stage and role of time since menopause at initiation of HT/ET may work in preventing the mental deterioration due to aging [17], and perimenopause can be the “critical window” for opportunity in neuroprotection with steroids. The perimenopausal transition might also represent a “window of opportunity” to prevent age-related neurological diseases [10, 18, 19].

Advertisement

2. Hormonal and genetic data on perimenopausal neuroaging

The data of this chapter are regarding brain aging in perimenopause—a part in woman’s reproductive life, systematized in the stages of reproductive aging (STRAW) [20].

Menopausal transition starts with the variation of cycles duration and ends with the last period (recognized only after 12 months of amenorrhea), natural menopause at the average age 51 years, premature menopause [premature ovarian failure (POF)] before 40 years, and early menopause between 40 and 45 years [21, 22]. Women with POF have been reported to have more anxiety, depression, somatization, sensitivity, hostility, and psychological distress than women with normal ovaries [23]. Perimenopause or “near menopause” starts from the stage −2 of menopausal transition and ends at 12 months after last menstruation, may be of 10–15 years. During this period, there are important variations of sex steroids, summarized by low ovarian inhibin [24], which in turn reduces the restraint on both the hypothalamus and pituitary gland, and results in elevated pituitary gonadotropin FSH, increased also by the hypothalamic gonadotropin-releasing hormone (GnRH). During the late menopause transition and a part of perimenopause, despite occasional episodes of normal cycling, women are exposed to periods of estrogen withdrawal, fewer ovulatory cycles, and prolonged hypogonadism, ultimately leading to the last menstrual period, after which is an elevated level of gonadotrophin secretion (only tonic, not phasic) [25]. During this phase, besides the low ovarian E2 and progesterone, there are productions of androgens and growth factors, which will decline in future years of postmenopause [26]. The ovaries are stimulated during menopausal transition and early postmenopausal years by gonadotropins, but the pulsatile GnRH pattern is different in different species before reproductive failure. It is a decrease of GnRH gene expression in many middle-aged rats [27], and an increase in perimenopausal rhesus monkeys [28].

Premature menopause/early menopause can be spontaneous or induced; after medical interventions such as chemotherapy/radiotherapy or surgery. The most common cause of premature/early menopause is bilateral oophorectomy with/without hysterectomy. Primary ovarian failure (POF) may be a cause of early/premature menopause, for ischemic stroke [29], as for all cardiovascular diseases risks [30], and these conditions were first described as a cause of neurological disturbances in different European, North American, and Japanese populations [21, 31, 32, 33].

Bilateral oophorectomy at premenopausal ages is inducing drops of E2 and testosterone levels, by 40–50%, and an abruptly rise in FSH levels, the levels of androgen being lower than in natural menopause at ages of 65 years, when women in normal or in premature/early menopause continue to have some levels of androgens [34]. As it is shown in the Table 1, the hazard ratios reached statistical significance in cases with bilateral oophorectomy: at the age < 34 years for dementia and <38 years for PD. Perimenopause is a “fragile” period in woman’s life, comparable to the fragility of the adolescence, if we speak about “hormonal storms,” but the hormonal pyramid is upside down, and more than these, the North American neurologists are considering perimenopause as a neurological transition state [35], because the characteristic symptoms regarding thermoregulation, sleep, circadian rhythms, and sensory processing are of neurological nature, besides the changes of cognitive function [36].

Adjusted odd ratio for dementia after unilateral oophorectomy
Age at surgery (years) Hazard ratio CI 95% P value
<43 1.74 0.97–3.14 0.06
43–48 1.68 1.06–2.66 0.03
>48 1.09 0.74–1.61 0.66
Adjusted odd ratio for dementia after bilateral oophorectomy
<34 4.61 2.52–8.43 <0.0001
34–41 1.23 0.67–2.26 0.51
>41 1.50 1.05–2.13 0.03
Adjusted odd ratio for PD after bilateral oophorectomy
<38 2.85 1.28–6.35 0.001
38–45 1.38 1.28–6.35 0.42
>45 1.38 0.92–3.03 0.09

Table 1.

Utero-ovarian surgery and neurological disturbances in premenopause.

Cases of cognitive impairment/dementia and Parkinson disease (PD) in women with unilateral (813) and bilateral (676) oophorectomy: For a nonmalignant disease, in Olmsted County, Minnesota (USA) during 1950–1987, followed up the death or the finish of study at 2001–2006 (Rocca et al. [32]).

The central and peripheral hormonal changes in menopausal transition and perimenopause were assessed in many research centers from Western Europe [37], Australia [24, 38], North America [25], and besides these, the rat models on gene expression analyses demonstrated that there are two distinct aging programs: chronological and endocrine, regarding bioenergetic gene expression involved in brain metabolism and synaptic plasticity [39].

The endocrine transition marked by changing from regular to irregular menstrual cycles is characterized by the impairment of the energy metabolism, glucose hypometabolism, and chronic oxidative stress, which were demonstrated by gene expression in brain metabolism, mitochondrial function, and long-term potentiation. Rat model analysis on brain energetic metabolism in menopausal transition demonstrated that insulin/insulin-like growth factor 1 and adenosine monophosphate-activated protein kinase/peroxisome proliferator-activated receptor gamma coactivator-1-alpha (AMPK/PGC1α) signaling pathways are upstream regulators [39], and these pathways suggest the critical role of E2 in neuronal survival. E2 stimulates the mitochondrial sequestration of Ca2+ and protects neurons against adverse consequences of excess cytoplasmic Ca2+ and subsequent dysregulation of Ca2+ homeostasis, with concomitant preservation of mitochondrial respiratory capacity [40].

