Skip to main content
American Journal of Neurodegenerative Disease logoLink to American Journal of Neurodegenerative Disease
. 2012 Nov 18;1(3):266–279.

Interactions between oestrogen and the renin angiotensin system - potential mechanisms for gender differences in Alzheimer’s disease

Thomas Simon O’Hagan 1, Whitney Wharton 2,3,4, Patrick Gavin Kehoe 1
PMCID: PMC3560469  PMID: 23383397

Abstract

Interactions between oestrogen and the renin angiotensin system (RAS) are reviewed and explored from the perspective where these interactions may modulate risk of developing Alzheimer’s disease (AD). AD is more prevalent in women than men, partly attributed to women’s increased life expectancy; however underlying vascular differences may also contribute to AD risk. The RAS is a key regulator of blood pressure (BP). Pharmacological inhibition of angiotensin converting enzyme (ACE) and blockade of angiotensin II type 1 receptors (AT1R) are widely used to treat hypertension. Variation in components of the RAS such as ACE, neprilysin (NEP) and AT1R have been reported in AD, some of which may also directly alter AD neuropathology with changes in amyloid beta (Aβ) levels, cognitive decline and neuroinflammation. Recently, RAS inhibiting drugs have been shown to attenuate the incidence, progression and pathology of AD. Oestrogen is also thought to prevent hypertension by reducing the vasoconstrictive actions of the RAS. Reduced oestrogen levels in women during the menopausal transition may therefore increase their risk of hypertension and/or RAS-mediated changes to cerebrovascular or AD pathology. Specifically, oestrogen prevents the production and action of angiotensin II (Ang II), thought to exert harmful effects of the RAS in both hypertension and AD, while also potentially facilitating RAS-mediated Aβ degradation. These oestrogen-RAS interactions may partly explain current conflicting findings regarding oestrogen depletion and hormone therapy with respect to AD risk. Clinical trials targeting either the RAS or oestrogen systems for AD prevention and treatment should perhaps give closer attention to key biochemical components of these pathways as potential confounders to primary and secondary outcome measures.

Keywords: Oestrogen, renin angiotensin system (RAS), gender difference, Alzheimer’s disease

Introduction

Alzheimer’s disease (AD) is the most common form of dementia [1]. An estimated 26.6 million people worldwide have AD, with numbers expected to quadruple by 2050 [2]. Midlife vascular risk factors including hypercholesterolemia and hypertension increase the risk of AD in later life. Anti-hypertensive drugs show promise in helping prevent or treat AD [1].

This paper reviews how oestrogen levels fluctuate and decline throughout a woman’s life, focussing on how these changes relate to the the renin angiotensin system (RAS) and the impact of reversing oestrogen depletion via hormone therapy (HT). The RAS is implicated in AD, and and hence post-menopausal oestrogen depletion may contribute to RAS-mediated gender differences in AD development. Inhibition of RAS and oestrogen pathways will be discussed and the resultant implications for future treatment of AD.

The amyloid component of AD

The peptide amyloid beta (Aβ) is formed from sequential cleavage of the amyloid precursor protein (APP) by β- and γ- secretases [3]. The most common isoforms of Aβ are Aβ1-40 and Aβ1-42 (the numbers refer to the number of constitutive amino acids) [3]. The accumulation of Aβ in late onset AD is likely due to Aβ clearance dysfunction rather than Aβ overproduction.

Throughout life, soluble Aβ levels decrease and insoluble Aβ levels increase. In AD, both soluble, oligomeric and insoluble Aβ levels increase, particularly the Aβ1-42 variant, which leads to the formation of AD senile plaques [4]. Aβ (mainly Aβ1-42) is also toxic to neuronal cells, partly due to induction of reactive oxygen species and inflammation as well as apoptosis of neurones via tumour necrosis factor (TNF) type I receptor signalling and intracellular calcium level disruption [5,6].

Enhanced β-secretase activity on APP gives rise to increased insoluble and decreased soluble Aβ formation which has been shown in AD [4]. Yet numerous enzymes including neprilysin (NEP) and ACE degrade Aβ (Figure 1). Changes to the activities of these Aβ degrading enzymes (ADEs) are likely to alter the pathology of AD [7].

Figure 1.

Figure 1

Formation of Aβ from APP. Aβ formation is determined by α-, β- or γ- secretase cleavage of APP. Equilibrium throughout life is disrupted in AD, with increased β- secretase processing. Aβ is initially produced as a soluble monomer, but can combine with other monomers to form multimeric complexes of varying solubility before forming insoluble fibrils. Sites of secretase and ACE cleavage of Aβ are shown. Taken from [3].

The Renin Angiotensin System (RAS)

The RAS (see Figure 2) is best known for maintaining blood pressure (BP) through water and electrolyte balance, and preservation of vascular tone [8]. Vasoconstrictive properties of RAS are mediated by Ang II acting on the angiotensin type 1 receptor (AT1R) and angiotensin type 2 receptor (AT2R). Ang II binding to AT1R promotes vasoconstrictive (i.e. pro-hypertensive) responses, which AT2R (vasodilatory) binding counters [9]. The AT1R is also important in cell proliferation, hypertrophy, generation of oxidative radicals and inflammation [9,10]. The long-standing view of a single circulatory RAS (cRAS) has more recently been replaced by acceptance of numerous organ based systems, including the brain RAS [9].

Figure 2.

Figure 2

Angiotensins in the RAS. (A) Schematic of the RAS pathway. In the vasoconstrictive arm, Ang I is cleaved from angiotensinogen (AGT) by renin. Ang II is produced from Ang I by ACE. Ang II acts as a vasoconstrictor while ACE-2 cleavage of AGT and in turn the eventual production of Ang (1-7) works to vasodilate and oppose the actions of Ang II. Ang=angiotensin; AP-A=aminopeptidase-A; AP-N=aminopeptidase-N; DAP=dipeptidyl aminopeptidase. Not all intermediary metabolites and enzymes are shown. (B) Schematic of angiotensins (including their amino acid composition) derived from the cleavage of the decapeptide Ang I at various amino acid positions. Adapted from [80,86].

Angiotensins, like Aβ, may affect memory formation. Both Ang II and angiotensin III (Ang III) inhibit long term potentiation (LTP), however angiotensin IV (Ang IV) enhances LTP [11]. A role of Ang IV in memory formation and recall is also supported by high densities of the Ang IV receptor (also known as insulin-regulated aminopeptidase (IRAP)) in the hippocampus, amygdala and prefrontal cortex [12]. IRAP agonists improve spatial learning, whereas antagonists cause noticeable cognitive deficits [13].

Oestrogen levels fluctuate throughout life

Multiple factors contribute to the increased incidence of AD in women, including longevity and oestrogen decline during the peri- and post-menopausal transition [14,15]. With increasing life expectancy, a third of women’s lives are now spent in a post-menopausal, low oestrogen state [16].

Prior to menopause oestradiol (E2) is the predominant circulating oestrogen (Figure 3). At menopause, E2 levels fall precipitously to approximately 10% of premenopausal levels, while levels of oestrone (E1) decline to a lesser extent [17]. E1 and E2 continue to be synthesized by peripheral conversion of adrenal androstenedione and aromatization of testosterone, causing a shift in the E1/E2 ratio with E1 becoming the predominant oestrogen post-menopause [18].

Figure 3.

Figure 3

Monthly fluctuations of endogenous oestrogen levels. Oestrogenic fluctuations across the menstrual cycle and the post-menopausal state are shown such that E1 becomes the predominant oestrogen post- menopause.

Oestrogen and the RAS

Hypertension is more prevalent in men than in women pre-menopause, but this pattern changes during the menopausal transition [19,20]. This is partly due to the ability of oestrogens to reduce hypertensive effects of the RAS [21,22].

Effects on RAS-mediated vasoconstriction

Oestrogen affects AGT gene expression and synthesis through modulation of regulatory elements in the gene promoter [23,24]. This increases the concentrations of AGT found in women [25]. Promotion of AGT is countered by a number of downstream mechanisms to prevent hypertensive effects.

