Schematic presentation of possible mechanisms of hypertensive dementia, including amyloid beta metabolism (A), NVU dysfunction (B), vulnerability of the hippocampus (C), and activation of RAS (D), and possible new therapeutic approaches for discovering antihypertensive drugs with anti-dementia actions (E). See text for details. Aβ, amyloid β; APP, amyloid β precursor protein; BACE1, β-site amyloid precursor protein cleaving enzyme 1; BBB, blood-brain barrier; CBF, cerebral blood flow; eNOS, endothelial nitric oxide synthase; FDA, Food and Drug Administration; IL, interleukin; NOX, NADPH oxidase; NVU, neurovascular unit; RAS, renin-angiotensin system; ROS, reactive oxygen species
In Japan, there is an urgent need to respond to dementia, and the Basic Act on Dementia to Promote an Inclusive Society [1] was enacted in January 2024. Among the basic policies, the importance of promoting research on dementia and preventing dementia is emphasized. Hypertension is one of the modifiable risk factors for dementia, and, in particular, midlife hypertension is attracting attention from an epidemiological perspective [2]. It has become clear that hypertension-induced dementia (“hypertensive dementia”) is associated with disturbance in the balance of the neurovascular unit (NVU), which is composed of the three elements of blood vessels, astrocytes, and neurons, in not only vascular dementia (VaD) but also Alzheimer’s disease (AD) [3]; thus, it has been proposed that the accumulation of cardiovascular burden over the life course induces dementia in older age [4]. Although the concept of “hypertensive dementia” has been established clinically, basic research is also progressing to elucidate the mechanisms and to find emerging concepts in particular, since the American Heart Association issued a scientific statement on blood pressure and dementia in 2016 [5]. After introducing the statement, a number of papers related to hypertensive dementia have been reported each year, and attention to this condition has been increasing. The Japanese Society of Hypertension has focused on the importance of hypertension management for the prevention of dementia, and established the Prevention of Cognitive Impairment by Hypertension Management (PCIHM) Working Group to investigate the relationship between hypertension and dementia through both basic and clinical research. This review summarizes the latest developments in basic research on hypertensive dementia. We hope that this will be useful not only for basic researchers in dementia but also for general practitioners who treat hypertension worldwide.
Hypertensive dementia
Hypertension is widely recognized as a major risk factor for cardiovascular disease, particularly stroke, and is consequently considered a contributing factor to VaD. Recent advances in understanding the NVU highlight the intricate interplay among blood vessels, neurons, and astrocytes [3]. Hypertension, by inducing structural and functional alterations in the cerebral vasculature, is increasingly regarded as a potential driver of NVU dysfunction and subsequent neurodegeneration through vascular injury. From this integrated perspective, hypertension is thought to influence both the onset and progression of various forms of dementia, including AD. Notably, prolonged hypertension results in accumulation of vascular stress over time. Numerous epidemiological studies have shown that midlife hypertension significantly increases the risk of developing dementia in later life [6, 7]. While recent evidence suggests that appropriate antihypertensive treatment initiated in midlife may have a preventive effect on dementia, definitive conclusions have yet to be reached [4, 7,8,9]. Conversely, the relationship between hypertension and dementia in older adults remains inconsistent, confounded by factors such as age-related heterogeneity and the potential cognitive consequences of hypotension [7, 10] or blood pressure variability [11,12,13,14]. Both low blood pressure and fluctuating blood pressure levels have been associated with reduced cerebral perfusion, which may exacerbate cognitive decline.
When examining dementia subtypes, dementia with Lewy bodies (DLB) frequently presents with orthostatic hypotension due to autonomic dysfunction. Javanshiri et al. reported that cardiovascular comorbidities, including hypertension, were significantly less prevalent in DLB compared to VaD [15]. Nonetheless, a systematic review by Chin et al. identified an association between cerebral microbleeds (CMBs) and dementia, reporting that in AD, CMBs were linked to both a history of hypertension and amyloid-β (Aβ) burden. Similarly, a relationship between hypertension and CMBs has also been observed in DLB [16]. Thus, while the role of hypertension may be less prominent in certain dementia types, such as DLB, its contribution, particularly through blood-brain barrier (BBB) disruption, cannot be dismissed when considering neurodegenerative diseases more broadly.
Hypertension-induced BBB disruption may trigger neuroinflammation, increase oxidative stress via activation of the renin-angiotensin system (RAS), and impair Aβ clearance, collectively worsening the neural environment and contributing to dementia pathogenesis. Therefore, we propose the term “hypertensive dementia” to encompass dementia arising from vascular impairment and hypertension-related neuropathological mechanisms, such as inflammation and BBB breakdown. In this review, we present recent basic research on the pathophysiology of dementia associated with hypertension and its related factors. We further re-examine the mechanisms underlying hypertensive dementia and explore the potential for its prevention through optimized blood pressure control.
Possible mechanisms of hypertensive dementia
Neurovascular unit dysfunction
Impact of hypertension on cerebral circulation
Hypertension significantly alters the cerebral circulation through changes in both the structure and function of blood vessels. It induces arteriolosclerosis, which is characterized by thickening of the arterial wall and narrowing of the lumen due to hypertrophy and hyperplasia of vascular smooth muscle cells (VSMC). Such changes include the phenotypic switching of VSMC from a contractile form toward a synthetic state. Consequently, vessels become stiffer and less responsive to fluctuations in blood pressure, compromising the blood supply to the brain. This structural rigidity increases the brain’s susceptibility to ischemic events, especially when cerebral autoregulation fails [3]. The functional impairment of cerebral autoregulation in hypertension stems from a loss of endothelial function and increased vascular stiffness. Under normal conditions, the NVU maintains cerebral blood flow (CBF) through a balance of vasodilation and vasoconstriction. In hypertension, this balance skews in favor of vasoconstrictor factors such as angiotensin II (Ang II) and endothelin-1 (ET-1), leading to excessive vasoconstriction, reduced CBF, and exacerbation of conditions like white matter lesions and cognitive decline [17]. Even modest sustained hypertension induces cerebrovascular remodeling and inflammatory changes, driven by mechanical and oxidative stress, inflammation, and neurohumoral factors like Ang II, leading to neuronal injury and cognitive decline through hypoxia and oxidative stress [18]. Hypertension impairs vasodilation by increasing protein kinase C (PKC) activity, which inhibits Rab11A-mediated trafficking of large conductance calcium-activated potassium (BK) channel β1 subunits in arterial myocytes. This leads to reduced nitric oxide (NO)-induced BK channel activation and vasodilation in hypertensive rats, altering arterial contractility and contributing to the risk of cerebrovascular diseases associated with hypertension [19]. Impaired cerebral vasoreactivity and autoregulation have also been observed in transgenic mouse models expressing mutant neurogenic locus notch homolog protein 3 (NOTCH3), which mimic features of cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL), highlighting the role of vascular dysfunction in exacerbating ischemic brain injury [20]. Transient receptor potential vanilloid 4 (TRPV4) channels are non-selective Ca2+ permeable channels expressed in endothelial cells, smooth muscle cells, astrocytes and neurons [21], all of which are included in the NVU and act together to regulate cerebral perfusion [22]. Hypertension impairs TRPV4-mediated dilation of cerebral parenchymal arterioles, reducing cerebral perfusion and contributing to cognitive decline, as shown in both TRPV4 knockout rats and hypertensive mice. TRPV4 channels are influenced by mineralocorticoid receptor activation, and blocking this receptor improves cerebrovascular function and cognition, highlighting its therapeutic potential [23, 24]. Notably, agonistic autoantibodies targeting the alpha 1-adrenergic receptor, which are detected in patients with widespread conditions like hypertension and type 2 diabetes, induce cerebrovascular damage in rats. Magnetic resonance imaging (MRI) revealed reduced vascular area and blood flow in the brain in antibody-exposed rats, suggesting that these antibodies may contribute to central nervous system diseases such as stroke and dementia [25]. Neurovascular coupling (NVC), the mechanism that links neuronal activity to localized increases in CBF, plays a critical role in ensuring the metabolic demands of active neurons are met. Capillary endothelial cells are central to NVC, using inward-rectifier K+ (Kir2.1)-mediated retrograde hyperpolarization to regulate CBF in response to neural activity [26]. In hypertension, this critical Kir2.1 channel-mediated signaling pathway is disrupted by angiotensin II type 1 receptor blocker (ARB)-induced aldosterone breakthrough, impairing functional hyperemia and exacerbating neurovascular uncoupling [27]. This mechanism highlights the importance of Kir2.1 channels in maintaining NVC and underscores their therapeutic potential in preventing cognitive decline linked to vascular dysfunction.
Hypertension-induced neurovascular unit dysfunction
The NVU plays a vital role in maintaining brain homeostasis, controlling the entry of molecules into the brain via the BBB, regulating cerebral blood flow, and responding to neural activity. It consists of vascular endothelial cells, pericytes, astrocyte endfeet, microglia, neurons, and the extracellular matrix. The integrity of the NVU is essential for protecting the brain from potentially harmful substances in the bloodstream and for regulating cerebral perfusion in response to neuronal demands. Hypertension profoundly affects the NVU through multiple mechanisms, including endothelial dysfunction, vascular remodeling, BBB breakdown, and chronic cerebral hypoperfusion [28].
Endothelial cells in the NVU are highly sensitive to changes in blood pressure. Under hypertensive conditions, these cells experience oxidative stress, which disrupts NO signaling—a critical regulator of vascular tone. Reduced NO bioavailability leads to impaired vasodilation, increased vascular resistance, and cerebral hypoperfusion. This contributes to cognitive decline, as sustained reductions in CBF result in neuronal injury and accumulation of toxic metabolites [28]. Hyperhomocysteinemia has been shown to impair regional CBF by reducing NO bioavailability and increasing oxidative stress, exacerbating endothelial dysfunction and NVU damage in hypertensive conditions [29]. Hypertension models have consistently demonstrated that hypertension reduces endothelial nitric oxide synthase (eNOS) expression and activity, leading to a deterioration in cerebrovascular function. This reduction in NO availability contributes to increased vascular resistance, BBB disruption, and impaired Aβ clearance, mechanisms that are closely associated with the pathophysiology of AD. The compromised vascular function exacerbates the accumulation of Aβ, thereby linking cerebrovascular dysfunction to the progression of AD [30].