Genetic analyses demonstrated that the menstrual cycles acyclicity is accompanied by a rise in genes required for fatty acid metabolism, a decline of genes required for mitochondrial function, β-amyloid degradation, and neuroinflammation including increased number in microglia population in aging hippocampus [41], plus the shift of microglia activation with predominant production of inflammatory cytokines [42], and a higher basal level of complement cascade genes and interleukin 1 receptor-like 1 in women versus men [43].

There are neuroimmune modulation differences in normal memory processes and memory dysregulation, in the roles of cytokines, astrocytes, and microglia in females and males [44]. These differences are from early development and differentiation of the brain [5], making women’s brain inherently vulnerable to neurodegenerative diseases, to a higher risk of mild cognitive impairment and AD in advanced ages [45] (though not all studies are in agreement on this point, [1]), and non-neurodegenerative cognitive impairments fact that drive to the deleterious/beneficial consequences for estrogen therapy. The metabolic and neuroinflammatory changes are connected via redox regulation during normal brain aging, and may be predictive for later-life vulnerability to hypometabolic conditions of AD [46].

There are new animal studies on female neuroaging, regarding the microglia involvement in neurogenesis [47], to innate immune system [48], being revealed the microglia sensome by direct RNA sequencing [49]. Molecular studies on mice aging [50] revealed a central role of gender in the transcriptomic response in hippocampal and cortex aging, demonstrating sexually divergent changes of neuroinflammation, mainly an increase of microglia-specific genes, and C1q protein expression of the complement system, in the activation of astrocytes, and in cytokine release and function in aging. C1qa induction is a driver of synapse loss with greater C1qa induction associated with poorer cognitive performance. It is considered that the age-related changes in inflammatory hippocampal genes amplified in women after estrogen failure may contribute to sex differences in age-related neurological diseases. There are classes of genes in which inductions and reductions in gene expression are acting synergistically in female aging hippocampus [50].

The rise of microglia-specific genes in aging females is interrelated to a significant decrease in the activation of two pro-neurogenesis pathways evident in aging hippocampus: Notch1 and Presenilin 1 and 2 (PSEN1 and PSEN2) regulated genes [51]: Notch1 is necessary for neural stem cell maintenance [52], the PSEN1 expression regulates neuroprogenitor cell differentiation [53], and the defects in PSEN1 expression are associated with the manifestation of AD in old age [54]. Another change of neuroinflammatory genes in aging women is that of Tyrobp known as TREM2, as a causal regulator in microglia-associated changes in AD [55], and its proper mechanism in AD etiology is still being determined [56].

Advertisement

3. Hypothesis on brain aging and neurodegeneration during perimenopause

The months/years of perimenopause represent an important moment during women’s aging, when steroids and their receptors decline is evident in the hippocampal and cortical neurons, after estrogen exposure during the reproductive years. The estrogens decline is associated/acts synergic to other factors as hypertension, diabetes, hypoxia/obstructive sleep apnea, obesity, vitamin B12/folate deficiency, depression, and traumatic brain injury to promote different pathological mechanisms involved in brain aging, memory impairment, and AD.

The Californian and Australian neuroscientists had shown that chronic cerebral hypoperfusion deprives the brain from its two paramount trophic substances, oxygen and glucose, and consequently, the brain suffers from synaptic dysfunction and neuronal degeneration/loss, leading to both gray and white matter atrophy. The magnetic resonance imaging of the head used in the North American studies from Kronos Early Estrogen Prevention Study (KEEPS) showed a brain volume decrease with an average of 0.30–0.35% per year, and an increase of 3.59–3.73% in the ventricular volumes in the first 18 months of menopause [57], with a regional reduction of volume, which is more important in the hippocampus [58].

There are two hypotheses regarding neurodegeneration in brain aging, connected to low energy fuel supply, glucose hypometabolism and its complications for normal functioning [59], and microglia activation with associated secondary effect. In these hypothetical conditions, there are sexually divergent differences in gene expression in aging brain with comparing the number of gene expression changes in both males and females, and separating gene expression profiles based on up or downregulation.

The first hypothesis regards to the deficiency in glucose availability and mitochondrial dysfunction well-known as hallmarks of brain aging, which are particularly accentuated in neurodegenerative disorders, and the shift from an aerobic glycolytic to a ketogenic phenotype of bioenergetic metabolism. The model on female rat brain aging revealed that bioenergetic decline is starting from perimenopausal transition, which is followed by the decrease of brain synaptic plasticity [39]. The mouse female transgenic model of familial AD revealed that ovariectomy induces a shift in fuel availability and metabolism in the hippocampus, with an increase of enzymes required for long-chain fatty acid and ketone body metabolism, to obtain brain energy [46, 60]. Glucose hypometabolism associated to cerebral hypoperfusion initiated with perimenopausal atherosclerosis [61], hypercholesterolemia, nitric oxide, and impairment of redox homeostasis is considered as the key pathophysiologic promoter of neurodegeneration [59], and the known differences in regional brain metabolism make some women prone to AD [62].