Conflicting evidence surrounds oestrogen and renin. Pre-menopausal women exhibit lower renin levels than both their post-menopausal counterparts and men [21]. While oestrogen can interfere with regulatory elements in the renin gene promoter in a similar fashion to AGT, the gene expression of renin may be controlled to a larger extent by β-adrenergic stimulated rises in cyclic adenosine monophosphate (cAMP) [21,26]. Oestrogen decreases circulating adrenaline and noradrenaline, which may therefore indirectly decrease plasma renin concentrations [21].

E2 attenuated ACE activity in vivo [22]. In humans, age-related decreases in ACE activity in girls between 8 and 18 years have been reported, possibly caused by inhibition of ACE mRNA [27].

The net effect of E2 on Ang II is unclear [28,29]. While AGT levels may increase, renin and ACE activities are reduced by oestrogen. The rate of Ang II synthesis may be dependent on the extent of renin and ACE inhibition over the rise in AGT.

Oestrogen also downregulates AT1R gene expression in rats [30-32]. Additionally, oestrogen sensitive binding proteins are thought to post-transcriptionally regulate AT1R mRNA [33]. These two mechanisms allow oestrogen to attenuate These two mechanisms allow oestrogen to attenuate AT1R responses to Ang II. Indeed, the vasoconstrictor response in the aortic rings and mesenteric vessels to Ang II is weaker in women [32].

Effects on RAS-mediated vasodilation

AT2R and ACE-2 have been mapped to the X chromosome, providing scope for gender differences [34,35]. AT2R mRNA expression is raised during oestrous of spontaneously hypertensive rats (SHRs) and increased ACE-2 expression in response to oestrogen is suggested [32]. Oestrogen increases circulating Ang (1-7), with female SHRs particularly sensitive to its vasodilatory effects [19,36]. Oestrogen may augment the effect of Ang (1-7) further by facilitating nitric oxide (NO) and prostaglandin release [19,36]. NEP protein level and activity is also substantially elevated in female hypertensive and control models, suggesting preferential NEP derived Ang (1-7) effects may occur in women [28]. Although beyond the scope of this paper, it is noteworthy this picture is complicated with the crossover of the RAS and the kallikrein-kinin system, with NEP able to degrade the vasodilator bradykinin [37]. However, additional evidence suggests that oestrogen may serve to restore any bradykinin-related changes to vascular tone resulting from upregulation of NEP by enhancing the vasodilatory effect of bradykinin [38].

Thus, while oestrogen increases AGT synthesis, the attenuation of renin and ACE results in an overall downregulation of the RAS. Although there remains ambiguous data on Ang II levels, its vasoconstrictive actions are reduced due to oestrogen-associated decreases in AT1R and increases in AT2R production. Further vasodilatory effects of oestrogen on the RAS may derive from NEP increases with downstream Ang (1-7) action but it is noteworthy there are possible additional bradykinin-related changes to the vasculature.

Oestrogen throughout life

Menstrual cycle

The menstrual cycle is characterized by cyclic changes in oestrogen which peak during the luteal phase and decline in the follicular phase [39]. In rats, brain AGT secretion correlates with oestrogen fluctuations in the ovarian and peripheral plasma [40]. Despite the activation of some RAS components in the luteal phase, increases in plasma Ang II and increase in BP are not apparent [41,42]. Instead there is decreased peripheral vascular resistance, suggesting a reduced response to Ang II, which may reflect oestrogen-mediated effects on ACE activity, AT1R and AT2R expression, as well as augmenting NEP and its associated vasodilatory responses. Thus oestrogen can contribute to regulatation and fine tuning of BP via specific actions on mediators of the RAS and their receptors. This well evolved system maintains homeostasis throughout the reproductive life of a woman.

Menopause

The loss of oestrogens during menopause is associated with increasing BP such that elderly women are more likely to be hypertensive than age matched men [20,43]. Animal models suggest that post-menopause, the behaviour of the RAS is opposite to that seen pre-menopause.

Effects on RAS-mediated vasoconstriction

Ovariectomised rats have lower AGT in hypothalamic and thalamic regions [40]. This should contribute to an overall lowering of BP, but increased BP is seen which may be partly due to increased sympathetic drive and renin release from postmenopausal weight gain [44]. Downstream feedback mechanisms in the RAS, such as increased ACE activity and binding densities could also be involved [45].

Despite increased BP in menopause, plasma Ang II levels appear to be paradoxically reduced [44]. This may be explained by an alteration in the AT1R:AT2R ratio, promoting AT1R binding. Indeed, AT1R binding density post ovariectomy is increased with subsequent enhanced vasoconstriction [32,45].

Data from human studies support in vivo findings. In post-menopausal women, AGT levels are significantly lower due to reduced oestrogen-mediated modulation of the AGT promoter [25]. Renin levels are also significantly lower in HT users and men, suggesting oestrogen loss raises renin levels and promotes vasoconstriction [21].

Effects on RAS-mediated vasodilation

Post-menopausal oestrogen loss causes reduced AT2R with an associated raised risk of hypertension [32,46]. There is also RAS-mediated downregulation of vasodilators including atrial natriuretic peptide (ANP) and NO [44].

The vasodilatory arm of the RAS is generally suppressed with loss of oestrogens decreasing ACE-2 activity and protein expression in hypertensive rats [47]. The decrease in ACE-2 protein is likely due to a loss of oestrogen-mediated transcription of ACE-2 [47].

Overall, the menopausal decline in oestrogen counters vasodilatory effects seen in the RAS pre-menopause. Although there is decreased AGT synthesis, this is countered by increases in: renin levels; ACE activity; vasoconstrictive effects of Ang II via increased AT1R and decreased AT2R expression; as well as reduced vasodilatory responses through decreased ACE-2 activity and protein expression.

Hormone therapy

Hormone therapy (HT) is used to reduce symptoms associated with decreases in E2 such as osteoporosis, coronary heart disease, hot flushes and mood disturbances [16,48]. Some evidence suggests that HT in women reduces AD risk [49]. After menopause, the predominant form of oestrogen is E1; thus, for HT to mimic premenopausal physiology, it is necessary to administer E2. However, many clinical studies evaluating the therapeutic efficacy of HT in healthy older women, (i.e. the Women’s Health Initiative), used conjugated equine oestrogen (CEE), a preparation rich in E1. In general, studies utilising E2 formulations of HT, particularly when administered during the menopausal transition, have shown a beneficial cognitive effect in comparison to studies utilising E1 preparations [50].

Effects on RAS-mediated vasoconstriction

Oral HT induces a hepatic first pass effect-mediated increase in plasma AGT [51].

Transdermal oestrogen increases total blood renin, probably at the level of gene expression [26,52]. However this increase in total blood renin is not accompanied by increases in active renin or plasma renin activity [52]. Suppression of β-adrenergic activity is a potential explanation for this discrepancy [21,52]. Increases in Ang II may further attenuate renin secretion, acting as negative feedback on the RAS [53]. Thus, oral HT stimulates the renin substrate AGT, while transdermal and oral HT downregulate the conversion of AGT to Ang I by inhibiting the rate-limiting step of the system.

Animal studies show that high E2 concentrations reverse and prevent the effects of ovariectomy on ACE activity in the periphery and heart [19,45,54]. Oestrogen therapy also reduced ACE gene expression, and therefore decreased ACE activity [22]. Indeed, in monkeys, oral oestrogen increases Ang I, but not Ang II [54].

Oestrogen alters the balance of AT1R:AT2R, lowering AT1R expression in vascular smooth muscle cells, the hypothalamus, adrenal cortex, and kidney [31,53,55]. Oestrogen also lowers AT1R binding [45,56]. Oestrogen may directly interfere with AT1R gene transcription via modulation of oestrogen response elements and activator protein-1 (AP-1) [31,32]. Oestrogen-mediated increases in NO is another proposed mechanism to downregulate AT1R expression [57,58].

Effects on RAS-mediated vasodilation

In female mice undergoing HT, AT2R gene expression and binding is upregulated [32,46,59]. The mechanisms of action of oestrogen on the AT2R are equivocal – there is no oestrogen response element on the AT2R promoter so altered expression may be via influences on AP-1, or by inhibition of Na+, K+-ATPase [46].