The BBB is a selective barrier that prevents the entry of potentially harmful substances into the brain. BBB dysfunction is recognized as an early biomarker of cognitive dysfunction in AD, associated with hippocampal damage and occurring independently of amyloid and tau biomarkers [31]. Hypertension-induced mechanical and oxidative stress weaken tight junctions between endothelial cells, leading to increased BBB permeability. This allows plasma proteins, inflammatory mediators, and other neurotoxic substances to infiltrate the brain parenchyma, initiating neuroinflammation and contributing to neuronal damage [28]. Hypertension-induced BBB damage is particularly pronounced in the hippocampus, while chronic hypoperfusion leads to white matter lesions, both of which contribute to cognitive decline and link vascular dysfunction to AD [32]. Capillary dysfunction plays a significant role in impairing oxygen extraction and CBF, further linking microvascular damage to the progression of both stroke and dementia [33]. Over-expression of endothelial ET-1 also exacerbates BBB breakdown, increases oxidative stress, and promotes neuronal apoptosis, further contributing to cognitive deficits in ischemic conditions [34]. The role of perivascular macrophages in BBB dysfunction has been highlighted in hypertension models, where crosstalk between endothelial cells and macrophages aggravates BBB permeability through Ang II receptor signaling, underscoring the immune-vascular interaction in NVU damage [35]. Furthermore, in Ang II-induced hypertensive mice, depletion of microglia and perivascular macrophages using a colony-stimulating factor 1 receptor inhibitor, PLX5622, has been shown to attenuate cognitive decline by suppressing pro-inflammatory activation at sites of BBB disruption [36]. This indicates that targeting immune cells may provide a therapeutic avenue to mitigate hypertension-induced NVU damage and cognitive impairment. Notably, elevated endothelial β-site amyloid precursor protein cleaving enzyme 1 (BACE1) induced by hypertension contributes to cerebral small vessel damage by cleaving tight junction proteins, leading to BBB breakdown, endothelial dysfunction, and cognitive impairment [37]. Intriguingly, chronic hypertension has also been shown to cause more pronounced defects in the integrity of the blood–cerebrospinal fluid (CSF) barrier than in the BBB, particularly in the spontaneously hypertensive rat (SHR). Studies have demonstrated that SHR exhibit increased permeability between the choroid plexus and CSF, indicating significant blood-CSF barrier disruption. These findings highlight the extensive impact of hypertension on NVU dysfunction and underscore the greater susceptibility of the blood-CSF barrier to hypertensive damage compared to the BBB [38].
Mechanisms linking NVU dysfunction to dementia
The cumulative effects of hypertension on NVU-endothelial dysfunction, BBB disruption, and cerebral hypoperfusion create a pro-inflammatory, hypoxic environment conducive to neurodegeneration [39]. Hypertension-induced NVU dysfunction results in reduced CBF, leading to neuronal injury and accumulation of toxic metabolites. One of the key factors contributing to NVU dysfunction is the age-related decline in circulating insulin-like growth factor 1 (IGF-1) level. IGF-1 deficiency exacerbates hypertension-induced microvascular rarefaction in the hippocampus and neocortex, decreasing microvascular density and promoting an anti-angiogenic gene expression profile [40]. This impairment leads to neurovascular uncoupling, increased BBB disruption, and neuroinflammation, contributing to the pathogenesis of vascular cognitive impairment (VCI) [40, 41]. IGF-1 deficiency also enhances cerebral microhemorrhages by increasing oxidative stress-mediated matrix metalloproteinase activation and impairing vascular remodeling, thus increasing arteriolar fragility [42]. VSMC are integral to IGF-1’s vasoprotective effects; VSMC-specific IGF-1 receptor deficiency impairs cerebral myogenic autoregulation and leads to cognitive dysfunction, highlighting the role of VSMC dysfunction in VCI [43]. Additionally, eNOS downregulation associated with IGF-1 deficiency contributes to hypertension-induced AD pathology; long-term exercise promotes eNOS and IGF-1 expression, activating the PI3K/Akt pathway and ameliorating cognitive impairment in the stroke-prone spontaneously hypertensive (SHRSP) rat [44]. These findings underscore IGF-1’s crucial role in cerebrovascular health and suggest targeting IGF-1 signaling as a therapeutic strategy to prevent or mitigate VCI and AD in elderly hypertensive individuals (Fig. 1).
Age-related decline of circulating IGF-1 and neurovascular unit (NVU) dysfunction in hypertension. IGF-1 deficiency exacerbates 1) hypertension-induced microvascular rarefaction in the hippocampus and neocortex, leading to reduced microvascular density and an anti-angiogenic gene expression profile; 2) impaired neurovascular coupling, increased BBB disruption, and neuroinflammation, contributing to VCI pathogenesis; 3) enhanced cerebral microhemorrhages due to oxidative stress-mediated matrix metalloproteinase activation and arteriolar fragility; 4) VSMC dysfunction, impairing cerebral autoregulation and contributing to cognitive decline; and 5) eNOS downregulation, exacerbating hypertension-induced AD pathology, with exercise-mediated IGF-1/PI3K/p-Akt pathway activation promoting eNOS expression to mitigate cognitive impairment. AD Alzheimer’s disease, BBB blood-brain barrier, eNOS endothelial nitric oxide synthase, IGF-1 insulin-like growth factor I, VCI vascular cognitive impairment
Moreover, the phenotypic switching of VSMC from a contractile form toward a synthetic state plays an important role in the progression of atherosclerosis with vascular remodeling such as vascular arterial wall thickening and narrowing of the lumen which reduces CBF and alter CBF autoregulation. A recent study highlighted the role of protein arginine methyltransferase 5 (PRMT5) in VSMC phenotypic switching. PRMT5 expression is upregulated in response to platelet-derived growth factor (PDGF) stimulation and vascular injury. This upregulation leads to histone modifications that suppress the expression of contractile genes while promoting proliferation and migration of VSMC, facilitating the transition to a synthetic phenotype. Inhibition of PRMT5 was found to attenuate neointimal formation, suggesting its potential as a therapeutic target [45]. Another study demonstrated that the combination of increased blood pressure and matrix stiffness synergistically induces VSMC phenotypic switching. This mechanical stress leads to cytoskeletal remodeling and the formation of matrix-degrading structures called podosomes, which are characteristic of the synthetic phenotype. These findings underscore the importance of mechanical cues in vascular remodeling associated with hypertension [46].
Adducin 3 (ADD3) is a subunit of the adducin protein complex involved in actin cytoskeleton regulation, particularly in VSMC. The K572Q mutation in ADD3 disrupts its actin-binding domain, destabilizing the cytoskeleton and impairing autoregulation of CBF. This dysfunction exacerbates NVU damage, increases BBB permeability, and contributes to cognitive decline, particularly in models of hypertension-induced dementia [47].
Hypertension induces chronic oxidative stress in the NVU, primarily through activation of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX) and the production of reactive oxygen species (ROS). ROS directly damage endothelial cells and promote the activation of inflammatory pathways, exacerbating vascular dysfunction and contributing to neuronal injury. Chronic oxidative stress, driven by NOX and inducible nitric oxide synthase (iNOS), exacerbates vascular dysfunction and neuronal injury. Pharmacological inhibition of iNOS and NOX has been shown to attenuate oxidative damage and cognitive deficits in hypertension-induced VaD models [48].
Effects of Alzheimer’s disease pathology
Hypertension and amyloid beta metabolism
Stress-induced hormonal and behavioral reactions may also participate in the development of hypertension, atherosclerosis, insulin resistance, and other peripheral disturbances that may indirectly induce neuropathological processes participating in the development and progression of AD [49, 50]. Microanatomical, neurochemical and behavioral data on SHR favor the hypothesis that this strain is a suitable model of vascular brain disorder [51]. SHR, along with patients with established hypertension, also develop astrogliosis in the brain [52]. Importantly, stress-induced detrimental effects are attractive as etiological factors in AD because they can be reduced by several approaches including behavioral intervention such as exercise [50] and pharmacological interventions such as antihypertensive agents. Antihypertensive intervention is considered to be effective because hypertension is known to induce intracerebral hemorrhage in Tg2576 mice, a model of cerebral amyloid angiopathy (CAA) that recapitulates common vascular changes in AD [53, 54]. CAA and hypertension-induced cerebral small vessel disease (CSVD) share some features but are mechanistically different because a transgenic rat model of CAA type 1 (rTg-DI) and the SHRSP, a rat model of hypertension, show distinct brain proteomic signatures at 12 months of age [55]. In SHRSP, age-dependent extracellular deposition of Aβ is observed from approximately 20-30 weeks onwards [56, 57]. SHRSP treated with N-acetylcysteine (NAC) show an age-dependent increase in BBB breakdown and small perivascular bleeds, accompanied by increased Aβ plaque load in the cerebral cortex [58]. The microvascular and brain injury observed in SHR-SP may be explained by a combination of neurological, inflammatory and vascular functional pathways [59]. In a mouse model of hypertension induced by chronic pressure overload, early cerebrovascular damage develops and is sustained by CD8+IFN-gamma+T lymphocytes [60]. Depletion of perivascular macrophages (PVM) via intracerebroventricular injection of clodronate in two models of hypertension—chronic fourteen-day Ang II infusion and BPH/2 J mice that have life-long hypertension—revealed that PVM are critical for impairing cerebral cortical arteriolar function in hypertension. These effects involve crosstalk between PVM and cerebral endothelial cells, interaction with circulating immune cells, and/or production of reactive oxygen species (ROS) [61]. An experimental study showed that Ang II-induced hypertension with aging (24 months) upregulated expression of Aβ precursor protein (APP), beta secretase, and gamma-secretase in C57BL/6 mice but rather changed hippocampal expression of several APP binding proteins [62]. However, another study showed that Ang II in the Tg2576 model did induce microvascular amyloid deposition and enhanced beta-secretase APP cleavage [63]. In different APP-overexpressing mice bearing Swedish and Indiana mutations, Ang II enhanced Aβ generation and gamma-secretase APP cleavage [64]. In addition, chronic hypertension induces impairment of blood–brain barrier permeability with deposition of Aβ in brain tissue, and passive immunotherapy prevents this latter phenomenon [65]. Thus, the conflicting results regarding the association between spontaneous or Ang II-induced hypertension and Aβ accumulation suggest that hypertension accelerates AD pathology in a context-dependent manner.