Posterior cingulated and prefrontal cortex, which closely resembles the hypometabolic profile of AD brains are the postmenopausal women’s brain areas with reduced cerebral blood flow, with alteration of brain blood barrier glucose transport, and with significant decline in glucose metabolism [63].

It was demonstrated that brain aging is associated with a decrease of central insulin concentration [64, 65, 66], with an impairment of insulin receptor binding ability, resulting in an increase in deterioration of glucose homeostasis in the brain. Brain insulin resistance [67] is associated to peripheral insulin resistance–a typical feature of elder ages, associated to atherogenic dyslipidemia [65], and ET influences insulin resistance in medial prefrontal gyrus metabolism.

The second hypothesis is focusing on neuroinflammation specifically after low estrogen levels, connected to the shift of microglia activation, with the changing rate of microglia after activation M1 (classical) to M2 (alternative) type [68] or it is a maladaptive microglia activation [69], or a shift from neuroprotection to neurotoxicity, underlining chronic neuroinflammation and parainflammation, which is different in women and men [70, 71]. The shift is connected to proinflammatory cytokines and oxidative-nitrosative stress, which plus elevated levels of complement pathway components and other immune factors plays a key pathophysiological role in promoting cognitive dysfunction by enhancing endothelin, Amyloid-β deposition, cerebral amyloid angiopathy, aberrant synapse elimination in the hippocampus [72], and blood-brain barrier disruption.

AD is characterized by the loss of neurons and synapses from the cerebral cortex and certain subcortical regions of the temporal and parietal lobes, and parts of the frontal cortex and cingulated gyrus [73], and accumulation of plaque made up of small peptides called β-amiloid (also written as A-beta or Aβ). β-amyloid is a fragment from a larger protein called amyloid precursor protein (APP), a transmembrane protein that penetrates through the neuron’s membrane. The Italian studies from Florence have demonstrated that estradiol is restoring in menopause the neuroprotective gene, seladin-1 (for SELective Alzheimer’s Disease INdicator-1), or the gene DHCR24, which is downregulated in AD [74]. This gene inhibits the activation of caspase-3, a key modulator of apoptosis, and the gene encodes 3β-hydroxysterol, which catalyzes the conversion of desmosterol into cholesterol, and an appropriate amount of membrane cholesterol plays a pivotal role to protect nerve cells against Aβ toxicity and counteracts the synthesis of Aβ in AD [75, 76].

Microglia, a type of glial cell derived from myeloid precursors in the bone marrow that populate the CNS during development, as well as a brain resident innate immune cell, is the first line of defense in the CNS, as a monitor/sensor of neuronal activity in normal brain [77], protecting the local environment against invading pathogens, helping recovery from injury, and also in synapse pruning and neurodevelopment [78]. It is crucial in clearing debris, apoptotic/necrotic cells, or products from necrotic cells, infiltration of infectious agents, mediating the brain’s inflammatory and repair response to traumatic injury, stroke, or neurodegeneration [79]. It was suggested that age-dependent and senescence-driven impairments of microglia functions and responses play essential roles during onset and progression of neurodegenerative diseases as AD and PD, in which molecular changes on microglia senescence are similar [80]. The unique nature and developmental origin of microglia causing microglial self-renewal and telomere shortening led to the hypothesis that these CNS-specific innate immune cells become senescent [81]. There are two important characteristics of human brain microglia: their heterogeneity observed in brain regions, and their different sensitivity to aging; the microglia from cortex, basal forebrain, and hippocampus are more sensible [81].

Microglia is activated from its normal state of a functionally “resting” resident immune cell of the CNS, and upon activation, microglia may proliferate and undergo a morphological transformation from a ramified to amoeboid appearance, and movement to sites of injury or stress can occur along with a release soluble immune mediators [82]. The activated microglia are functioning like a phagocyte or macrophage, having toll-like receptors (TLRs), that recognize specific molecular patterns as complement, mannose, scavenger, C-type lectin, nucleotide-binding oligomerization domain-like, and this specific action of microglia is called autophagy. The autophagy is crucial for neuronal health and survival, the delivery of toxic molecules and organelles from neuronal apoptotic cells to microglia lysosomes may be acutely and/or chronically dysregulated by senescence, affecting phagocytosis and inflammation-innate immune functions in all age-associated neurodegenerative diseases [83]. There are two phenotypes of activated microglia: M1 (the cells become more cytotoxic by releasing additional pro-inflammatory cytokines- TNF, Il-1β, Il-6, and free radicals [84]), and M2, which becomes more anti-inflammatory, by secreting anti-inflammatory cytokines and neurotrophic factors and helps repair local damage [82]. The mouse model on AD is showing a distinct shift in activated microglia phenotypes, that occurs between the beginning of Aβ pathology (alternative phenotype), and advanced stages (classical phenotype), the latter may cause disease-associated neuron loss.

In this context, there are comments/discussions on microglia: if it is a scapegoat, a saboteur, or something else. A multicenter research group has discovered the presence of microglia amylin receptors mediating Aβ inflammation and neurodegeneration on primary cultures of fetal human and rats microglia [84], these receptors being common to neurons and microglia. It was proposed a model of microglia activation for AD, and neuronal death, involving these receptors, microglia, neurons, inflammation, amyloid precursor protein, and Aβ (Figure 1).