Oestrogen administration increases Ang (1-7) in transgenic animals [19]. Promoting Ang (1-7) lowers BP by preventing the retention of sodium ions, thus countering the effects of Ang II. Oestrogen increases Ang (1-7) relative to Ang II, possibly by inhibiting ACE activity [19]. It is also possible Ang (1-7) levels increase due to oestrogen effects on ACE-2.

In animal models of oestrogen replacement there were increases in ACE-2 and NEP activity [47]. However, in the renal cortex of mRen(2). Lewis rat models of hypertension, as well as in normotensive Lewis controls, ACE-2 activity was higher in males [28]. Conversely, Ang (1-7) is significantly higher in females, with amplified depressor effects in the presence of oestrogen [19,28]. Changes to Ang (1-7) levels likely reflect subtle shifts between ACE, NEP and ACE-2.

Oral and transdermal HT decrease systolic and diastolic BP, which is likely explained by the effects of oestrogen outlined above [43]. The effects of HT largely mimic those of endogenous oestrogen. Discrepant findings are likely due to methodological differences, such differential HT formulation and administration [51]. HT increases plasma AGT synthesis with oral administration, however there is reduced plasma renin activity due to indirect inhibition of β-adrenergic activity and ACE activity. There is limited evidence of an effect of HT on Ang II, however reported decreases in AT1R and increases in A2TR expression and binding have a net effect of reducing vasoconstriction. Conversely, vasodilation is upregulated via increases in Ang (1-7), possibly due to raised ACE-2 or NEP activity or inhibition of ACE. The Kronos Early Estrogen Prevention Study (KEEPS) however, did not show a significant relationship between blood pressure and either CEE or transdermal E2, in recently menopausal women over the 4 year clinical trial [60]. Additional follow up data is required to elicit longer term effects which may support animal models.

Relevance to AD

The brain RAS and communication with the cRAS

The brain RAS contains molecules present in the cRAS including AGT, renin, ACE and Ang II. The brain RAS regulates hormone release, central and peripheral BP, water and sodium levels and sympathetic nerve activity [61].

The cRAS and brain RAS communicate possibly by AT1R activation or via the circumventricular organs [61,62]. AGT is synthesised by astrocytes and its production may be enhanced by increased systemic Ang II binding to the AT1R on brain vessels [63]. This is supported by observed reductions in AGT, ACE mRNA expression and Ang II in response to Angiotensin Receptor Blocker (ARB) treatment. The cRAS may also influence the gene expression of the brain RAS [64].

Hypertension and Aβ

Hypertension is a major risk factor for cognitive dysfunction and subsequent AD [65]. Reduced cerebral blood flow is one explanation where hypoxia can directly affect β-secretase regulation, raising Aβ1-40 and Aβ1-42 levels and possibly promoting tangle formation [66,67].

Ang II, the blood brain barrier (BBB) and inflammation

Sustained hypertension can disrupt the normal separation of molecules from the vasculature and brain. Ang II may directly cause BBB leakage via AT1R and promote the entry of immune cells to the brain, activating resident microglia and astrocytes, features common to AD [68-70]. BBB dysregulation also releases cytokines such as TNF-α and interleukin-6 (IL-6), enhancing inflammatory processes resembling AD [71].

A secondary role of Ang II is as a proinflammatory mediator of increased vascular permeability, ROS formation, recruitment of inflammatory molecules and tissue repair remodelling [69,72]. Ang II causes upregulation of NF-κB, a transcription factor that maintains an immune response via pro-inflammatory cytokine release and adhesion molecules such as VCAM-1 and ICAM-1 [73,74].

Since oestrogen downregulates AT1R expression, thereby attenuating the response to Ang II, menopausal oestrogen loss may exacerbate BBB permeability and thus be a potential risk factor for AD [30-33,45].

Anti-hypertensive drugs, ACE and AD

Many large-scale studies have shown that antihypertensive drugs reduce dementia incidence and progression [75-77]. Beyond cardiovascular benefits, there is now a theoretical BP-independent mechanism of cognitive improvement involving blocking the formation of Ang II, thereby reducing inflammatory responses and maintaining BBB integrity [78,79].

Ang II-targeting anti-hypertensives, ACE-inhibitors (ACE-Is) as well as ARBs, may also counter the extent to which Ang II inhibits acetylcholine (ACh) release [80,81].

For ACE-Is, cognitive benefits are dependent on whether they cross the BBB, for example captopril and perindopril (centrally acting drugs). Non-centrally acting drugs such as enalapril and imidapril have shown no effect and in some instances, may increase the risk of dementia compared to participants not taking antihypertensives [78,82]. However for ACE-Is, there may be a caveat. Increased ACE levels have been correlated with decreased Aβ aggregation, fibril formation, deposition and cell survival [83]. Thus ACE-I mediated benefits on cognition via inhibition of Ang II may coincide with adverse elevations of Aβ. As yet, definitive evidence of ACE mediated degradation of Aβ is lacking, although recent findings show how long term exposure of people to ARBs has a very positive effect on AD-related pathology [84]. Incidentally, women who have lower (gene-variant associated) circulating ACE levels are at increased risk of developing AD [85].

There are possible explanations for these unresolved discrepancies. Firstly, centrally acting drugs may reduce brain Ang II and hence increase ACh release [86]. Secondly, central inhibition of Ang II increases cerebral perfusion [87]. Thirdly, inhibition of Ang II formation by ACE-Is raises Ang I, potentially favouring increased production of the vasodilators Ang (1-9), Ang (1-7) and Angiotensin (3-7) (Ang (3-7)) [88]. Ang (3-7) activates the AT4 receptor, which potentiates potassium-mediated ACh release and hence improves cognition [88]. Fourthly, there are increased concentrations of ACE, and Ang II and AT1R in the parietal cortex of AD brain [89]. ACE upregulation may thus act as a compensatory response to control Aβ levels which ACE-Is (but not ARBs) could interfere with [90,91]. Hence, the menopausal loss of oestrogen and subsequence increase of ACE activity may predispose women to cognitive decline in later life [45].

ARBs therefore may have a lot to offer in AD. Not only do they reduce BBB permeability, they also prevent the potentially harmful effects of angiotensin II by selectively targeting AT1R and do not interfere with ACE [92].

The effect of ARBs on cognition have been proposed, as with ACE-Is, to be partly independent of their anti-hypertensive action [93]. ARBs are marginally less effective than ACE-Is at combating cardiovascular disease, but may be more effective in prevention of AD [94,95]. This differential effect may be due to the interaction of ACE with Aβ. Furthermore, blockage of Ang II binding may aid greater Ang IV formation which can lead to further cognitive enhancement, particularly in executive function [88,96].

Although ACE-Is and ARBs co-administered have been shown to offer the greatest protection from dementia due to their additive effect, such co-prescribing is now thought to be deleterious because of renal complications [94]. Ang IV analogues may also be useful, given their ability to enhance cognition, however, as yet these molecules are too large and hydrophobic to cross the BBB and are rapidly degraded by aminopeptidases [88]. Efforts to correct this are currently underway [12].

NEP and AD

NEP is important in the RAS as it modulates vasodilation via changes to Ang I and bradykinin. However, NEP also degrades monomeric and oligomeric forms of Aβ1-40 and Aβ1-42 [97]. In vivo NEP deficiency coincides with raised Aβ, particularly in the hippocampus [98,99]. Examination of post-mortem normal and AD patients supports this, with an age related decline in NEP protein levels that inversely correlates with increasing Aβ [100].

Females have increased NEP protein and activity [28]. In human and animal models, oestrogen levels are proportional to NEP activity. Oestrogen deficiency decreases NEP activity, which is reversed by HT [101,102]. The regulation of NEP by oestrogen appears to be dependent on its receptors ER-α and ER-β. Once these receptors are activated it is proposed NEP mRNA transcription is induced [102]. This therefore provides a mechanism for oestrogen-mediated Aβ degradation in pre-menopausal women which is lost later in life.