Plausible mechanisms of hypertension-induced AD
Several different mechanisms by which hypertension induces AD pathology are proposed. In SHR, water content in the brain is increased, at least partially due to decreased expression of aquaporin 4 [66], a molecule involved in the glymphatic Aβ clearance pathway. In the same model, a high fluorescent molecular weight tracer released passively into the hippocampus showed markedly enhanced spreading and accumulated at interstitial fluid-CSF borders, around arteries surrounded by astrocytes positive for aquaporin 4 [67]. Sieving of high molecular weight solutes at these sites may promote aggregation of Aβ. In the hippocampus of SHR, expression of occludin and zonula occludens-1 was decreased with blood brain barrier disruption, which may facilitate Aβ influx into the brain [68]. Aβ influx may be facilitated by the receptor for advanced glycation end product (RAGE), which may be upregulated by hypertension [69]. RAGE activation in cerebral vessels is a crucial pathogenetic event in a hypertension-induced Aβ accumulation model (transverse aortic coarctation (TAC) mice), suggesting that inhibiting this target could limit the onset of vascular-related AD [21]. In addition, Tg-SwDI mice, a CAA model, showed exacerbation of microvascular Aβ accumulation, vascular inflammation, blood-brain barrier disruption, and pericyte loss, thus accelerating cognitive deficits after 3-6 months of chemically induced chronic hypertension [70]. Transient hypertension, induced by one-month treatment with L-NG-nitroarginine methyl ester (L-NAME), also aggravated CAA and induced loss of cortical myelin in the cingulate cortex in F344TgAD [71]. Rho-associated coiled-coil-containing protein kinase (ROCK) 1 and ROCK2 were found to modulate different aspects of vascular remodeling following hypertension, suggesting that ROCK1 and ROCK2 signaling are a molecular signature of hypertension-induced vascular degeneration in AD [71].
Immune response represents a pivotal and genetically variable modifying factor that may influence vulnerability to neurodegeneration. Microglial cells in SHR overexpressing human truncated tau protein showed lowered expression of MHCII level with a robust phagocytic phenotype compared to those in counterpart WKY rats overexpressing the same construct [72, 73]. At the early stage of hypertension in SHR-SP, hippocampal microglia exhibited upregulated inflammation-associated markers CD11b/c, CD200R, and CD86 [74]. In a TAC model of hypertension, microglial cell activation and interleukin (IL)-1beta upregulation occurred at 3 weeks, followed by Aβ deposition at 4 weeks. However, immune system activation by lipopolysaccharide (LPS) significantly suppressed amyloid burden, suggesting that stimulation in the appropriate time window may be protective [75]. Pro-inflammatory IL-1beta [76, 77] and Toll-like receptor 4 [76] were significantly increased in the hippocampus of SHR [77]. These results suggest that targeting inflammation can either exacerbate or limit vascular-triggered AD pathology in a time- and context-dependent manner [78].
Endothelial nitric oxide synthase and AD
eNOS may be involved in hypertension-induced AD pathology because beta secretase/BACE1 is upregulated in the eNOS-deficient mouse brain, potentially leading to Aβ accumulation. BACE1 is highly expressed in cerebral microvessels from patients with hypertension, and BACE1 cleaves a tight junction protein, occludin, and attenuates eNOS activity [37] (Fig. 2). Consistent with this, endothelial-specific BACE1 transgenic mice show chronic hypoperfusion, BBB leakage, tau hyperphosphorylation, synaptic loss, and cognitive impairment, while inhibition of aberrant BACE1 activity ameliorates tight junction loss, endothelial dysfunction, and memory deficits [37]. eNOS constitutes a feedback signaling loop with Pin1 and Aβ: Pin1 inhibits Aβ production and enhances eNOS activity, and Aβ and eNOS form a mutual inhibition system [79]. Once such a feedback signaling loop is disturbed, it may result in Aβ deposition, hypertension and microbleeds, synchronously leading to AD and CAA. Impaired NO bioavailability exacerbates the pathophysiology of AD in APP/PS1 (amyloid precursor protein/presenilin-1) mice, essentially impacting amyloid load and cognitive impairment, independently of L-NAME-induced hypertension, although only CAA seems to be dependent on hypertension [80].
Plausible mechanism underlying development of hypertension-induced cerebral small vessel disease (CSVD) through BACE1 upregulation. Hypertension is associated with increased expression of endothelial BACE1, which then cleaves APP (resulting in Aβ synthesis; amyloid-dependent pathway) and occludin (resulting in tight junction disruption; amyloid-independent pathway); both of these phenomena culminate in eNOS inactivation and induction of CSVD. The amyloid-dependent pathway is associated with the amyloid form of CSVD (left), which may be more common in Western populations, and the amyloid-independent pathway is associated with hypertension-related CSVD (right), which may be more common in Asian populations. Aβ amyloid β, APP Aβ precursor protein, BACE1 β-site amyloid precursor protein cleaving enzyme 1, CSVD cerebral small vessel disease, eNOS endothelial nitric oxide synthase
Renin-angiotensin system
Direct effects of angiotensin II
Hypertension-induced brain damage is not just a direct result of damage to blood vessels, but may also be caused by a vicious cycle involving increased oxidative stress, neuroinflammation, and sympathetic nerve activation, which may intensify the damage. It is thought that activation of RAS is involved in these processes. Examination of autopsy samples from the frontal cortex showed that protein levels of Ang II type 1 (AT1) receptor and phosphorylated extracellular signal-regulated kinase (ERK) were higher in AD. Protein level of AT1 receptor correlated with an oxidative stress marker (protein carbonyl level) in both AD and cognitive impairment groups [81], indicating activation of RAS in the brain of AD patients. In a study using diabetic mice, swelling of astrocyte endfeet was observed, suggesting disruption of the BBB. Such a pathological change was suppressed by an ARB, telmisartan, indicating involvement of Ang II in the BBB disruption [82]. Ang II reduces the endothelium-dependent response in cerebral blood vessels by inducing oxidative stress such as NOX-dependent ROS, especially peroxynitrite [83, 84]. This endothelial dysfunction may inhibit NVC and induce microvascular damage, resulting in reduced CBF [85] and reduce the increased blood-flow response to external stimuli [86], ultimately impairing cognitive function. A more detailed mechanism has been reported by Iadecola’s group. Daily subcutaneous administration of a sub-pressor dose of Ang II gradually increased blood pressure. After discontinuation of Ang II administration, blood pressure decreased; however, microvascular dysfunction did not quickly recover, indicating that Ang II-induced oxidative stress in the microvasculature significantly disrupts cerebrovascular blood flow regulation and attenuates the increase in CBF induced by neural activity and endothelium-dependent vasodilators [87]. They also showed that suppression of free radicals in the subfornical organ (SFO) by overexpression of Cu/Zn-superoxide dismutase (Cu/Zn-SOD) in a similar model reduced microvascular dysfunction, suggesting that SFO and its hypothalamic centrifugal pathway are involved in Ang II-induced cerebrovascular changes [88]. Faraco et al. also reported that perivascular macrophages (PVM) induce neurovascular dysfunction due to hypertension, leading to cognitive dysfunction [89]. Iadecola et al. summarized one of the underlying mechanisms of hypertension-induced cognitive impairment by Ang II in a review published in 2019, that Ang II activates the AT1 receptor in PVM, and then activates NOX2 leading to vascular oxidative stress and neurovascular dysfunction. This indicates that PVM are an abundant source of vascular ROS production [90] (Fig. 3). Recently, Chen et al. reported a mechanism by which direct binding of Aβ42 to the AT1 receptor in astrocytes activates β-arrestin signaling and induces autophagy dysfunction, leading to Aβ accumulation and inducing synaptotoxicity [91]. On the other hand, a study using two mouse models, AD, hAPP23(+/-) and hAPPswe/PSEN1dE9 mice, demonstrated that Ang II administration for 28 days was too short to induce cognitive decline and brain pathology, suggesting that long-term Ang II administration may be required to impair brain and cognitive function [92].