Figure 1.

Model of neurodegeneration in AD proposed by Fu et al. [84]: Through the involvement microglia and neural amylin receptors in mediating the Aβ-induced neurodegeneration. Legend: The expression of amylin receptors of resting microglia, increased in response to inflammatory triggers like LPS, induces microglial cells activation. The interaction of Aβ with amylin receptors of the activated microglia leads to increased production and release of cytokines (TNF, Il-1β, and Il-6), which act directly on neurons to produce cell death and additionally augment the production of Aβ via processing of the amyloid precursor protein (APP). The Aβ, in turn, interacts with neuronal and microglial amylin receptors to produce cell death. Adapted from Fu et al. [84]. Open access to this article is distributed under the terms of the creative commons attribution 4.0 international license (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the creative commons license, and indicate if changes were made. The creative commons public domain dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

The amylin receptors are increasing as microglia responds to inflammatory triggers, such as lipopolysaccharide, resulting in microglia activation. The interaction of Aβ with amylin receptors of the activated microglia leads to increased production and release of cytokines, which act directly on neurons to produce cell death, with additionally increased production of Aβ via processing the amyloid precursor protein. The Aβ interacts with neurons and microglia amylin receptors to produce cell death [84].

The microglia activation is via the release of ATP, neurotransmitters, growth factors or cytokines, ion changes, special of Ca+2 in the CNS environment, or loss of inhibitor molecules displayed by healthy neurons, or when microglia cells encounter molecules not normally found in the healthy CNS, as blood clotting factors, intracellular constituents released by necrotic cells (hypomethylated mammalian DNA, RNA), externalized phosphatidylserine on apoptotic cells, immunoglobulin-antigen complexes, opsonizing complement, abnormally folded proteins or pathogen-related structures. When microglia activation occurs, the activation is correlated to the severity degree of the stressor, being recorded the disruptions of microglia functions causing synaptic dysfunction and excess synapse loss early in abnormalities of learning and memory [77].

Being a debate about the initiator from the two hypotheses: first, the bioenergetic hypothesis based on mitochondrial dysfunction, and the second on the microglia activation as the driving force for neuroinflammation, which is “a lesson learned from microglia depletion models” [85], there are multiple evidences that these abnormalities exacerbate each other, and these mechanistic diversities have cellular redox dysregulation as a common denominator and connector [86]. According to these, one may consider a metabolic inflammatory axis during brain aging and in neurodegenerative diseases [42]. In conditions of hypoglycemia, lactate can serve as an auxiliary fuel by metabolism of glycogen stores to generate glucose and subsequently lactate; some studies revealed that glial cells are likely to produce lactate in excess to its utilization by neurons [46].

Neurological symptoms that emerge during perimenopause are indicative of disruption in multiple estrogen-regulated systems, and affect multiple domains of cognitive and memory functions [87].

Estrogens (special E2) are appreciated as a master regulator of bioenergetic systems in the body and brain [88].

The hypothesis on estrogen action or “healthy-cell bias” hypothesis [87, 89] is similar to the understanding of the different cardiovascular protective/harmful effects of estrogens at different women ages—protective before 60 years and harmful after 65 years. The protective effect of E2 is altered in the presence of the APOE4 genotype, which alters the response of microglia and macrophages to 17β-E2 [90], and this fact may be an explanation of some studies showing better results with ET on memory recall in women aged around 70 and 20 years postmenopausal, if women have not demonstrated memory impairment [91].

It was demonstrated how during reproductive ages the estrogen-induced signaling pathways in hippocampal and cortical neurons converge upon the mitochondria to enhance aerobic glycolysis coupled to the citric acid cycle, mitochondrial respiration, and ATP generation, and in senescence when estrogens are missing, it is a chronic oxidative stress due to the shift from an aerobic glycolytic to a ketogenic profile/phenotype/ [35, 60], and this shift is preceded by the early, already mentioned decline in glucose transport and metabolism [46]. In mouse model, the mitochondrial bioenergetic deficit precedes AD [92]. The estrogen decline in perimenopause is associated to the decline in mitochondria bioenergetics and together with the shift to ketogenetic profile are steps to Aβ depositions in AD [93, 94]. Hexokinase, the first rate limiting step in glycolysis, interacts with mitochondria and prevents mitochondria-mediated apoptosis and through this mechanism, is promoting survival in neurons and other cell types [95], but AD patients exhibit declined hexokinase activity in the brain, cerebral microvessels, leukocytes, and fibroblasts.

Calcium dynamics play a pivotal and mandatory role in the estradiol-inducible cascade that leads to neurotrophic and neuroprotective benefit [89]. Dynamics of Ca2+ homeostasis are tightly regulated in healthy neurons and dysfunctional in degenerating neurons at elder ages.

The emergence of glucose hypometabolism, microglia activation, and impaired synaptic function in brain provide plausible mechanisms of neurological symptoms of perimenopause and can be predictive of later-life vulnerability to hypometabolic conditions such as AD. The alteration in the bioenergetic profile of the brain in the months/years of perimenopause may be an explanation for the controversies on estrogen therapy/hormone therapy divergent outcomes, beneficial [18, 19] or harmful (WHI Memory Study) effects on neural health, on memory and cognition [46].