It should also be noted Miners et al. (2010) reported NEP increases throughout life, and, after adjusting for neuronal loss, NEP activity was increased in AD [4]. This may however, like with ACE, be a compensatory upregulation to try to limit the extent of Aβ pathology. More recently, the same group report NEP may protect against the occurrence of cerebral amyloid angiopathy (the deposition of Aβ in blood vessel walls) [103].

Therapeutic potential of Oestrogen in AD?

Oestrogen could have potential as a therapy to treat or reduce the incidence of AD. Oestrogen levels fluctuate through reproductive life (Figure 4) but drop off markedly around mid-life which coincides with the timeframe in which the earliest stages of AD are thought to take root [104] and which may increase women’s vulnerability to developing the disease and how oestrogen supplementation may be of therapeutic benefit [105]. An important consideration remains the risk of endometrial carcinoma and breast cancer which must be considered when contemplating oral formulations [102]. The cancer risk is thought to be mediated through ER-α, and therefore ER-β specific drug targets have more potential [106]. Although ER-β is able to independently regulate NEP expression, many benefits of oestrogen on the RAS may be through ER-α [102,107]. Gene therapy also offers possibilities. Using lentivirus vectors to deliver NEP into mice cells already expressing human APP and secreting Aβ had a marked decrease in extracellular Aβ1-42, while cells expressing NEP were less susceptible to Aβ mediated cell death. These effects of NEP were also observed in vivo [98].

Figure 4.

Figure 4

Fluctuations of endogenous oestrogen levels throughout life and proposed interactions between oestrogen and both blood pressure and Alzheimer’s disease. (A) Shows the changes of oestrogen throughout life and illustrates how oestrogen oscillates throughout a woman’s reproductive life, as well as the overall gradual decline in the hormone as women age. (B) The Venn diagram shows the key changes oestrogen has on the RAS and the overlap between effects on BP and AD. The red arrow indicates the change post-menopause, and the black arrow the effect of oestrogen.

Concluding remarks

The RAS is a complex interaction of vasomodulatory molecules which regularly interact and are partly regulated by oestrogen. The effect of oestrogen may protect against AD by inhibiting Ang II levels (via ACE) and stimulating NEP which could minimise the development of Aβ pathology. The ability of oestrogen to interfere with Ang II signalling may protect the vasculature from pro-inflammatory and hypoxic damage, although the role of bradykinin is as yet unclear. We propose that changes in the modulation of RAS molecules between pre- and post-menopause could alter the risk of hypertension and AD in women compared to men. These mechanisms may go some way towards explaining the higher incidence and prevalence of AD seen in women. Additionally, the complex interaction of the RAS with both BP and Aβ degrading enzymes may explain why there has been variance in the outcomes of clinical trials conducted to date of HT for the prevention of AD.