Possible mechanisms of Ang II-induced cognitive impairment via Ang II- receptors. Angiotensin II (Ang II) binds to Ang II type 1 (AT1) receptors on vascular peripheral cells of the blood brain barrier (BBB) (e.g., perivascular macrophages) and produces oxidative stress, inducing cerebral vascular endothelial cell (EC) damage. This leads to disruption of the BBB, which induces neuroinflammation in the brain through the invasion of inflammatory cells and Ang II itself, which does not pass through the normal BBB. BBB disruption induces swollen astrocyte endfeet, which leads to dysfunction of the neurovascular unit. This is a possible mechanism leading to RAS-activated neurological damage. Aβ amyloid beta, BBB blood-brain barrier, CBF cerebral blood flow, Hippo hippocampus, HP hyperphosphorylation, LTP long-term potentiation, NOX nitric oxide, ROS reactive oxygen species
Actions of ARB administration on RAS
Many basic experiments have investigated the effects of a sub-depressor dose of RAS inhibitors. Intranasal administration of losartan decreased inflammatory cytokines such as IL-12p40/p70, IL-1β, and granulocyte macrophage colony-stimulating factor (GM-CSF) while increasing IL-10, which suppressed inflammation and expression of tyrosine hydroxylase in the striatum and locus coeruleus in APP/PS1 mice [93]. Intranasal administration of losartan also increased IL-10, resulting in a decrease in perivascular β-amyloid and a decline in neurogenesis in SHRSP [94]. Coatl-Cuaya et al. showed that losartan improved cognitive function by regulating hypertension and improving neuroplasticity in chronic hypertensive mice [95]. Campos et al. showed that losartan was effective in reducing cognitive decline in ovariectomized female mice [96]. Takeda et al. reported that olmesartan treatment reduced microvascular oxidative stress in APP23 mice and suppressed cognitive decline in a blood pressure-independent manner [97]. Treatment with olmesartan also suppressed cognitive decline via suppression of oxidative stress in a 5XFAD mouse model in which bilateral common carotid artery occlusion (BCCAO) was applied for 17 minutes to induce ischemia [98]. On the other hand, telmisartan suppressed learning and memory deficits, endothelial dysfunction, and changes in various biochemical parameters in deoxycorticosterone acetate (DOCA)-saline hypertension, as did donepezil [99]. Shindo et al. also reported that telmisartan’s anti-inflammatory effects mediated by the partial agonistic action of peroxisome proliferator-activated receptor (PPAR) γ prevented spatial cognitive impairment in a rat model of oligomeric or aggregated Aβ administration [100]. Liu et al. reported that telmisartan increased Aβ40 and Aβ42 production but decreased the Aβ42/Aβ40 ratio, while losartan, valsartan, and candesartan showed no change in Aβ generation and olmesartan showed a significant increase in Aβ42 generation [101]. Telmisartan has also been shown to be neuroprotective in ovariectomized mice by increasing the protective arm of RAS (angiotensin converting enzyme (ACE) 2/Ang-(1-7)/Mas receptor) when used in combination with 17β-estradiol [102]. Valsartan has been reported to potentiate spinogenesis by enhancing α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor trafficking and synaptic plasticity signaling [103]. Furthermore, candesartan has been reported to improve hippocampal neurogenesis by inhibiting astrocyte and microglial activation independent of blood pressure reduction [104]. Moreover, intranasal administration of irbesartan effectively prevented LPS-induced cognitive decline by activation of PI3K/Akt pathways through neuroprotection and anti-inflammation [105]. Thus, ARB administration in experimental systems has shown brain protective effects based on different mechanisms depending on the drug (Table 1). Since no drug-drug comparisons have been made, this may indicate multiple aspects of Ang II action rather than drug-to-drug differences.
Protective arms of RAS (AT2 agonists and ACE2/Ang-(1-7)/Mas axis)
Ang II type 2 (AT2) receptor activation by ARB administration has been previously reported to protect the brain and prevent cognitive impairment via relative stimulation of the AT2 receptor by unbound Ang II. Thus, a direct AT2 receptor stimulator has previously been expected to be a multiple-organ protective agent. An AT2 receptor agonist (Compound 21, C21) has been reported to have no antihypertensive effect but has shown organ-protective effects in models of AD, kidney disease, stroke, and myocardial infarction [106], and prevention of cognitive impairment [107,108,109]. Treatment of a 60 min transient middle cerebral artery occlusion model with C21 and/or candesartan reduced cytotoxicity and prevented chronic reactive microgliosis in SHR after stroke [110]. Although AT2-related brain protective effects in female mice have been previously reported [111, 112], in ovariectomized (OVX) female SHR subjected to middle cerebral artery occlusion (MCAO), C21 prevented cognitive dysfunction after stroke, presumably through a mechanism involving vascular protection and recovery [113]. Alshammari et al. suggested that modulation of AT2 receptor signaling via C21 may be a useful therapeutic option to improve sensori-motor and cognitive outcomes in aged animals [114]. Although it has been expected that Ang II, which cannot bind to the AT1 receptor, would stimulate the AT2 receptor and cause cerebroprotection in response to ARB administration, some reports suggest that AT2 receptor-targeted therapy is not an ideal strategy to restore Aβ-associated cognitive impairment and cerebrovascular damage, considering the effects of an ARB with PD123319, an AT2 receptor antagonist. [115]. The effects of Ang IV and angiotensin II type 4 (AT4) receptor signaling on cognitive function have been investigated. For example, insulin-regulated aminopeptidase (IRAP), which is also the receptor for Ang IV, co-localized with the insulin-responsive glucose transporter type 4 (GLUT4), and Ang IV promoted activity-dependent glucose uptake in hippocampal sections [116], indicating that Ang IV acts as a cognitive enhancer [117]. Recent reviews have demonstrated the preventive effects of ARBs on cognitive dysfunction via activation of not only the Ang II/AT2 receptor pathway but also the Ang IV/AT4 receptor, Ang-(1-7)/Mas receptor pathways through other unbound Ang II metabolites by AT1 receptor blockade [118, 119]. The cognitive function-related effects induced by receptor stimulation of each RAS component are summarized in Fig. 3, with reference to the content mentioned thus far and past reviews [120].
Also, the protective arms of the RAS, such as the ACE2/Ang-(1-7)/Mas axis, have been expected to prevent dementia via brain protective effects. The brain ACE2/Ang-(1-7)/Mas axis may be involved as protective factors in the etiology and progression of AD, possibly through regulation of tau hyperphosphorylation [121]. ACE2 enhancement through administration of diminazene aceturate (DIZE) improved amyloid-related hippocampal pathology and cognitive deficits in Tg2576 mice [122]. Moreover, systemic administration of Ang (1-7) to 5XFAD mice improved Ang II-induced cognitive impairment, and brain damage and skeletal muscle damage were suppressed [123]. Glycosylated angiotensin-(1-7) peptide Ang-1-6-O-Ser-Glc-NH2 (PNA5) suppressed inflammation and showed beneficial effects on vascular dementia [124]. Furthermore, chronic subcutaneous administration of iodine Ang (1-7) (non-radioactive iodine isotope) to SHR showed that neither Ang (1-7) nor iodine Ang (1-7) affected blood pressure or cognitive function [125]. If AT2 receptors are present, the absence of Mas receptors may have a protective effect against chronic cerebral ischemia [126]. Tran et al. reviewed the effects of neuroinflammation induced by chronic RAS activation on cognitive impairment with consideration of the classical arms and protective arms in the brain [127].
Other RAS components
The nuclear factor-kappa B (NF-kB) inhibitor, natrium diethyldithiocarbamate trihydrate (NDDCT), suppressed vascular dementia in DOCA-induced hypertensive mice as did the ACE inhibitor (ACEI), lisinopril [128]. Lisinopril also significantly reduced brain total tau and phosphorylated tau (pTau)-181 levels [129]. On the other hand, overexpression of ACE in bone marrow monocytic cells alters immune responses that defend against many different types of pathological attacks, including the cognitive decline observed in animal models of AD [130]. Cells overexpressing ACE also showed enhanced immune responses [131]. Increased expression of ACE by macrophages and neutrophils enhances the ability of these cells to respond to immune challenges such as infections, neoplasms, AD, and atherosclerosis [132].
Koide et al. found that chronic hypertension caused neurovascular uncoupling via decreased activity of Kir2.1, an inwardly rectifying potassium channel in capillary endothelial cells. This effect was prevented by amlodipine and not by losartan, but was preventable by eplerenone, suggesting the effects of aldosterone [27]. Moreover, eplerenone alleviated aldosterone-induced brain damage with increased cortical thinning and apoptosis in SHR [133].
This review’s final focus is on neprilysin inhibitors. Some protease inhibitors may affect Aβ metabolism [134], such as through inhibition of neprilysin [135]. The long-term effects of neprilysin inhibitors on amyloid metabolism have been a concern because neprilysin is an amyloid-degrading enzyme [136], and it has been previously reported that neprilysin expression and activity are decreased in the cortex of elderly AD patients [137]. However, Langenickel et al. showed that spinal fluid Aβ40 and Aβ42 were unchanged after sacubitril/valsartan treatment compared to placebo in healthy volunteers [138]. Also, Brum et al. recently showed that plasma Aβ42 and Aβ40 increased significantly from baseline in the sacubitril/valsartan group, while the plasma Aβ42/Aβ40 ratio decreased significantly [139]. Moreover, from a sub-study of a recent clinical study, PARAGON-HF (Prospective Comparison of Angiotensin Receptor Neprilysin Inhibitor with Angiotensin Receptor Blocker Global Outcomes in Heart Failure with Preserved Ejection Fraction), no difference in cognitive change in patients was observed between treatment with sacubitril/valsartan and treatment with valsartan during a median follow-up of 32 months [140]. Thus, considering that sacubitril is thought to hardly cross the blood-brain barrier, the effect of sacubitril/valsartan on Aβ metabolism may not be clinically relevant.
Basic approach to hypertensive dementia
Mystery in dementia approach with animal models—Why is the hippocampus vulnerable?