References

  1. 1. McCarrey AC, An Y, Kitner-Triolo MH, Ferrucci L, Resnick SM. Sex differences in cognitive trajectories in clinically normal older adults. Psychology and Aging. 2016;31(2):166-175. DOI: 10.1037
  2. 2. Vina J, Lloret A. Why women have more Alzheimer’s disease than men: Gender and mitochondrial toxicity of amyloid-beta peptide. Journal of Alzheimer’s Disease. 2010;20(Suppl 2):S527-S533. DOI: 10.3233
  3. 3. Alzheimer's A. Alzheimer’s disease facts and figures. Alzheimer's & Dementia. 2013;9(2):208-245. DOI:10.1016
  4. 4. Appelros P, Stegmayr B, Terent A. Sex differences in stroke epidemiology: A systematic review. Stroke. 2009;40(4):1082-1090. DOI: 10.1161
  5. 5. Pike CJ. Sex and the development of Alzheimer's disease. Journal of Neuroscience Research. 2017 Jan 2;95(1-2):671-680. DOI: 10.1002
  6. 6. Prince M, Wimo A, Prina M, et al. Alzheimer's Disease International. World Alzheimer Report 2015: The Global Impact of Dementia: An Analysis of Prevalence, Incidence, Cost and Trend. https://www.alz.co.uk/research/world-report-2015
  7. 7. Marin R, Guerra B, Diaz M, et al. Estrogen activates classical and alternative mechanisms to orchestrate neuroprotection. Current Neurovascular Research. 2005 Oct;2(4):287-301
  8. 8. Arnold AP, Lusis AJ. Understanding the sexome: Measuring and reporting sex differences in gene systems. Endocrinology. 2012;153(6):2551-2555. DOI: 10.1210
  9. 9. Baron AM, Pike CJ. Sex hormones, aging, and Alzheimer's disease. Frontiers in Bioscience. 2012;4:976-997
  10. 10. Henderson VW. Cognition and cognitive aging. Climacteric. 2007 Oct;10(Suppl 2):88-91
  11. 11. Blacker D, Lee H, Muzikansky A, et al. Neuropsychological measures in normal individuals that predict subsequent cognitive decline. Archives of Neurology. 2007;64(6):862-871
  12. 12. Levey J, Lah J, Goldstein F, Steenland K, Bliwise D. Mild cognitive impairment: An opportunity to identify patients at high risk for progression to Alzheimer's disease. Clinical Therapeutics. 2006;28(7):991-1001
  13. 13. Gauthier S, Reisberg B, Zaudig M, Winblad B, et al. Mild cognitive impairment. Lancet 2006;367:1262-1270
  14. 14. Yan Y, Cheng L, Chen X, Ai J, et al. Estrogen deficiency is associated with hippocampal morphological remodeling of early postmenopausal mice. Oncotarget. 2017 Mar 28;8(13):21892-21902. DOI: 10.18632
  15. 15. Zhang Y, Champagne N, Beitel KL, Goodyer GC, Trifiro M, LeBlanc A. Estrogen and androgen protection of human neurons against intracellular amyloid toxicity through heat shock protein 70. The Journal of Neuroscience. 2004;24(23):5315-5321
  16. 16. Kohannim O, Hua X, Hibar DP, et al. Boosting power for clinical trials using classifiers based on multiple biomarkers. Neurobiology of Aging. 2010;31(8):1429-1442
  17. 17. Dye VR, Miller JK, Singer JE, Levine JA. Hormone replacement therapy and risk for neurodegenerative diseases. International Journal of Alzheimer's Disease. 2012:18. Jagannatha Rao KS, editor. Article ID 258454. DOI: doi.org/10.1155/2012/258454
  18. 18. Maki PM et al. Perimenopausal use of hormone therapy is associated with enhanced memory and hippocampal function later in life. Brain Research. 2011;1379:232-243
  19. 19. Maki PM. Minireview: Effects of different HT formulations on cognition. Endocrinology. 2012;153:3564-3570
  20. 20. Soules MR, Sherman S, Parrott E, Rebar R, Santoro N, Utian W, et al. Executive summary: Stages of reproductive aging workshop (STRAW). Fertility and Sterility. 2001;76(5):874-878
  21. 21. Shuster LT, Rhodes DJ, Rocca WA, et al. Premature menopause or early menopause: Long-term health consequences. Maturitas 2010;65:161-166
  22. 22. North American Menopause Society Menopause Practice–A Clinician’s Guide. 4. Cleveland, OH: North American menopause Society; 2010
  23. 23. van der Stege JG, Groen H, van Kasteren YM, et al. Decreased androgen concentrations and diminished general and sexual well-being in women with premature ovarian failure. Menopause. 2008;15:23-31
  24. 24. Burger HG, Dudley EC, Robertson DM, Dennerstein L. Hormonal changes in the menopause transition. Recent Progress in Hormone Research. 2002;57:257-275
  25. 25. Butler L, Santoro N. The reproductive endocrinology of the menopausal transition. Steroids. 2011 Jun;76(7):627-635. DOI: 10.1016
  26. 26. Davison SL, Bell R, Davis SR, et al. Androgen levels in adult females: Changes with age, menopause, and oophorectomy. The Journal of Clinical Endocrinology and Metabolism. 2005;90:3847-3853