References

  • 1.Kehoe PG, Passmore PA. The Renin-Angiotensin system and antihypertensive drugs in Alzheimer's disease: current standing of the Angiotensin hypothesis? Journal of Alzheimer's disease: JAD. 2012;30:S251–268. doi: 10.3233/JAD-2012-111376. [DOI] [PubMed] [Google Scholar]
  • 2.Brookmeyer R, Johnson E, Ziegler-Graham K, Arrighi HM. Forecasting the global burden of Alzheimer's disease. Alzheimer's and Dementia. 2007;3:186–191. doi: 10.1016/j.jalz.2007.04.381. [DOI] [PubMed] [Google Scholar]
  • 3.Kehoe PG. Angiotensins and Alzheimer's disease: a bench to bedside overview. Alzheimers Res Ther. 2009;1:3. doi: 10.1186/alzrt3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Miners JS, van Helmond Z, Kehoe PG, Love S. Changes with Age in the Activities of beta-Secretase and the A beta-Degrading Enzymes Neprilysin, Insulin-Degrading Enzyme and Angiotensin-Converting Enzyme. Brain Pathology. 2010;20:794–802. doi: 10.1111/j.1750-3639.2010.00375.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Alberdi E, Sánchez-Gómez MV, Cavaliere F, Pérez-Samartín A, Zugaza JL, Trullas R, Domercq M, Matute C. Amyloid [beta] oligomers induce Ca2+ dysregulation and neuronal death through activation of ionotropic glutamate receptors. Cell Calcium. 2010;47:264–272. doi: 10.1016/j.ceca.2009.12.010. [DOI] [PubMed] [Google Scholar]
  • 6.Li R, Yang LB, Lindholm K, Konishi Y, Yue X, Hampel H, Zhang D, Shen Y. Tumor necrosis factor death receptor signaling cascade is required for amyloid-beta protein-induced neuron death. Journal of Neuroscience. 2004;24:1760–1771. doi: 10.1523/JNEUROSCI.4580-03.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Miners JS, Barua N, Kehoe PG, Gill S, Love S. A beta-Degrading Enzymes: Potential for Treatment of Alzheimer Disease. Journal of Neuropathology and Experimental Neurology. 2011;70:944–959. doi: 10.1097/NEN.0b013e3182345e46. [DOI] [PubMed] [Google Scholar]
  • 8.Gradman AH. Evolving understanding of the renin-angiotensin-aldosterone system: Pathophysiology and targets for therapeutic intervention. American Heart Journal. 2009;157:S1–S6. doi: 10.1016/j.ahj.2009.04.005. [DOI] [PubMed] [Google Scholar]
  • 9.Fyhrquist F, Saijonmaa O. Renin-angiotensin system revisited. Journal of Internal Medicine. 2008;264:224–236. doi: 10.1111/j.1365-2796.2008.01981.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Santos RAS, Ferreira AJ, Silva ACE. Recent advances in the angiotensin-converting enzyme 2-angiotensin(1-7)-Mas axis. Experimental Physiology. 2008;93:519–527. doi: 10.1113/expphysiol.2008.042002. [DOI] [PubMed] [Google Scholar]
  • 11.Wayner MJ, Armstrong DL, Phelix CF, Wright JW, Harding JW. Angiotensin IV enhances LTP in rat dentate gyrus in vivo. Peptides. 2001;22:1403–1414. doi: 10.1016/s0196-9781(01)00475-2. [DOI] [PubMed] [Google Scholar]
  • 12.Chai SY, Yeatman HR, Parker MW, Ascher DB, Thompson PE, Mulvey HT, Albiston AL. Development of cognitive enhancers based on inhibition of insulin-regulated aminopeptidase. BMC Neuroscience. 2008;9(Suppl 2):S14. doi: 10.1186/1471-2202-9-S2-S14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Wright JW, Stubley L, Pederson ES, Kramar EA, Hanesworth JM, Harding JW. Contributions of the brain angiotensin IV-AT4 receptor subtype system to spatial learning. Journal of Neuroscience. 1999;19:3952–3961. doi: 10.1523/JNEUROSCI.19-10-03952.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Giacobini E. Aging, Alzheimer's disease, and estrogen therapy. Experimental Gerontology. 1998;33:865–869. doi: 10.1016/s0531-5565(98)00041-2. [DOI] [PubMed] [Google Scholar]
  • 15.Greendale GA, Lee NP, Arriola ER. The menopause. The Lancet. 1999;353:571–580. doi: 10.1016/S0140-6736(98)05352-5. [DOI] [PubMed] [Google Scholar]
  • 16.Bhavnani BR. Estrogens and menopause: pharmacology of conjugated equine estrogens and their potential role in the prevention of neurodegenerative diseases such as Alzheimer's. The Journal of Steroid Biochemistry and Molecular Biology. 2003;85:473–482. doi: 10.1016/s0960-0760(03)00220-6. [DOI] [PubMed] [Google Scholar]
  • 17.Rannevik G, Jeppsson S, Johnell O, Bjerre B, Laurell-Borulf Y, Svanberg L. A longitudinal study of the perimenopausal transition: altered profiles of steroid and pituitary hormones, SHBG and bone mineral density. Maturitas. 1995;21:103–113. doi: 10.1016/0378-5122(94)00869-9. [DOI] [PubMed] [Google Scholar]
  • 18.Gruber C, Tschugguel W, Schneeberger C, Huber J. Production and actions of estrogens. N Engl J Med. 2002;346:340–352. doi: 10.1056/NEJMra000471. [DOI] [PubMed] [Google Scholar]
  • 19.Brosnihan KB, Li P, Ganten D, Ferrario CM. Estrogen protects transgenic hypertensive rats by shifting the vasoconstrictor-vasodilator balance of RAS. American Journal of Physiology-Regulatory Integrative and Comparative Physiology. 1997;273:R1908–R1915. doi: 10.1152/ajpregu.1997.273.6.R1908. [DOI] [PubMed] [Google Scholar]
  • 20.Taddei S. Blood pressure through aging and menopause. Climacteric. 2009;12:36–40. doi: 10.1080/13697130903004758. [DOI] [PubMed] [Google Scholar]
  • 21.Schunkert H, Danser AHJ, Hense HW, Derkx FHM, Kurzinger S, Riegger GAJ. Effects of estrogen replacement therapy on the renin-angiotensin system in postmenopausal women. Circulation. 1997;95:39–45. doi: 10.1161/01.cir.95.1.39. [DOI] [PubMed] [Google Scholar]
  • 22.Gallagher PE, Li P, Lenhart JR, Chappell MC, Brosnihan KB. Estrogen regulation of angiotensin-converting enzyme mRNA. Hypertension. 1999;33:323–328. doi: 10.1161/01.hyp.33.1.323. [DOI] [PubMed] [Google Scholar]
  • 23.Feldmer M, Kaling M, Takahashi S, Mullins JJ, Ganten D. Glucocorticoid- and estrogen-responsive elements in the 5'-flanking region of the rat angiotensinogen gene. Journal of Hypertension. 1991;9:1005–1012. doi: 10.1097/00004872-199111000-00005. [DOI] [PubMed] [Google Scholar]
  • 24.Stavréus-Evers A, Parini P, Freyschuss B, Elger W, Reddersen G, Sahlin L, Eriksson H. Estrogenic influence on the regulation of hepatic estrogen receptor-[alpha] and serum level of angiotensinogen in female rats. The Journal of Steroid Biochemistry and Molecular Biology. 2001;78:83–88. doi: 10.1016/s0960-0760(01)00077-2. [DOI] [PubMed] [Google Scholar]
  • 25.Fischer M, Baessler A, Schunkert H. Renin angiotensin system and gender differences in the cardiovascular system. Cardiovascular Research. 2002;53:672–677. doi: 10.1016/s0008-6363(01)00479-5. [DOI] [PubMed] [Google Scholar]
  • 26.Fukamizu A, Nishi K, Cho T, Saitoh M, Nakayama K, Ohkubo H, Nakanishi S, Murakami K. Structure of the rat renin gene. Journal of Molecular Biology. 1988;201:443–450. doi: 10.1016/0022-2836(88)90151-9. [DOI] [PubMed] [Google Scholar]
  • 27.Landazuri P, Granobles C, Loango N. Gender Differences in Serum Angiotensin-Converting Enzyme Activity and Blood Pressure in Children: an Observational Study. Arquivos Brasileiros De Cardiologia. 2008;91:352–357. doi: 10.1590/s0066-782x2008001800005. [DOI] [PubMed] [Google Scholar]
  • 28.Pendergrass KD, Pirro NT, Westwood BM, Ferrario CM, Brosnihan KB, Chappell MC. Sex differences in circulating and renal angiotensins of hypertensive mRen(2). Lewis but not normotensive Lewis rats. American Journal of Physiology-Heart and Circulatory Physiology. 2008;295:H10–H20. doi: 10.1152/ajpheart.01277.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Sullivan JC, Semprun-Prieto L, Boesen EI, Pollock DM, Pollock JS. Sex and sex hormones influence the development of albuminuria and renal macrophage infiltration in spontaneously hypertensive rats. American Journal of Physiology-Regulatory Integrative and Comparative Physiology. 2007;293:R1573–R1579. doi: 10.1152/ajpregu.00429.2007. [DOI] [PubMed] [Google Scholar]