Cerebrovascular network, energy supply and waste removal processes in hippocampus
The brain hippocampus is an important region that plays a central role in memory and learning, but it is also vulnerable to damage from a variety of factors. This chapter details the mechanisms of vulnerability of the brain hippocampus revealed by experimental animal models. The hippocampus is particularly dependent on blood flow in the brain and is highly vulnerable to lack of blood flow, ischemia [141, 142]. Even short periods of ischemia can damage hippocampal neurons, leading to cell death in the worst cases. Because the hippocampus has a very high energy demand compared to other brain regions, probably due to its complex functions such as memory formation and information processing, if the blood flow supply is cut off, energy deficiency quickly sets in and cellular function is impaired. Neurons in the hippocampus utilize the excitatory neurotransmitter glutamate in abundance. Ischemia causes over-release of glutamate into the extracellular space, leading to over-excitation of neurons. This is known as ‘excitotoxicity’ and is a major cause of neuronal death [143]. In addition, the hippocampus is a particularly frequent focus of epileptic seizures, or seizure origin, probably due to its excitotoxicity and complicated and dense neuronal network. An experimental rat model of eclampsia showed that seizures during pregnancy can cause long-lasting hippocampal dysfunction [144]. On the other hand, ageing causes a decrease in the number and function of neurons in the hippocampus, due to the effects of various age-related factors, including oxidative stress and inflammation, on the hippocampus, eventually leading to reduced memory and cognitive function [145]. The same level of hypertension can lead to significantly earlier onset and increased cerebral microhemorrhages in aged mice than in younger mice [146]. These findings suggest that impairment of the cerebrovascular network can have a strong impact on energy supply and waste removal processes in the hippocampus. Bennette et al. performed mesoscale microscopy methods and in vivo imaging to compare aged and younger murine cerebrovascular networks. They found that vasculature and pericyte densities showed significant reductions in not only the hippocampal network but also basal forebrain areas, which corresponds well with areas of obvious brain atrophy in elderly humans. In addition, the aged brain exhibited increased blood extravasation with leakage of immunoglobulin, indicating compromise of the blood-brain barrier. Of note, the oxygen-carrying capacity of blood is also reduced in the aged mouse brain [147]. Other research groups reported that mitochondrial function of isolated brain microvessels is impaired in aged mice, which could be one reason for the reduced oxygen-carrying capacity of blood [148]. What kind of mechanism contributes to the impaired cerebrovascular networks? Some evidence suggests a contribution of the immune system to cerebrovascular network damage, especially under hypertension. A study in hypertensive rhesus monkeys showed widespread activation of microglial cells and astroglial cells, with leakage of the blood-brain barrier [149]. Recent data demonstrated that perivascular macrophages are the major source of ROS, inducing the neurovascular dysfunction [61]. Kaiser et al. evaluated SHR and reported that both T cells and natural killer cells were significantly increased in the SHR brain microvasculature [150]. Microarray analysis also showed that endothelial proliferation-related genes including those for CD151 antigen and activin A receptor were upregulated in SHR [151]. Hypertension can damage the vascular network as well as the glymphatic network and result in white matter lesions. Mortensen et al. infused paramagnetic contrast into the cisterna magna under dynamic MRI to quantify glymphatic transport kinetics in SHR and age-matched normotensive Wistar-Kyoto rats. They demonstrated that glymphatic system clearance was suppressed in SHR, indicating that hypertension and aging impaired glymphatic transport, resulting in impairment of the removal processes for waste or proteins including amyloid beta in the brain [152]. In vivo MRI clearly showed that the perivascular space is enlarged in SHRSP [153]; meanwhile, white matter volume, including that of the corpus callosum, is decreased in SHR, compared with WKY rats [154]. These findings suggest that the hippocampus has a propensity to require a large energy and nutritional supply, which in itself leads to vulnerability to ageing and hypertension. Therefore, the cerebrovascular network is critically important to maintain the homeostasis of the hippocampus (Fig. 4).
Proposed mechanism for vulnerability of hippocampus. The hippocampus, which is essential for memory and learning, has a high energy demand and requires a large amount of blood flow. On the other hand, aging, hypertension, and decline in estrogen exacerbate oxidative stress and inflammation, leading to disruption of the cerebrovascular network, including BBB leakage and pericyte damage. This mismatch between high demand and low supply ultimately renders the hippocampus highly vulnerable. IL-1β interleukin-1β, IL-17 interleukin-17, BBB blood-brain barrier
Experimental models for assessment of hypertensive dementia
Animal models of NVU damage in hypertension
Animal models have played a crucial role in elucidating the mechanisms of NVU damage and cognitive impairment associated with hypertension. The SHR and SHRSP models closely replicate key features of human VCI, including white matter damage, oxidative stress, neuroinflammation, and BBB disruption [155]. Chronic hypertension induces early BBB dysfunction in the hippocampus of young SHR and SHRSP rats, suggesting BBB impairment in the initial stages of vascular dementia [156]. Similarly, stroke-prone renovascular hypertensive rats (RHRSP), established using the two-kidney, two-clip method, exhibit cerebral hypoperfusion, BBB disruption, white matter damage, and cognitive impairment, mirroring human CSVD pathology and validating RHRSP as a suitable preclinical model [157].
The SHR model is also characterized by sleep apnea, which is associated with microvascular rarefaction in critical brain regions, such as the pre-Bötzinger complex. This condition leads to neuronal loss, neuroinflammation, and cognitive decline. Notably, sleep apnea exacerbates hypertension, further aggravating neurovascular damage, especially in small vessels, and contributing to the development of vascular dementia [158].
Rodent models of chronic hypoperfusion, such as bilateral carotid artery occlusion, are instrumental in replicating the white matter damage and cognitive deficits observed in VCI. These models highlight essential aspects of ischemic injury, microvascular alterations, and behavioral impairments, making them effective tools for studying NVU dysfunction in the context of hypertension-induced VCI [159]. In models of cerebral hypoperfusion, SHR show more severe cognitive deficits and white matter pathology due to worsened BBB breakdown [160] and neurodegenerative outcomes [161]. Chronic hypoxia in SHR disrupts tight junction proteins, leading to increased BBB permeability and triggering angiogenesis in white matter. Minocycline treatment has been shown to reduce BBB damage, enhance angiogenesis, and protect oligodendrocytes [162]. Recent studies utilizing models of gradual cerebral hypoperfusion in SHR have demonstrated slowly progressing white matter damage and working memory impairment, effectively modeling subcortical ischemic vascular dementia while reducing mortality rates compared to traditional models [163]. Moreover, chronic hypoxia in hypertensive rat models exacerbates NVU dysfunction by inducing tau hyperphosphorylation, neuroinflammation, and neurodegeneration, paralleling the structural brain damage and cerebral hypoperfusion seen in human vascular dementia patients [164].
Aging, hypertension, and diabetes have been shown to perturb the brain vasculome in unique ways, with significant alterations in gene expression that influence neurovascular function. Hypertension, in particular, activates pathways related to mitochondrial responses and apoptosis in the brain, contributing to vascular damage and cognitive decline [165]. However, limitations of rodent models, such as their limited white matter, highlight the need for larger species that more accurately replicate human VCI pathology, including white matter damage and cognitive impairment [166].
Transgenic models that combine hypertension with genetic mutations linked to AD, such as the APP/PS1 mouse model, have provided valuable insights into the interaction between hypertension and amyloid pathology. In aged APP/PS1 mice, increased systolic blood pressure decreases CBF and connectivity, leading to neuroinflammation and cognitive impairment, modeling the early vascular contributions to AD [167]. Hypertension accelerates amyloid plaque deposition, reduces cerebral microvessel density, and impairs NO production, thereby hastening cognitive decline in AD models [168].
The chronic elevation of pulse pressure, induced by the transverse aortic constriction model, has also been shown to promote neurovascular damage, increase Aβ accumulation, and prevent cognitive improvement in AD models [169].
Collectively, these animal models offer critical insights into the complex interplay between hypertension, NVU dysfunction, and cognitive impairment. The SHR, SHRSP, and transgenic models, in particular, demonstrate how chronic hypertension induces NVU damage through BBB disruption, neuroinflammation, white matter pathology, and ischemic injury. Such findings underscore the importance of developing therapeutic strategies targeting NVU integrity in the context of hypertension-related VCI and AD. Future research should also aim to refine these models, incorporating species with more extensive white matter to better approximate human pathology.
In vitro models of NVU dysfunction
In vitro models have advanced our understanding of NVU dysfunction under hypertensive conditions. Co-culture systems of endothelial cells, astrocytes, and pericytes recreate the NVU and help explore the effects of hypertension on NVU function. These models reveal that hypertensive conditions increase endothelial permeability, reduce NO production, and alter astrocyte-endothelial interactions, all contributing to NVU dysfunction [159, 170].
In vitro BBB models, including co-culture systems and synthetic microvessels, simulate BBB permeability and NVU dysfunction. However, they are limited in replicating the full complexity of the NVU. Newer synthetic microvessel models show promise for mimicking vascular conditions, such as BBB dynamics, but still face challenges in reproducing full 3D interactions [159, 170].
Organ-on-a-chip technology integrates microfluidics with cell culture to replicate NVU dynamics under hypertensive conditions. These models simulate mechanical stress from elevated blood pressure, providing insights into how hypertension leads to NVU damage and cognitive decline. They also serve as platforms for testing potential therapeutic interventions aimed at preserving NVU function in hypertensive patients [171]. Although organ-on-a-chip technology is a valuable tool that more closely mimics physiological conditions, there are currently no reports of studies related to hypertension and dementia using this system. It is anticipated that future research utilizing this experimental platform will enable more detailed examination of the pathophysiology involved.
Aβ injection as a model of hypertension
Intraarterially injected Aβ constricts cerebral vessels, which may contribute to cerebral hypoperfusion as observed in early AD [172]. Besides, freshly solubilized Aβ enhances endothelin-1-induced vasoconstriction in isolated human middle cerebral and basilar arteries [173]. These observations are consistent with the finding that Aβ (25-35) induced vasoconstriction of mouse aortic rings and coronary arteries in Langendorff-perfused rat hearts, which resulted in decreased coronary flow through alpha1-adrenergic receptor activation [174]. In addition, when injected into the cerebroventricular space, Aβ induces a progressive rise in blood pressure in rats with pre-existing hypertension produced by a high-salt diet through modulation of autonomic activity, which may be interpreted as a physiological response to hypoperfusion complicating the accumulation of Aβ within the brain [175]. Beta-adrenergic receptors may also be involved in AD pathogenesis through effects on Aβ production or inflammation [176]. Aβ-induced vasoconstriction is ameliorated by NS-398, a selective cyclooxygenase-2 inhibitor, or by SB202190, a specific p38 mitogen activated protein kinase inhibitor, and these two agents preserved cerebral blood flow in the double transgenic APPsw AD model (PS1/APPsw) [173]. Aβ-induced hypertension with evidence of reduced baroreflex responsiveness and accumulation of Aβ and elevated endothelin-1 in Wistar rats was ameliorated by the endothelin A receptor antagonist zibotentan (ZD4054) [177].
Therapeutic approach to hypertensive dementia
Prevention of dementia with antihypertensive drugs
Evidence from epidemiological studies and clinical trials suggests that certain antihypertensive drugs may reduce the risk of developing dementia. Notably, some of these drugs are reported to reduce the incidence of both vascular dementia and AD through mechanisms independent of their blood pressure-lowering effects. Recently, the world’s first disease-modifying therapy against AD, an immunotherapy targeting AD-related Aβ protein, has obtained full approval in East Asia, including Japan, Europe, and the United States; however, its efficacy is limited. The anti-dementia actions of generic antihypertensive medications would have a huge impact on the worldwide dementia-related socioeconomic burden.
The cellular and molecular mechanisms underlying the favorable effects of antihypertensive drugs have been extensively investigated in a variety of in vitro and in vivo models of dementia. Some researchers have focused on specific types of antihypertensive drugs and investigated their mechanisms in detail using validated experimental models. This classical approach is straightforward and enables us to elucidate multiple aspects of the actions of certain types of antihypertensive drugs. However, this kind of approach requires a priori knowledge of clinical evidence demonstrating that the target drug has been proven to exert favorable effects on dementia; therefore, this approach cannot be used to discover new drugs with unknown therapeutic potential.