  27. 27. Gore AC, Oung T, Woller MJ, et al. Neuroendocrine mechanisms for reproductive senescence in the female rat: Gonadotropin-releasing hormone neurons. Endocrine. 2000;13:315-323
  28. 28. Gore AC, Windsor-Engnell BM, Terasawa E. Menopausal increases in pulsatile gonadotropin-releasing hormone (GnRH) release in a non-human primate (Macaca mulatta). Endocrinology. 2004;145:4653-4659
  29. 29. Rocca AW, Miller MV, Brown DR, et al. Premature menopause or early menopause and risk of ischemic stroke. Menopause. 2012 Mar;19(3):271-277. DOI: 10.1097
  30. 30. Rivera CM, Grossardt BR, Rhodes DJ, et al. Increased cardiovascular mortality after early bilateral oophorectomy. Menopause. 2009;16:15-23
  31. 31. Nappi ER, Sinforiani E, Nappi G, et al. Memory functioning at menopause: Impact of age in ovariectomized women. Gynecologic and Obstetric Investigation 1999;47:29-36
  32. 32. Rocca WA, Grossardt BR, Melton LJ 3rd, et al. Survival patterns after oophorectomy in premenopausal women: A population-based cohort study. The Lancet Oncology. 2006;7:821-828
  33. 33. Baba Y, Ishikawa S, Kajii E, et al. Premature menopause is associated with increased risk of cerebral infarction in Japanese women. Menopause 2010;17:506-510
  34. 34. De Vos M, Devroey P, Fauser BC. Primary ovarian insufficiency. Lancet. 2010;376:911-921
  35. 35. Brinton DR, Yao J, Cadenas E, et al. Perimenopause as a neurological transition state. Nature Reviews Endocrinology. 2015;11:393-405. DOI:10.1038
  36. 36. Weber MT, Rubin LH, Maki PM. Cognition in perimenopause: The effect of transition stage. Menopause 2013;20:511-517
  37. 37. Genazzani AR, Bernardi F, Luisi M, et al. Endocrinology of menopausal transition and its brain implications. CNS Spectrums. 2005 Jun;10(6):449-457
  38. 38. Burger HG et al. Nomenclature and endocrinology of menopause and perimenopause. Expert Review of Neurotherapeutics. 2007;7(11 Suppl):S35-S43
  39. 39. Yin F, Yao J, Sancheti H, Cadenas E, Brinton RD, et al. The perimenopausal aging transition in the female rat brain: Decline in bioenergetic systems and synaptic plasticity. Neurobiology of Aging. 2015 Jul;36(7):2282-2295. DOI: 10.1016
  40. 40. Nilsen J, Irwin RW, Brinton RD, et al. Estradiol in vivo regulation of brain mitochondrial proteome. The Journal of Neuroscience. 2007 Dec 19;27(51):14069-14077. DOI: 10.1523
  41. 41. Mouton PR, Long JM, Ingram DK, et al. Age and gender effects on microglia and astrocyte numbers in brains of mice. Brain Research. 2002;956(1):30-35. DOI: 10.1016
  42. 42. Yin F, Sancheti H, Patil I, Cadenas E. Energy metabolism and inflammation in brain aging and Alzheimer’s disease. Free Radical Biology and Medicine. 2016 Nov;100:108-122. DOI: 10.1016
  43. 43. Trabzuni D, Ramasamy A, Ryten M, et al. North American Brain Expression C Widespread sex differences in gene expression and splicing in the adult human brain. Nature Communications. 2013;4:2771. DOI: 10.1038
  44. 44. Tronson NC, Collette KM. (Putative) sex differences in neuroimmune modulation of memory. Journal of Neuroscience Research. 2017 Jan 2;95(1-2):472-486. DOI: 10.1002
  45. 45. Zhang Z. Gender differentials in cognitive impairment and decline of the oldest old in China. Journals of Gerontology. Series B, Psychological Sciences and Social Sciences. 2006;61(2):S107-S115. DOI: 10.1093
  46. 46. Ding F, Yao J, Brinton RD, et al. Early decline in glucose transport and metabolism precedes shift to ketogenic system in female aging and Alzheimer's mouse brain: Implication for bioenergetic intervention. PLoS One. 2013 Nov 11;8(11):e79977. DOI: 10.1371
  47. 47. Holtman IR, Raj DD, Kamphuis W, et al. Induction of a common microglia gene expression signature by aging and neurodegenerative conditions: A co-expression meta-analysis. Acta Neuropathologica Communications. 2015;3:31. DOI: 10.1186
  48. 48. Ziv Y, Ron N, Schwartz M, et al. Immune cells contribute to the maintenance of neurogenesis and spatial learning abilities in adulthood. Nature Neuroscience. 2006;9(2):268-275. DOI: 10.1038
  49. 49. Hickman SE, Kingery ND, El Khoury J, et al. The microglial sensome revealed by direct RNA sequencing. Nature Neuroscience. 2013;16(12):1896-1905. DOI: 10.1038
  50. 50. Mangold AC, Wronowski B, Freeman MW, et al. Sexually divergent induction of microglial-associated neuroinflammation with hippocampal aging. Journal of Neuroinflammation. 2017;14:141. DOI: 10.1186