  • 30.Nickenig G, Baumer AT, Grohe C, Kahlert S, Strehlow K, Rosenkranz S, Stablein A, Beckers F, Smits JFM, Daemen M, Vetter H, Bohm M. Estrogen modulates AT(1) receptor gene expression in vitro and in vivo. Circulation. 1998;97:2197–2201. doi: 10.1161/01.cir.97.22.2197. [DOI] [PubMed] [Google Scholar]
  • 31.Wu Z, Maric C, Roesch DM, Zheng W, Verbalis JG, Sandberg K. Estrogen regulates adrenal angiotensin AT(1) receptors by modulating AT (1) receptor translation. Endocrinology. 2003;144:3251–3261. doi: 10.1210/en.2003-0015. [DOI] [PubMed] [Google Scholar]
  • 32.Silva-Antonialli MM, Tostes RCA, Fernandes L, Fior-Chadi DR, Akamine EH, Carvalho MHC, Fortes ZB, Nigro D. A lower ratio of AT(1)/AT (2) receptors of angiotensin II is found in female than in male spontaneously hypertensive rats. Cardiovascular Research. 2004;62:587–593. doi: 10.1016/j.cardiores.2004.01.020. [DOI] [PubMed] [Google Scholar]
  • 33.Krishnamurthi K, Verbalis JG, Zheng W, Wu Z, Clerch LB, Sandberg K. Estrogen regulates angiotensin AT(1) receptor expression via cytosolic proteins that bind to the 5 ' leader sequence of the receptor mRNA. Endocrinology. 1999;140:5435–5438. doi: 10.1210/endo.140.11.7242. [DOI] [PubMed] [Google Scholar]
  • 34.Koike G, Horiuchi M, Yamada T, Szpirer C, Jacob HJ, Dzau VJ. Human Type 2 Angiotensin II Receptor Gene: Cloned, Mapped to the X Chromosome, and Its mRNA Is Expressed in the Human Lung. Biochemical and Biophysical Research Communications. 1994;203:1842–1850. doi: 10.1006/bbrc.1994.2402. [DOI] [PubMed] [Google Scholar]
  • 35.Crackower MA, Sarao R, Oudit GY, Yagil C, Kozieradzki I, Scanga SE, Oliveira-dos-Santos AJ, da Costa J, Zhang LY, Pei Y, Scholey J, Ferrario CM, Manoukian AS, Chappell MC, Backx PH, Yagil Y, Penninger JM. Angiotensin-converting enzyme 2 is an essential regulator of heart function. Nature. 2002;417:822–828. doi: 10.1038/nature00786. [DOI] [PubMed] [Google Scholar]
  • 36.Eatman D, Wang M, Socci RR, Thierry-Palmer M, Emmett N, Bayorh MA. Gender differences in the attenuation of salt-induced hypertension by angiotensin (1-7) Peptides. 2001;22:927–933. doi: 10.1016/s0196-9781(01)00404-1. [DOI] [PubMed] [Google Scholar]
  • 37.Deddish PA, Marcic BM, Tan F, Jackman HL, Chen Z, Erdos EG. Neprilysin inhibitors potentiate effects of bradykinin on b2 receptor. Hypertension. 2002;39:619–623. doi: 10.1161/hy0202.103298. [DOI] [PubMed] [Google Scholar]
  • 38.Madeddu P, Emanueli C, Varoni MV, Demontis MP, Anania V, Gorioso N, Chao J. Regulation of bradykinin B-2-receptor expression by oestrogen. British Journal of Pharmacology. 1997;121:1763–1769. doi: 10.1038/sj.bjp.0701255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Chidambaram M, Duncan JA, Lai VS, Cattran DC, Floras JS, Scholey JW, Miller JA. Variation in the renin angiotensin system throughout the normal menstrual cycle. Journal of the American Society of Nephrology. 2002;13:446–452. doi: 10.1681/ASN.V132446. [DOI] [PubMed] [Google Scholar]
  • 40.Greenland KJ, Sernia C. Oestrogenic regulation of brain angiotensinogen. Journal of Neuroendocrinology. 2004;16:508–515. doi: 10.1111/j.1365-2826.2004.01194.x. [DOI] [PubMed] [Google Scholar]
  • 41.Szmuilowicz ED, Adler GK, Williams JS, Green DE, Yao TM, Hopkins PN, Seely EW. Relationship between aldosterone and progesterone in the human menstrual cycle. Journal of Clinical Endocrinology & Metabolism. 2006;91:3981–3987. doi: 10.1210/jc.2006-1154. [DOI] [PubMed] [Google Scholar]
  • 42.Chapman AB, Zamudio S, Woodmansee W, Merouani A, Osorio F, Johnson A, Moore LG, Dahms T, Coffin C, Abraham WT, Schrier RW. Systemic and renal hemodynamic changes in the luteal phase of the menstrual cycle mimic early pregnancy. American Journal of Physiology-Renal Physiology. 1997;273:F777–F782. doi: 10.1152/ajprenal.1997.273.5.F777. [DOI] [PubMed] [Google Scholar]
  • 43.Karjalainen AH, Ruskoaho H, Vuolteenaho O, Heikkinen JE, Bäckström A-C, Savolainen MJ, Kesäniemi YA. Effects of estrogen replacement therapy on natriuretic peptides and blood pressure. Maturitas. 2004;47:201–208. doi: 10.1016/S0378-5122(03)00279-2. [DOI] [PubMed] [Google Scholar]
  • 44.Xu X, Xiao J-C, Luo L-F, Wang S, Zhang J-P, Huang J-J, Liu M-L, Liu C-G, Xu K-Q, Li Y-J, Song H-P. Effects of ovariectomy and 17[beta] -estradiol treatment on the renin-angiotensin system, blood pressure, and endothelial ultrastructure. International Journal of Cardiology. 2008;130:196–204. doi: 10.1016/j.ijcard.2007.08.041. [DOI] [PubMed] [Google Scholar]
  • 45.Dean SA, Tan JH, O'Brien ER, Leenen FHH. 17 beta-estradiol downregulates tissue angiotensin-converting enzyme and ANG II type 1 receptor in female rats. American Journal of Physiology-Regulatory Integrative and Comparative Physiology. 2005;288:R759–R766. doi: 10.1152/ajpregu.00595.2004. [DOI] [PubMed] [Google Scholar]
  • 46.Macova M, Armando I, Zhou J, Baiardi G, Tyurmin D, Larrayoz-Roldan IM, Saavedra JM. Estrogen Reduces Aldosterone, Upregulates Adrenal Angiotensin II AT(2) Receptors and Normalizes Adrenomedullary Fra-2 in Ovariectomized Rats. Neuroendocrinology. 2008;88:276–286. doi: 10.1159/000150977. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Ji H, Menini S, Zheng W, Pesce C, Wu X, Sandberg K. Role of angiotensin-converting enzyme 2 and angiotensin(1-7) in 17 beta-oestradiol regulation of renal pathology in renal wrap hypertension in rats. Experimental Physiology. 2008;93:648–657. doi: 10.1113/expphysiol.2007.041392. [DOI] [PubMed] [Google Scholar]
  • 48.Hunter MS, Mann E. A cognitive model of menopausal hot flushes and night sweats. Journal of Psychosomatic Research. 2010;69:491–501. doi: 10.1016/j.jpsychores.2010.04.005. [DOI] [PubMed] [Google Scholar]
  • 49.Panidis D, Rousso D, Kourtis A, Giannoulis C, Mavromatidis G, Stergiopoulos K. Hormone replacement therapy at the threshold of 21st century. European Journal of Obstetrics & Gynecology and Reproductive Biology. 2001;99:154–164. doi: 10.1016/s0301-2115(01)00405-5. [DOI] [PubMed] [Google Scholar]
  • 50.Rossouw JE, Anderson GL, Prentice RL, LaCroix AZ, Kooperberg C, Stefanick ML, Jackson RD, Beresford SAA, Howard BV, Johnson KC, Kotchen M, Ockene J. Risks and benefits of estrogen plus progestin in healthy postmenopausal women - Principal results from the Women's Health Initiative randomized controlled trial. Jama-Journal of the American Medical Association. 2002;288:321–33. doi: 10.1001/jama.288.3.321. [DOI] [PubMed] [Google Scholar]
  • 51.Oelkers WKH. Effects of estrogens and progestogens on the renin-aldosterone system and blood pressure. Steroids. 1996;61:166–171. doi: 10.1016/0039-128x(96)00007-4. [DOI] [PubMed] [Google Scholar]
  • 52.Seely EW, Walsh BW, Gerhard MD, Williams GH. Estradiol with or without progesterone and ambulatory blood pressure in postmenopausal women. Hypertension. 1999;33:1190–1194. doi: 10.1161/01.hyp.33.5.1190. [DOI] [PubMed] [Google Scholar]
  • 53.Harvey PJ, Morris BL, Miller JA, Floras JS. Estradiol induces discordant angiotensin and blood pressure responses to Orthostasis in healthy postmenopausal women. Hypertension. 2005;45:399–405. doi: 10.1161/01.HYP.0000157161.78721.5c. [DOI] [PubMed] [Google Scholar]
  • 54.Brosnihan KB, Weddle D, Anthony MS, Heise C, Li P, Ferrario CM. Effects of chronic hormone replacement on the renin-angiotensin system in cynomolgus monkeys. Journal of Hypertension. 1997;15:719–726. doi: 10.1097/00004872-199715070-00003. [DOI] [PubMed] [Google Scholar]
  • 55.Owonikoko TK, Fabucci ME, Brown PR, Nisar N, Hilton J, Mathews WB, Ravert HT, Rauseo P, Sandberg K, Dannals RF, Szabo Z. In vivo investigation of estrogen regulation of adrenal and renal angiotensin (AT(1)) receptor expression by PET. Journal of Nuclear Medicine. 2004;45:94–100. [PMC free article] [PubMed] [Google Scholar]