To eliminate this bottleneck, some researchers have adopted comprehensive drug screening methodology to newly identify antihypertensive agents with potential therapeutic effects on dementia, focusing on their specific molecular mechanisms, such as the modulation of Aβ metabolism, neuroinflammation, axonal injury, and protein degradation.
Mechanisms underlying anti-dementia actions of antihypertensive drugs
For certain types of antihypertensive drugs, there is considerable clinical evidence supporting their favorable effects on the pathogenesis of dementia. The cellular and molecular mechanisms underlying those effects have been investigated in a variety of experimental models.
RAS-acting drugs
Among all pharmacological classes of antihypertensive drugs, the mechanisms of the anti-dementia actions of ACEIs and ARBs have been most thoroughly investigated, as the cerebral RAS is known to play a critical role in cognition and the pathophysiology of dementia. Evidence from a variety of in vitro and in vivo models of AD [178,179,180,181,182], the hypertensive [183,184,185,186]/vascular dementia model [187,188,189,190,191], the inflammation-induced cognitive impairment model [192], and the age-related anxiety mouse model [193] supports the concept that RAS-acting drugs could prevent the progression of dementia. The role of the cerebral RAS and its contribution to the pathogenesis of dementia are described in detail elsewhere in this review article [see section 3.1]. Of note, RAS-acting drugs do not always exert neuroprotective effects in experimental mouse models [194], and ARB-induced hypotension is reported to induce cognitive decline associated with tau phosphorylation and reduced spin density [195].
Recent studies have shown an association of ACE with AD, suggesting a potential role of ACE in Aβ degradation. Whether ACEIs exacerbate the cerebral accumulation of neurotoxic Aβ by preventing its degradation or not is a critical issue in the long-term management of hypertension in people at risk of developing dementia [196]. Indeed, some researchers have demonstrated that certain types of ACEI, such as captopril, increased Aβ level in cell culture models and in the brain of AD mouse models at a clinical dose [196]. However, this effect of captopril on Aβ production was hardly observed in other studies [197, 198]. The discrepancy in the results may be attributable to differences in the experimental systems used in each study. More evidence is needed to determine whether certain ACEIs have the property of preventing Aβ degradation and, consequently, exacerbating AD neuropathology in the brain.
Calcium channel blockers (CCBs)
Paris et al. compared dihydropyridine (DHP) calcium channel blockers (CCBs) (felodipine, nifedipine, amlodipine, nilvadipine, nitrendipine, nimodipine, and isradipine) and non-DHP CCBs (verapamil and diltiazem) for their potential effect of modulating Aβ metabolism in both in vitro and in vivo AD models. They reported that DHP CCBs, but not non-DHP CCBs, significantly reduced Aβ accumulation [199]. Another type of DHP CCB (nicardipine) prevented neurodegeneration in the brain of hypertensive rats through mechanisms independent of its blood pressure-lowering effect [200]. Reducing blood pressure variability through DHP CCB (amlodipine) treatment successfully ameliorated cognitive impairment and breakdown of the blood–brain barrier in an aged hypertensive mouse model (BPH/2 J) [201]. DHP CCBs (lercanidipine, manidipine, and nimodipine) repaired hypertension-induced brain damage, such as hyperplasia of glial fibrillary acidic protein (GFAP)-positive astrocytes in the brain of hypertensive rats [202]. Memory deficit and neuroinflammation in a mouse model of traumatic brain injury (TBI) could be ameliorated by treatment with a DHP CCB (nilvadipine) through inhibition of splenic tyrosine kinase (SYK) [203]. Ischemia-induced delayed neuronal death in hippocampal neurons was prevented by treatment with a DHP CCB (lercanidipine) in a hypertensive rat model [204]. Long-term treatment with a DHP CCB (amlodipine), but not a β-blocker (atenolol), attenuated the increase in cerebrovascular resistance observed in a hypertensive rat model [205].
β-blockers
β-adrenergic antagonists are reported to ameliorate cognitive impairment induced by chronic administration of corticosterone in mice [206] and reduce the amyloid burden in an AD mouse model [207]. However, another study using an AD mouse model reported that chronic administration of β-blockers potentiated neuroinflammation and impaired cognitive behavior [208].
Other compounds with antihypertensive effects
Other than the above-mentioned antihypertensive medications, drugs, or compounds with a potential blood pressure-lowering effect, such as traditional Chinese medical compounds [209,210,211,212,213,214], antioxidant carotenoids [215], and other plant-related products [216], are reported to have favorable effects against dementia in cellular and animal models. Modification of Aβ metabolism and reduction of oxidative stress are proposed as putative mechanisms underlying the anti-dementia actions of these compounds.
Unbiased screening-based approach to identify antihypertensive drugs with anti-dementia properties
In contrast to the above-mentioned classical approach targeting a specific type of antihypertensive drug, the screening-based approach enables us to uncover the unknown effects of antihypertensive drugs on the pathogenesis of dementia without a priori knowledge of clinical evidence. Here, we review recent findings derived from such unbiased screening-based approaches (Fig. 5).
Approaches to identify antihypertensive drugs with potential therapeutic effects on dementia. The classical approach focuses on specific types of antihypertensive drugs that have been proven to exert favorable effects on dementia in clinical studies and investigates their mechanisms using validated experimental models. The screening-based approach enables us to uncover unknown effects of antihypertensive drugs on the pathogenesis of dementia without a priori knowledge of clinical evidence. APP amyloid precursor protein
Wang et al. [217] aimed to identify antihypertensive drugs, among 55 currently prescribed antihypertensive medications, that can reduce Aβ generation and its oligomerization to toxic species via a high-throughput screening system. They first found that seven of 55 antihypertensive drugs significantly reduced Aβ generation in a primary neuronal cell culture derived from an APP-Tg AD mouse model (Tg2576). These seven drugs represented six pharmacological subclasses, including a β-blocker (propranolol), an α/β-blocker (carvedilol), ARBs (valsartan and losartan), a CCB (nicardipine), a diuretic (amiloride), and a vasodilator (hydralazine). Among the seven antihypertensive drugs, valsartan was able to prevent oligomerization of Aβ into a neurotoxic species. Then, the authors evaluated the efficacy of valsartan in vivo in the APP-Tg AD mouse model (Tg2576). Valsartan treatment successfully prevented the development of amyloid neuropathology and attenuated cognitive impairment. Importantly, these effects were achieved at below the recommended human equivalent dosage of the drug, with minimal impact on blood pressure.
Furthermore, the same group of researchers has conducted more comprehensive screening assays. They screened 1600 Food and Drug Administration (FDA)-approved drugs, which included 115 cardiovascular drugs representing all pharmacological classes, to identify drugs that could potentially affect Aβ production and cerebral accumulation [197]. In the initial screening with a primary neuronal culture derived from an APP-Tg AD mouse model (Tg2576), they identified 184 FDA-approved drugs that significantly reduced Aβ production, 13 of which were related to cardiovascular treatment. They observed that seven anti-hypertensive drugs (carvedilol, propranolol, valsartan, losartan, hydralazine, nicardipine, and amiloride), representing different pharmacological subclasses, were able to reduce Aβ production in a dose-dependent manner. Among the candidate drugs, nicardipine and propranolol decreased the cerebral accumulation of both Aβ40 and Aβ42 species in the brain of APP-Tg AD mice following a one-month course of treatment.
Pauls et al. identified some anti-hypertensive drugs with therapeutic potential against AD pathogenesis using a unique unbiased data-driven approach [218]. First, they performed comprehensive transcriptomic and proteomic analyses of brain tissues from three distinct AD mouse models (AppNL-F, AppNL-G-F, and 3xTg-AD mice) at different time points and found common molecular patterns representing progression of AD pathogenesis, which they called the “biological signature of AD”. Then, they screened more than 800,000 bioactive compounds to identify small molecules with the potential to reverse the biological AD signature using a computer-based machine-learning approach. This approach identified 8250 candidate compounds that could potentially reverse the AD signature; this included a number of antihypertensive drugs. Among the candidate antihypertensive drugs, bendroflumethiazide and penbutolol had the highest scores in terms of AD-signature reversing properties. Indeed, treatment with these antihypertensive drugs reversed cognitive impairment in AD mouse models. Penbutolol reversed the AD molecular signature and decreased the cerebral accumulation of Aβ in the treated animals. Notably, these drugs have not previously been tested in clinical trials or mouse models of AD.
A growing body of evidence supports the critical role of inflammation in the pathogenesis of dementia, including AD, and RAS may be involved in this neuroinflammation. Gouveia et al. [105] conducted comprehensive pharmacological in vitro and in vivo screening to investigate the effects of RAS-acting drugs on neuroinflammation. First, three ARBs (irbesartan, valsartan, and losartan) and three ACEIs (enalapril, lisinopril, and captopril) were screened in a BV-2 microglial cell culture model with LPS stimulation. Among these RAS-acting drugs, irbesartan most potently inhibited the production of inflammatory cytokines in vitro. Then, the authors evaluated the efficacy of irbesartan in vivo using an LPS-induced neuroinflammation mouse model with cognitive impairment. Intranasal administration of irbesartan significantly ameliorated LPS-induced cognitive impairment, which coincided with modification of the PI3/AKT pathway, dendritic spine density, and oxidative stress.
Siddiqi et al. [219] screened FDA-approved CCBs to identify drugs with autophagy-inducing properties that potentially degrade neurodegenerative disease-associated proteins such as tau (in tauopathy, including AD), α-synuclein (in Parkinson’s disease), and mutant huntingtin (in Huntington’s disease). Of note, they aimed to identify drugs capable of inducing autophagy in the brain at concentrations that are achievable in humans without side effects. They first selected five CCBs with a BBB-penetrant and a long half-time among all FDA-approved CCBs (nimodipine, isradipine, felodipine, diltiazem, and verapamil). In the primary in vitro screening using a cell culture model to evaluate autophagy activity, felodipine caused the greatest increase in the number of autophagosomes. They then confirmed that felodipine could decrease the amount of insoluble tau in two zebrafish transgenic models of tauopathy, demonstrating its in vivo efficacy. Finally, they demonstrated that felodipine reduced the cerebral accumulation of insoluble α-synuclein in a transgenic mouse model of Parkinson’s disease expressing A53T mutant α-synuclein. Notably, this reduction of α-synuclein was associated with improvement of motor impairment and prevention of neuronal cell death in the substantia nigra. Felodipine treatment also improved motor function in a mouse model of Huntington’s disease (B6HD mice).