  51. 51. Gebara E, Sultan S, Kocher-Braissant J, Toni N. Adult hippocampal neurogenesis inversely correlates with microglia in conditions of voluntary running and aging. Frontiers in Neuroscience 2013;7:145. DOI: 10.3389
  52. 52. Ables JL, Decarolis NA, Eisch AJ, et al. Notch1 is required for maintenance of the reservoir of adult hippocampal stem cells. The Journal of Neuroscience 2010;30(31):10484-10492. DOI: 10.1523
  53. 53. Gadadhar A, Marr R, Lazarov O. Presenilin-1 regulates neural progenitor cell differentiation in the adult brain. The Journal of Neuroscience. 2011;31(7):2615-2623. DOI: 10.1523
  54. 54. Bekris LM, Yu CE, Bird TD, Tsuang DW. Genetics of Alzheimer disease. Journal of Geriatric Psychiatry and Neurology . 2010;23(4):213-227. DOI: 10.1177
  55. 55. Zhang B, Gaiteri C, Bodea LG, Dobrin R, et al. Integrated systems approach identifies genetic nodes and networks in late-onset Alzheimer’s disease. Cell. 2013;153(3):707-720. DOI: 10.1016
  56. 56. Ma J, Jiang T, Tan L, Yu JT. TYROBP in Alzheimer’s disease. Molecular Neurobiology. 2015;51(2):820-826. DOI: 10.1007
  57. 57. Kantarci K, Miller VM, Jack Jr CR, et al. Brain Volume Changes in Recently Menopausal Women in a Hormone Replacement Trial.: International Menopause Society Meeting. May 21, 2008. Madrid, Spain
  58. 58. Raz N, Lindenberger U, Acker JD, et al. Regional brain changes in aging healthy adults: General trends, individual differences and modifiers. Cereberal Cortex. 2005;15(11):1676-1689. DOI: 10.1093
  59. 59. Daulatzai MA. Cerebral hypoperfusion and glucose hypometabolism: Key pathophysiological modulators promote neurodegeneration, cognitive impairment, and Alzheimer's disease. Journal of Neuroscience Research. 2017 Apr;95(4):943-972. DOI: 10.1002
  60. 60. Yao J, Hamilton RT, Cadenas E, Brinton RD. Decline in mitochondrial bioenergetics and shift to ketogenic profile in brain during reproductive senescence. Biochimica et Biophysica Acta. 2010 Oct;1800(10):1121-1126. DOI: 10.1016
  61. 61. Lerner DJ, Kannel WB. Patterns of coronary heart disease morbidity and mortality in the sexes. A 26-year follow up of the Framingham population. American Heart Journal. 1986;111:383-390
  62. 62. Silverman DHS, Geist CL, Kenna HA, et al. Differences in regional brain metabolism associated with specific formulations of hormone therapy in postmenopausal women at risk for AD. Psychoneuroendocrinology. 2010;36:502-513
  63. 63. Zhao L, Mao Z, Woody SK, Brinton RD. Sex differences in metabolic aging of the brain: Insights into female susceptibility to Alzheimer’s disease. Neurobiology of Aging. 2016;42:69-79. DOI: 10.1016
  64. 64. Rasgon NL, et al. Insulin resistance and hippocampal volume in women at risk for Alzheimer's disease. Neurobiology of Aging. 2011;32:1942-1948
  65. 65. Rasgon NL, et al. Insulin resistance and medial prefrontal gyrus metabolism in women receiving hormone therapy. Psychiatry Research. 2014;223:28-36
  66. 66. Baranowska – Bik A, Bik W. Insulin and brain aging. Przeglad Menopauzalny. 2017 Jun;16(2):44-46. DOI: 10.5114
  67. 67. Kullmann S, Heni M, Haring HU, et al. Brain insulin resistance at the crossroads of metabolic and cognitive disorders in humans. Physiological Reviews. 2016 Oct;96(4):1169-1209. DOI: 10.1152
  68. 68. Nikodemova M, Small AL, Kimyon RS, Watters JJ. Age-dependent differences in microglial responses to systemic inflammation are evident as early as middle age. Physiological Genomics. 2016;48(5):336-344. DOI: 10.1152/physiolgenomics.00129.2015
  69. 69. Hume DA. The many alternative faces of macrophage activation. Frontiers in Immunology. 2015;6:370. DOI: 10.3389/fimmu.2015.00370
  70. 70. Mirza MA, Ritzel R, Liu F, et al. Sexually dimorphic outcomes and inflammatory responses in hypoxic-ischemic encephalopathy. Journal of Neuroinflammation. 2015;12:32. DOI: 10.1186/s12974-015-0251-6
  71. 71. Lenz KM, McCarthy MM. A starring role for microglia in brain sex differences. The Neuroscientist. 2015;21(3):306-321. DOI: 10.1177/1073858414536468
  72. 72. Hong S, Beja-Glasser VF, Nfonoyim BM, Barres BA, et al. Complement and microglia mediate early synapse loss in Alzheimer mouse models. Science 2016;352(6286):712-716. DOI: 10.1126/science.aad8373
  73. 73. Wenk GL. Neuropathologic changes in Alzheimer's disease. The Journal of Clinical Psychiatry. 2003;64(Suppl 9):7-10
  74. 74. Luciani P, Deledda C, Peri A, et al. Seladin-1 is a fundamental mediator of the neuroprotective effects of estrogen in human neuroblast long-term cell cultures. Endocrinology 2008 Sep;149(9):4256-4266. DOI: 10.1210/en.2007-1795