  • 56.Rogers JL, Mitchell AR, Maric C, Sandberg K, Myers A, Mulroney SE. Effect of sex hormones on renal estrogen and angiotensin type 1 receptors in female and male rats. American Journal of Physiology-Regulatory Integrative and Comparative Physiology. 2007;292:R794–R799. doi: 10.1152/ajpregu.00424.2006. [DOI] [PubMed] [Google Scholar]
  • 57.Ichiki T, Usui M, Kato M, Funakoshi Y, Ito K, Egashira K, Takeshita A. Downregulation of angiotensin II type 1 receptor gene transcription by nitric oxide. Hypertension. 1998;31:342–348. doi: 10.1161/01.hyp.31.1.342. [DOI] [PubMed] [Google Scholar]
  • 58.Usui M, Ichiki T, Katoh M, Egashira K, Takeshita A. Regulation of angiotensin II receptor expression by nitric oxide in rat adrenal gland. Hypertension. 1998;32:527–533. doi: 10.1161/01.hyp.32.3.527. [DOI] [PubMed] [Google Scholar]
  • 59.Armando I, Jezova M, Juorio AV, Terron JA, Falcon-Neri A, Semino-Mora C, Imboden H, Saavedra JM. Estrogen upregulates renal angiotensin II AT(2) receptors. American Journal of Physiology-Renal Physiology. 2002;283:F934–F943. doi: 10.1152/ajprenal.00145.2002. [DOI] [PubMed] [Google Scholar]
  • 60.Saavedra JM. Brain angiotensin II: New developments, unanswered questions and therapeutic opportunities. Cellular and Molecular Neurobiology. 2005;25:485–512. doi: 10.1007/s10571-005-4011-5. [DOI] [PubMed] [Google Scholar]
  • 61.Rose JM, Audus KL. AT(1) receptors mediate angiotensin II uptake and transport by bovine brain microvessel endothelial cells in primary culture. Journal of Cardiovascular Pharmacology. 1999;33:30–35. doi: 10.1097/00005344-199901000-00005. [DOI] [PubMed] [Google Scholar]
  • 62.Sherrod M, Liu XB, Zhang XJ, Sigmund CD. Nuclear localization of angiotensinogen in astrocytes. American Journal of Physiology-Regulatory Integrative and Comparative Physiology. 2005;288:R539–R546. doi: 10.1152/ajpregu.00594.2004. [DOI] [PubMed] [Google Scholar]
  • 63.Pelisch N, Hosomi N, Ueno M, Masugata H, Murao K, Hitomi H, Nakano D, Kobori H, Nishiyama A, Kohno M. Systemic candesartan reduces brain angiotensin II via downregulation of brain renin-angiotensin system. Hypertension Research. 2010;33:161–164. doi: 10.1038/hr.2009.200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Iadecola C, Davisson RL. Hypertension and Cerebrovascular Dysfunction. Cell Metabolism. 2008;7:476–484. doi: 10.1016/j.cmet.2008.03.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Sun X, He G, Qing H, Zhou W, Dobie F, Cai F, Staufenbiel M, Huang LE, Song W. Hypoxia facilitates Alzheimer's disease pathogenesis by up-regulating BACE1 gene expression. Proceedings of the National Academy of Sciences of the United States of America. 2006;103:18727–18732. doi: 10.1073/pnas.0606298103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.de la Torre JC. Critically attained threshold of cerebral hypoperfusion: the CATCH hypothesis of Alzheimer's pathogenesis. Neurobiology of Aging. 2000;21:331–342. doi: 10.1016/s0197-4580(00)00111-1. [DOI] [PubMed] [Google Scholar]
  • 67.Saavedra JM, Sánchez-Lemus E, Benicky J. Blockade of brain angiotensin II AT1 receptors ameliorates stress, anxiety, brain inflammation and ischemia: Therapeutic implications. Psychoneuroendocrinology. 2011;36:1–18. doi: 10.1016/j.psyneuen.2010.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Zhang M, Mao Y, Ramirez SH, Tuma RF, Chabrashvili T. Angiotensin II induced cerebral microvascular inflammation and increased blood-brain barrier permeability via oxidative stress. Neuroscience. 2010;171:852–858. doi: 10.1016/j.neuroscience.2010.09.029. [DOI] [PubMed] [Google Scholar]
  • 69.Fleegal-DeMotta MA, Doghu S, Banks WA. Angiotensin II modulates BBB permeability via activation of the AT(1) receptor in brain endothelial cells. Journal of Cerebral Blood Flow and Metabolism. 2009;29:640–647. doi: 10.1038/jcbfm.2008.158. [DOI] [PubMed] [Google Scholar]
  • 70.Gentile MT, Poulet R, Pardo AD, Cifelli G, Maffei A, Vecchione C, Passarelli F, Landolfi A, Carullo P, Lembo G. [beta] -Amyloid deposition in brain is enhanced in mouse models of arterial hypertension. Neurobiology of Aging. 2009;30:222–228. doi: 10.1016/j.neurobiolaging.2007.06.005. [DOI] [PubMed] [Google Scholar]
  • 71.Suzuki Y, Ruiz-Ortega M, Lorenzo O, Ruperez M, Esteban V, Egido J. Inflammation and angiotensin II. The International Journal of Biochemistry & Cell Biology. 2003;35:881–900. doi: 10.1016/s1357-2725(02)00271-6. [DOI] [PubMed] [Google Scholar]
  • 72.Savoia C, Schiffrin EL. Vascular inflammation in hypertension and diabetes: molecular mechanisms and therapeutic interventions. Clinical Science. 2007;112:375–384. doi: 10.1042/CS20060247. [DOI] [PubMed] [Google Scholar]
  • 73.Dandona P, Dhindsa S, Ghanim H, Chaudhuri A. Angiotensin II and inflammation: the effect of angiotensin-converting enzyme inhibition and angiotensin II receptor blockade. Journal of Human Hypertension. 2007;21:20–27. doi: 10.1038/sj.jhh.1002101. [DOI] [PubMed] [Google Scholar]
  • 74.Khachaturian AS, Zandi PP, Lyketsos CG, Hayden KM, Skoog I, Norton MC, Tschanz JT, Mayer L, Welsh-Bohmer KA, Breitner JC. Antihypertensive medication use and incident Alzheimer disease: The cache county study. Neuropsychopharmacology. 2006;31:S39–S39. doi: 10.1001/archneur.63.5.noc60013. [DOI] [PubMed] [Google Scholar]
  • 75.in't Veld BA, Ruitenberg A, Hofman A, Stricker BHC, Breteler MMB. Antihypertensive drugs and incidence of dementia: the Rotterdam Study. Neurobiology of Aging. 2001;22:407–412. doi: 10.1016/s0197-4580(00)00241-4. [DOI] [PubMed] [Google Scholar]
  • 76.Forette F, Seux ML, Staessen JA, Thijs L, Babarskiene MR, Babeanu S, Bossini A, Fagard R, Gil- Extremera B, Laks T, Kobalava Z, Sarti C, Tuomilehto J, Vanhanen H, Webster J, Yodfat Y, Birkenhager WH, Syst-Eur I. The prevention of dementia with antihypertensive treatment. Archives of Internal Medicine. 2002;162:2046–2052. doi: 10.1001/archinte.162.18.2046. [DOI] [PubMed] [Google Scholar]
  • 77.Sink KM, Leng X, Williamson J, Kritchevsky SB, Yaffe K, Kuller L, Yasar S, Atkinson H, Robbins M, Psaty B, Goff DC Jr. Angiotensin-converting enzyme inhibitors and cognitive decline in older adults with hypertension: results from the Cardiovascular Health Study. Archives of Internal Medicine. 2009;169:1195–1202. doi: 10.1001/archinternmed.2009.175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Basso N, Paglia N, Stella I, de Cavanagh EMV, Ferder L, del Rosario Lores Arnaiz M, Inserra F. Protective effect of the inhibition of the renin-angiotensin system on aging. Regulatory Peptides. 2005;128:247–252. doi: 10.1016/j.regpep.2004.12.027. [DOI] [PubMed] [Google Scholar]
  • 79.Vauquelin G, Michotte Y, Smolders I, Sarre S, Ebinger G, Dupont A, Vanderheyden P. Cellular targets for angiotensin II fragments: pharmacological and molecular evidence. Journal of the Renin-Angiotensin-Aldosterone System. 2002;3:195–204. doi: 10.3317/jraas.2002.041. [DOI] [PubMed] [Google Scholar]
  • 80.Kehoe PG, Miners S, Love S. Angiotensins in Alzheimer's disease - friend or foe? Trends in Neurosciences. 2009;32:619–628. doi: 10.1016/j.tins.2009.07.006. [DOI] [PubMed] [Google Scholar]
  • 81.Yamada K, Uchida S, Takahashi S, Takayama M, Nagata Y, Suzuki N, Shirakura S, Kanda T. Effect of a centrally active angiotensin-converting enzyme inhibitor, perindopril, on cognitive performance in a mouse model of Alzheimer's disease. Brain Research. 2010;1352:176–186. doi: 10.1016/j.brainres.2010.07.006. [DOI] [PubMed] [Google Scholar]