Michaels et al. [220] investigated the molecular mechanisms underlying axonal loss occurring in the brain of AD and vascular dementia models and revealed that age-related microglial NOX activation plays a critical role. They conducted systematic screening aimed at identifying candidate drugs with antioxidant properties using the drug library of the US Drug Collection. Among over 1000 compounds, they found that indapamide, a diuretic with good evidence of BBB permeability, had a significant antioxidant property. Indapamide inhibited extracellular superoxide generation in a microglia-enriched culture. Furthermore, indapamide treatment in middle-aged mice reduced demyelinated lesion volume and axonal loss in a pharmacological model of axonal injury.
Various treatment strategies for AD in relation to hypertension
The effects of azelnidipine, a DHP CCB used for the treatment of hypertension, on oligomeric Aβ (oAβ)-induced calcium influx and its downstream pathway were assessed in immortalized mouse cerebral endothelial cells (bEND3); azelnidipine was shown to suppress oxidative stress and inflammatory responses in Aβ-stimulated microvasculature in an AD model [221]. Oxidative stress is a factor causing dementia such as AD and VaD. In VaD, NOXs are a molecular source of ROS, and inhibition of NOX activity can reduce cognitive impairment in animal models of VaD [222]. Oxidative stress-induced neuronal damage and expression of genes linked with neuroinflammation were ameliorated by African star apple fruit pulp in rats given cyclosporine (25 mg/kg.bw) to induce hypertension; it mitigated the effect of high blood pressure on brain neurochemicals associated with AD [223].
In the BPH/2 mouse, a polygenic model of hypertension, small cerebral resistance arteries that run across the surface of the brain (pial arteries) showed enhanced pressure-induced constriction due to diminished activity of BK channels—key vasodilatory ion channels of cerebral vascular smooth muscle cells [224]. This pathogenic mechanism is responsible for the observed increase in constriction and may be targeted as a possible avenue for restoring healthy CBF in vascular dementia.
SHR can be used as a model for studying learning impairment and reduced central nicotinic receptors, which persist and worsen with age [225]. SHR showed upregulation of cholinergic markers, vesicular acetylcholine transporter (VAChT) and choline acetyltransferase (ChAT), which suggests that AhE inhibition may exert beneficial effects on cholinergic neurotransmission in an animal model of VaD [226]. Galantamine and the cholinergic precursor choline alphoscerate showed neuroprotection in SHR [227,228,229]. The chromogranin A-derived peptide catestatin (CST) exerted potent antihypertensive and neuroprotective effects plausibly through gamma-aminobutyric acid (GABA)ergic output in SHR, and constitutes a novel potential therapeutic measure to correct impairment of blood flow control in disorders such as stroke and AD [230].
Intravenous infusion of mesenchymal stem cells (MSC) elicited functional recovery in SHRSP through restoration of BBB function via remodeling of the microvasculature and inhibition of Aβ accumulation [231]. The infused MSC activated both transforming growth factor-beta and angiopoietin 1 signaling pathways.
The anti-hypertensive agent ifenprodil has neuroprotective activity through its effects on N-methyl-D-aspartate (NMDA) receptors [232]. A centrally acting ARB, candesartan, was found to exert an anti-inflammatory effect in a model of brain inflammation induced by LPS treatment, which may be translatable to treatment of brain disorders such as AD [233]. Microglial TREM2 (triggering receptor expressed on myeloid cells 2) mitigates inflammatory responses and neuronal apoptosis in angiotensin II-induced hypertension in middle-aged mice [234]. By use of a transgenic C. elegans worm that expresses human Aβ1-42, atrial natriuretic peptide (ANP) was found to delay Aβ-induced worm paralysis, decrease Aβ plaques in the worm brain, and reduce ROS production, suggesting that ANP possesses a novel biological function as an Aβ inhibitor, in the nonaggregated state [235]. Irisin, a hormone cleaved from fibronectin type-III domain-containing protein 5 in response to exercise, was shown to repair damage caused by midlife cardiovascular risk factors such as hypertension, prevent neural Aβ aggregation, and reduce neuroinflammation. In transgenic htau mice, irisin reduced ptau in the hippocampus in only female htau mice, but not in male htau mice [236].
In silico computational strategies for drug candidate selection identified bumetanide, a loop diuretic which inhibits the kidney Na+-K+-2Cl- cotransporter isoform NKCC2 and is used for treatment of hypertension, as a top candidate agent that reverses APOE4 transcriptomic brain signatures and improves memory deficits in APOE4 animal models of AD [237].
2K1C (two-kidney, two-clip) renovascular hypertension-induced impairment of behavioral, biochemical, and endothelial parameters was attenuated by treatment with ulinastatin or quercetin alone as well as in combination [238]. The 2K1C hypertension model shows a significant increase in brain inflammation, oxidative stress, and endothelial dysfunction with memory impairment recapitulating VaD, which was ameliorated by agomelatine, a melatonergic MT1/MT2 agonist, nicorandil, a selective ATP sensitive potassium (KATP) channel opener, and donepezil [239]. Endothelin-1 antagonism with bosentan ameliorated renovascular hypertension-induced VaD [240]. Venlafaxine, a specific serotonin-norepinephrine reuptake inhibitor (SNRI), was also shown to be effective on brain pathology observed in the 2K1C hypertension VaD model [241].
Vasopeptidases, such as neprilysin (NEP), are known to degrade Aβ peptides; therefore, NEP inhibitors used as antihypertensive drugs may induce Aβ accumulation and influence AD risk [242, 243]. Thus, the safety of the use of sacubitril/valsartan (LCZ696), a drug approved for heart failure, for cognitive function should be assessed, especially in the case of long-term administration [244].
Other drug candidates for AD include phytochemicals [245], melatonin [246], and miR-124 inhibitors [247]. These interventions may represent an important strategy for prevention or attenuation of the progression of dementia.
Other therapeutic strategies for protection of cerebrovascular network
Bailey et al. performed mRNA microarray and found that salt exposure increased oxidative stress, activated microglia, and reduced myelin integrity in SHRSP [248]. Of note, dietary salt stimulates Th17 polarization in the gut, increased circulating IL-17. IL-17 can act as a suppressor of NO production, leading to endothelial dysfunction [249, 250]. A high-fat diet is also associated with cerebrovascular network dysfunction. In 5XFAD mice, a high-fat diet increased NOX, upregulated hippocampal oxidative stress, and enhanced cerebrovascular beta-amyloid deposition, leading to cognitive dysfunction [251]. Other research groups reported that a diet high in saturated fats, salt and refined sugar induced hypertension, blockage of brain microvessels and white matter atrophy in Sprague-Dawley rats [252]. On the other hand, a diet supplemented with high acetate and butyrate, releasing high amounts of short-chain fatty acids, regulated intestinal energy metabolism, suppressed immune cell recruitment, and delayed cognitive decline in 5XFAD mice [253].
A sex hormone, estrogen, is also related to cerebral blood flow and increases glucose transport across the blood-brain-barrier. In addition, estrogen decline can suppress the inflammatory response such as increased TNF-alpha and stimulation of TH17 T-cells [254]. Pradhyumnan et al. administered estrogen receptor subtype-beta in a transient MCAO (tMCAO) model of elderly Sprague-Dawley female rats, and found that this treatment decreased hippocampal neuronal death and improved cognitive function, suggesting that estrogen treatment has potential for future clinical investigation [255].
As another therapeutic agent to protect the cerebral vascular network, glucagon-like peptide 1 (GLP-1) has attracted attention. GLP-1 is involved in cellular energy metabolism, and some studies have shown that GLP-1 treatment improves brain glucose metabolism in AD [256]. Recently, Zheng et al. reported that GLP-1 treatment improved cognition in 5xFAD mice through enhancing aerobic glycolysis and suppression of oxidative stress [257]. Resveratrol is a natural polyphenol with anti-inflammatory properties. Interestingly, there is some evidence that resveratrol treatment increases GLP-1, which is secreted by intestinal L cells, attenuates the gut microbiome pattern, and suppresses inflammation in the brain [258]. Treatment with resveratrol attenuated hypertension-induced oxidative stress, suppressed vascular matrix metalloproteinase activation, and reduced cerebral microhemorrhages [146]. These findings indicate that both control of the oxidative stress/inflammatory response and maintenance of endothelial function are important to maintain the cerebrovascular network in a good condition. Potential drugs and compounds to prevent hypertensive dementia are summarized in Table 1, in the order in which they appear in the text.
Asian perspectives
Taking into account clinical considerations, there may be differences in the mechanism of induction of hypertensive dementia between Asians and Westerners, as shown in Fig. 2; there may be more amyloid-independent mechanisms underlying the development of small vessel injury in Asians. In addition, the incidence of stroke is higher in Asia than in Europe and the United States, suggesting that cerebrovascular disease is more likely to occur in Asians than in Westerners [259]. Moreover, RAS activity in Asian subjects may differ from that in Western subjects due to differences in salt intake and salt sensitivity [260]. It is possible that certain diseases may be more likely to occur in Asians, and the presence of these diseases, combined with hypertension, may lead to the development of dementia. We anticipate that more effective preventive measures will be developed in consideration of the basic mechanisms of hypertensive dementia.
Conclusions and perspectives
We herein present an update summary of basic research on hypertensive dementia, describing the factors and mechanisms and how they may induce hippocampal vulnerability, and present a new screening method for anti-dementia drugs. We present a summary that provides an integrative and factor-specific view of the mechanisms of hypertensive dementia (Fig. 6). Table 2 summarizes the animal models included in the text and the effects of stimulation or treatment related to hypertension, as well as a comprehensive review of the molecular and cellular mechanisms of hypertensive dementia from basic research to date (Fig. 7).