  75. 75. Peri A, Benvenuti S, Cellai I, et al. Membrane cholesterol as a mediator of the neuroprotective effects of estrogens. Neuroscience. 2011;191:107-117. DOI: 10.1016
  76. 76. Peri A. Neuroprotective effects of estrogens: The role of cholesterol. Journal of Endocrinological Investigation. 2016 Jan;39(1):11-18. DOI: 10.1007/s40618-015-0332-5
  77. 77. Morris GP, Clark IA, Zinn R, Vissel B. Microglia: A new frontier for synaptic plasticity, learning and memory, and neurodegenerative disease research. Neurobiology of Learning and Memory. 2013 Oct;105:40-53. DOI: 10.1016/j.nlm.2013.07.002
  78. 78. Conde JR, Streit WJ. Microglia in the aging brain. Journal of Neuropathology and Experimental Neurology. 2006;65(3):199-203. DOI: 10.1097/01.jnen.0000202887.22082.63
  79. 79. Lucin KM, Wyss-Coray T. Immune activation in brain aging and neurodegeneration: Too much or too little? Neuron. 2009 Oct 15;64(1):110-122. DOI: 10.1016
  80. 80. Spittau B. Aging microglia-phenotypes, functions and implications for age-related neurodegenerative diseases. Frontiers in Aging Neuroscience. 2017 Jun 14;9:194. DOI: 10.3389/fnagi.2017.00194
  81. 81. Grabert K, Michoel T, Karavolos MH, McColl BW, et al. Microglial brain region-dependent diversity and selective regional sensitivities to aging. Nature Neuroscience 2016;19(3):504-516. DOI: 10.1038/nn.4222
  82. 82. Ekdahl CT. Microglial activation—Tuning and pruning adult neurogenesis. Frontiers in Pharmacology. 2012;3:41. DOI: 10.3389/fphar.2012.00041
  83. 83. Plaza-Zabala A, Sirra-Torre V, Sierra A. Autophagy and microglia: Novel partners in neurodegeneration and aging. International Journal of Molecular Sciences. 2017 Mar 9;18(3). pii: E598. DOI: 10.3390/ijms18030598
  84. 84. Fu W, Vukojevic V, Patel A, Jhamandas J, et al. Role of microglial amylin receptors in mediating beta amyloid (Aβ)-induced inflammation. Journal of Neuroinflammation, 2017 Oct 6;14(1):199. DOI: 10.1186/s12974-017-0972-9
  85. 85. Spangenberg EE, Green KN. Inflammation in Alzheimer's disease: Lessons learned from microglia-depletion models. Brain, Behavior, and Immunity 2017 Mar;61:1-11. DOI: 10.1016/j.bbi.2016.07.003
  86. 86. Uchoa MF, Moser VA, Pike CJ. Interactions between inflammation, sex steroids, and Alzheimer's disease risk factors. Frontiers in Neuroendocrinology. 2016 Oct;43:60-82. DOI: 10.1016/j.yfrne.2016.09.001
  87. 87. Brinton RD. Investigative models for determining hormone therapy-induced outcomes in brain: Evidence in support of a healthy cell bias of estrogen action. Annals of the New York Academy of Sciences. 2005;1052:57-74
  88. 88. Rettberg JR, Yao J, Brinton RD. Estrogen: A master regulator of bioenergetic systems in the brain and body. Frontiers in Neuroendocrinology. 2014 Jan;35(1):8-30. DOI: 10.1016
  89. 89. Chen S, Nilsen J, Brinton RD. Dose and temporal pattern of estrogen exposure determines neuroprotective outcome in hippocampal neurons: Therapeutic implications. Endocrinology. 2006;147(11):5303-5313. DOI: 10.1210/en.2006-049
  90. 90. Brown CM, Colton CA, et al. The APOE4 genotype alters the response of microglia and macrophages to 17β-estradiol. Neurobiology of Aging. 2008;29(12):1783-1794
  91. 91. Dumas J, Hancur-Bucci C, Newhouse P, et al. Estradiol interacts with the cholinergic system to affect verbal memory in postmenopausal women: Evidence for the critical period hypothesis. Hormones and Behavior. 2008;53(1):159-169
  92. 92. Yao J, Irwin RW, Brinton RD, et al. Mitochondrial bioenergetic deficit precedes Alzheimer's pathology in female mouse model of Alzheimer's disease. Proceedings of National Academy of Sciences of United States of America. 2009;106:14670-14675
  93. 93. Yao J, Rettberg JR, Brinton RD, et al. Shift in brain metabolism in late onset Alzheimer's disease: Implications for biomarkers and therapeutic interventions. Molecular Aspects of Medicine. 2011;32:247-257
  94. 94. Yao J, Irwin R, Brinton RD, et al. Ovarian hormone loss induces bioenergetic deficits and mitochondrial beta-amyloid. Neurobiology of Aging. 2012;33:1507-1521
  95. 95. Gimenez-Cassina A, Lim F, Diaz-Nido J, et al. Mitochondrial hexokinase II promotes neuronal survival and acts downstream of glycogen synthase kinase-3. The Journal of Biological Chemistry. 2009;284:3001-3011

Written By

Manuela Cristina Russu and Alexandra Cristina Antonescu

Submitted: 07 August 2017 Reviewed: 24 January 2018 Published: 21 February 2018