  • 82.Hu J, Igarashi A, Kamata M, Nakagawa H. Angiotensin-converting enzyme degrades Alzheimer amyloid beta-peptide (A beta); retards A beta aggregation, deposition, fibril formation; and inhibits cytotoxicity. Journal of Biological Chemistry. 2001;276:47863–47868. doi: 10.1074/jbc.M104068200. [DOI] [PubMed] [Google Scholar]
  • 83.Hajjar I, Brown L, Mack WJ, Chui H. Impact of angiotensin receptor blockers on alzheimer disease neuropathology in a large brain autopsy series. Archives of Neurology. 2012:1–7. doi: 10.1001/archneurol.2012.1010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Sleegers K, den Heijer T, van Dijk EJ, Hofman A, Bertoli-Avella AM, Koudstaal PJ, Breteler MMB, van Duijn CM. ACE gene is associated with Alzheimer's disease and atrophy of hippocampus and amygdala. Neurobiology of Aging. 2005;26:1153–1159. doi: 10.1016/j.neurobiolaging.2004.09.011. [DOI] [PubMed] [Google Scholar]
  • 85.Kehoe PG, Wilcock GK. Is inhibition of the renin-angiotensin system a new treatment option for Alzheimer's disease? The Lancet Neurology. 2007;6:373–378. doi: 10.1016/S1474-4422(07)70077-7. [DOI] [PubMed] [Google Scholar]
  • 86.Ruitenberg A, den Heijer T, Bakker SLM, van Swieten JC, Koudstaal PJ, Hoftnan A, Breteler MMB. Cerebral hypoperfusion and clinical onset of dementia: The Rotterdam study. Annals of Neurology. 2005;57:789–794. doi: 10.1002/ana.20493. [DOI] [PubMed] [Google Scholar]
  • 87.Wright JW, Harding JW. The brain RAS and Alzheimer's disease. Experimental Neurology. 2010;223:326–333. doi: 10.1016/j.expneurol.2009.09.012. [DOI] [PubMed] [Google Scholar]
  • 88.Savaskan E, Hock C, Olivieri G, Bruttel S, Rosenberg C, Hulette C, Müller-Spahn F. Cortical alterations of angiotensin converting enzyme, angiotensin II and AT1 receptor in Alzheimer's dementia. Neurobiology of Aging. 2001;22:541–546. doi: 10.1016/s0197-4580(00)00259-1. [DOI] [PubMed] [Google Scholar]
  • 89.Miners JS, Baig S, Palmer J, Palmer LE, Kehoe PG, Love S. A beta-degrading enzymes in Alzheimer's disease. Brain Pathology. 2008;18:240–252. doi: 10.1111/j.1750-3639.2008.00132.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Miners S, Ashby E, Baig S, Harrison R, Tayler H, Speedy E, Prince JA, Love S, Kehoe PG. Angiotensin-converting enzyme levels and activity in Alzheimer's disease: differences in brain and CSF ACE and association with ACE1 genotypes. Am J Transl Res. 2009;1:163–177. [PMC free article] [PubMed] [Google Scholar]
  • 91.Kaya M, Kalayci R, Kucuk M, Arican N, Elmas I, Kudat H, Korkut F. Effect of losartan on the blood-brain barrier permeability in diabetic hypertensive rats. Life Sciences. 2003;73:3235–3244. doi: 10.1016/j.lfs.2003.06.014. [DOI] [PubMed] [Google Scholar]
  • 92.Fogari R, Mugellini A, Zoppi A, Derosa G, Pasotti C, Fogari E, Preti P. Influence of losartan and atenolol on memory function in very elderly hypertensive patients. Journal of Human Hypertension. 2003;17:781–785. doi: 10.1038/sj.jhh.1001613. [DOI] [PubMed] [Google Scholar]
  • 93.Li NC, Lee A, Whitmer RA, Kivipelto M, Lawler E, Kazis LE, Wolozin B. Use of angiotensin receptor blockers and risk of dementia in a predominantly male population: prospective cohort analysis. British Medical Journal. 2010;340:10. doi: 10.1136/bmj.b5465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Davies NM, Kehoe PG, Ben-Shlomo Y, Martin RM. Associations of Anti-Hypertensive Treatments with Alzheimer's Disease, Vascular Dementia, and Other Dementias. Journal of Alzheimer's Disease. 2011;26:699–708. doi: 10.3233/JAD-2011-110347. [DOI] [PubMed] [Google Scholar]
  • 95.Hajjar I, Hart M, Chen Y-L, Mack W, Milberg W, Chui H, Lipsitz L. Effect of Antihypertensive Therapy on Cognitive Function in Early Executive Cognitive Impairment: A Double-blind Randomized Clinical Trial. Archives of Internal Medicine. 2012;172:442–444. doi: 10.1001/archinternmed.2011.1391. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Kanemitsu H, Tomiyama T, Mori H. Human neprilysin is capable of degrading amyloid [beta] peptide not only in the monomeric form but also the pathological oligomeric form. Neuroscience Letters. 2003;350:113–116. doi: 10.1016/s0304-3940(03)00898-x. [DOI] [PubMed] [Google Scholar]
  • 97.Marr RA, Guan H, Rockenstein E, Kindy M, Gage FH, Verma I, Masliah E, Hersh LB. Neprilysin regulates amyloid Beta peptide levels. J Mol Neurosci. 2004;22:5–11. doi: 10.1385/JMN:22:1-2:5. [DOI] [PubMed] [Google Scholar]
  • 98.Farris W, Schutz SG, Cirrito JR, Shankar GM, Sun XY, George A, Leissring MA, Walsh DM, Qiao W, Holtzman DM, Selkoe DJ. Loss of neprilysin function promotes amyloid plaque formation and causes cerebral amyloid angiopathy. American Journal of Pathology. 2007;171:241–251. doi: 10.2353/ajpath.2007.070105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Hellström-Lindahl E, Ravid R, Nordberg A. Age-dependent decline of neprilysin in Alzheimer's disease and normal brain: Inverse correlation with A[beta] levels. Neurobiology of Aging. 2008;29:210–221. doi: 10.1016/j.neurobiolaging.2006.10.010. [DOI] [PubMed] [Google Scholar]
  • 100.Huang J, Guan H, Booze RM, Eckman CB, Hersh LB. Estrogen regulates neprilysin activity in rat brain. Neuroscience Letters. 2004;367:85–87. doi: 10.1016/j.neulet.2004.05.085. [DOI] [PubMed] [Google Scholar]
  • 101.Liang KW, Yang LQ, Yin C, Xiao ZM, Zhang JJ, Liu YM, Huang J. Estrogen Stimulates Degradation of beta-Amyloid Peptide by Upregulating Neprilysin. Journal of Biological Chemistry. 2010;285:935–942. doi: 10.1074/jbc.M109.051664. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Miners JS, Kehoe P, Love S. Neprilysin Protects against Cerebral Amyloid Angiopathy and A beta-Induced Degeneration of Cerebrovascular Smooth Muscle Cells. Brain Pathology. 2011;21:594–605. doi: 10.1111/j.1750-3639.2011.00486.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Bateman RJ, Xiong C, Benzinger TL, Fagan AM, Goate A, Fox NC, Marcus DS, Cairns NJ, Xie X, Blazey TM, Holtzman DM, Santacruz A, Buckles V, Oliver A, Moulder K, Aisen PS, Ghetti B, Klunk WE, McDade E, Martins RN, Masters CL, Mayeux R, Ringman JM, Rossor MN, Schofield PR, Sperling RA, Salloway S, Morris JC. Clinical and biomarker changes in dominantly inherited Alzheimer's disease. The New England journal of medicine. 2012;367:795–804. doi: 10.1056/NEJMoa1202753. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Wharton W, Gleason CE, Lorenze KR, Markgraf TS, Ries ML, Carlsson CM, Asthana S. Potential role of estrogen in the pathobiology and prevention of Alzheimer's disease. American journal of translational research. 2009;1:131–147. [PMC free article] [PubMed] [Google Scholar]
  • 105.Sun M, Paciga JE, Feldman RI, Yuan ZQ, Coppola D, Lu YY, Shelley SA, Nicosia SV, Cheng JQ. Phosphatidylinositol-3-OH kinase (PI3K)/AKT2, activated in breast cancer, regulates and is induced by estrogen receptor alpha (ER alpha) via interaction between ER alpha and PI3K. Cancer Research. 2001;61:5985–5991. [PubMed] [Google Scholar]
  • 106.Brosnihan KB, Hodgin JB, Smithies O, Maeda N, Gallagher P. Tissue-specific regulation of ACE/ACE2 and AT(1)/AT(2) receptor gene expression by oestrogen in apolipoprotein E/oestrogen receptor-alpha knock-out mice. Experimental Physiology. 2008;93:658–664. doi: 10.1113/expphysiol.2007.041806. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Wharton W, Gleason CE, Sandra O, Carlsson CM, Asthana S. Neurobiological Underpinnings of the Estrogen-Mood Relationship. Current Psychiatry Reviews. 2012;8:247–256. doi: 10.2174/157340012800792957. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from American Journal of Neurodegenerative Disease are provided here courtesy of e-Century Publishing Corporation

RESOURCES