Schematic presentation providing comprehensive overview of key mechanisms of hypertensive dementia. Hypertension affects the metabolism of Aβ and induces NVU dysfunction through vascular disorders, increased oxidative stress, and neuroinflammation, leading to vulnerability of the hippocampus. Decreased eNOS affects various aspects, and activation of the renin-angiotensin system modifies each mechanism. Aβ amyloid beta, APP Aβ precursor protein, BACE1 β-site amyloid precursor protein cleaving enzyme 1, eNOS endothelial nitric oxide synthase, NVU neurovascular unit, HP hyperphosphorylation, RAS renin-angiotensin system
Visual summary of complexity and variety of molecular and cellular mechanisms of hypertensive dementia categorized based on key mechanisms. Functional, pathophysiological, and molecular-cellular research findings that induce hypertensive dementia through each key mechanism have been reported. Here is a summary of each key mechanism. Aβ amyloid beta, AD Alzheimer’s disease, ADD3 adducin 3, Ang II angiotensin II, AT1R Ang II type 1 receptor, BBB blood brain barrier, BK channel large conductance calcium-activated potassium channel, IGF-1 insulin-like growth factor 1, iNOS inducible nitric oxide synthase, NO nitric oxide, NOX NADPH oxidase, NVU neurovascular unit, ROS reactive oxygen species, TRPV4 transient receptor potential vanilloid 4, VaD vascular dementia
Unfortunately, it is frustrating that there is currently no conclusive interventional trial that can demonstrate whether treating hypertension can reduce the onset or progression of dementia, but there are numerous studies that have demonstrated that endothelial BACE1 increases and eNOS decreases albumin leakage, microglial activation, and inflammatory cytokines such as IL-1 beta, which will contribute to elucidation of the pathophysiology and early treatment of hypertensive dementia. The establishment of characteristic biomarkers for hypertensive dementia is also desirable. We hope this review will help in this regard. To confront the ongoing dementia pandemic, we should continue to make progress in basic research and develop clinically beneficial prevention and treatment methods based on the results of such research.
References
House of Councillors, The National Diet of Japan. Basic Act for Dementia to promote the realisation of a symbiotic society. 2023. https://www.sangiin.go.jp/japanese/joho1/kousei/gian/211/meisai/m211090211024.htm.
Livingston G, Huntley J, Liu KY, Costafreda SG, Selbaek G, Alladi S, et al. Dementia prevention, intervention, and care: 2024 report of the Lancet standing Commission. Lancet. 2024;404:572–628.
Santisteban MM, Iadecola C, Carnevale D. Hypertension, neurovascular dysfunction, and cognitive impairment. Hypertension. 2023;80:22–34.
Qiu C, Fratiglioni L. A major role for cardiovascular burden in age-related cognitive decline. Nat Rev Cardiol. 2015;12:267–77.
Iadecola C, Yaffe K, Biller J, Bratzke LC, Faraci FM, Gorelick PB, et al. Impact of hypertension on cognitive function: a scientific statement from the American Heart Association. Hypertension. 2016;68:e67–e94.
Ninomiya T, Ohara T, Hirakawa Y, Yoshida D, Doi Y, Hata J, et al. Midlife and late-life blood pressure and dementia in Japanese elderly: the Hisayama study. Hypertension. 2011;58:22–8.
Ou YN, Tan CC, Shen XN, Xu W, Hou XH, Dong Q, et al. Blood pressure and risks of cognitive impairment and dementia: a systematic review and meta-analysis of 209 prospective studies. Hypertension. 2020;76:217–25.
Hughes D, Judge C, Murphy R, Loughlin E, Costello M, Whiteley W, et al. Association of blood pressure lowering with incident dementia or cognitive impairment: a systematic review and meta-analysis. JAMA. 2020;323:1934–44.
Lennon MJ, Lam BCP, Lipnicki DM, Crawford JD, Peters R, Schutte AE, et al. Use of antihypertensives, blood pressure, and estimated risk of dementia in late life: an individual participant data meta-analysis. JAMA Netw Open. 2023;6:e2333353.
Peters R, Xu Y, Fitzgerald O, Aung HL, Beckett N, Bulpitt C, et al. Blood pressure lowering and prevention of dementia: an individual patient data meta-analysis. Eur Heart J. 2022;43:4980–90.
de Heus RAA, Tzourio C, Lee EJL, Opozda M, Vincent AD, Anstey KJ, et al. Association between blood pressure variability with dementia and cognitive impairment: a systematic review and meta-analysis. Hypertension. 2021;78:1478–89.
Hayakawa T, McGarrigle CA, Coen RF, Soraghan CJ, Foran T, Lawlor BA, et al. Orthostatic blood pressure behavior in people with mild cognitive impairment predicts conversion to dementia. J Am Geriatr Soc. 2015;63:1868–73.
Jia P, Lee HWY, Chan JYC, Yiu KKL, Tsoi KKF. Long-term blood pressure variability increases risks of dementia and cognitive decline: a meta-analysis of longitudinal studies. Hypertension. 2021;78:996–1004.
Li C, Ma Y, Hua R, Yang Z, Zhong B, Wang H, et al. Dose-response relationship between long-term blood pressure variability and cognitive decline. Stroke. 2021;52:3249–57.
Javanshiri K, Haglund M, Englund E. Cardiovascular disease, diabetes mellitus, and hypertension in Lewy body disease: a comparison with other dementia disorders. J Alzheimers Dis. 2019;71:851–9.
Chin KS, Holper S, Loveland P, Churilov L, Yassi N, Watson R. Prevalence of cerebral microbleeds in Alzheimer’s disease, dementia with Lewy bodies and Parkinson’s disease dementia: a systematic review and meta-analysis. Neurobiol Aging. 2024;134:74–83.
Pacholko A, Iadecola C. Hypertension, neurodegeneration, and cognitive decline. Hypertension. 2024;81:991–1007.
Pannozzo MA, Holland PR, Scullion G, Talbot R, Mullins JJ, Horsburgh K. Controlled hypertension induces cerebrovascular and gene alterations in Cyp1a1-Ren2 transgenic rats. J Am Soc Hypertens. 2013;7:411–9.
Leo MD, Zhai X, Yin W, Jaggar JH. Impaired trafficking of beta1 subunits inhibits BK channels in cerebral arteries of hypertensive rats. Hypertension. 2018;72:765–75.
Lacombe P, Oligo C, Domenga V, Tournier-Lasserve E, Joutel A. Impaired cerebral vasoreactivity in a transgenic mouse model of cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy arteriopathy. Stroke. 2005;36:1053–8.
Iddings JA, Kim KJ, Zhou Y, Higashimori H, Filosa JA. Enhanced parenchymal arteriole tone and astrocyte signaling protect neurovascular coupling mediated parenchymal arteriole vasodilation in the spontaneously hypertensive rat. J Cereb Blood Flow Metab. 2015;35:1127–36.
Iadecola C. The neurovascular unit coming of age: a journey through neurovascular coupling in health and disease. Neuron. 2017;96:17–42.
Diaz-Otero JM, Yen TC, Fisher C, Bota D, Jackson WF, Dorrance AM. Mineralocorticoid receptor antagonism improves parenchymal arteriole dilation via a TRPV4-dependent mechanism and prevents cognitive dysfunction in hypertension. Am J Physiol Heart Circ Physiol. 2018;315:H1304–H15.
Diaz-Otero JM, Yen TC, Ahmad A, Laimon-Thomson E, Abolibdeh B, Kelly K, et al. Transient receptor potential vanilloid 4 channels are important regulators of parenchymal arteriole dilation and cognitive function. Microcirculation. 2019;26:e12535.
Karczewski P, Pohlmann A, Wagenhaus B, Wisbrun N, Hempel P, Lemke B, et al. Antibodies to the alpha1-adrenergic receptor cause vascular impairments in rat brain as demonstrated by magnetic resonance angiography. PLoS One. 2012;7:e41602.
Longden TA, Dabertrand F, Koide M, Gonzales AL, Tykocki NR, Brayden JE, et al. Capillary K(+)-sensing initiates retrograde hyperpolarization to increase local cerebral blood flow. Nat Neurosci. 2017;20:717–26.
Koide M, Harraz OF, Dabertrand F, Longden TA, Ferris HR, Wellman GC, et al. Differential restoration of functional hyperemia by antihypertensive drug classes in hypertension-related cerebral small vessel disease. J Clin Investig. 2021;131
Baggeroer CE, Cambronero FE, Savan NA, Jefferson AL, Santisteban MM. Basic mechanisms of brain injury and cognitive decline in hypertension. Hypertension. 2024;81:34–44.
Toda N, Okamura T. Hyperhomocysteinemia impairs regional blood flow: involvements of endothelial and neuronal nitric oxide. Pflugers Arch. 2016;468:1517–25.
Iadecola C. Hypertension and dementia. Hypertension. 2014;64:3–5.
Nation DA, Sweeney MD, Montagne A, Sagare AP, D’Orazio LM, Pachicano M, et al. Blood-brain barrier breakdown is an early biomarker of human cognitive dysfunction. Nat Med. 2019;25:270–6.
Ueno M, Chiba Y, Matsumoto K, Murakami R, Fujihara R, Kawauchi M, et al. Blood-brain barrier damage in vascular dementia. Neuropathology. 2016;36:115–24.
Ostergaard L, Jespersen SN, Engedahl T, Gutierrez Jimenez E, Ashkanian M, Hansen MB, et al. Capillary dysfunction: its detection and causative role in dementias and stroke. Curr Neurol Neurosci Rep. 2015;15:37.
Zhang X, Yeung PK, McAlonan GM, Chung SS, Chung SK. Transgenic mice over-expressing endothelial endothelin-1 show cognitive deficit with blood-brain barrier breakdown after transient ischemia with long-term reperfusion. Neurobiol Learn Mem. 2013;101:46–54.
Santisteban MM, Ahn SJ, Lane D, Faraco G, Garcia-Bonilla L, Racchumi G, et al. Endothelium-macrophage crosstalk mediates blood-brain barrier dysfunction in hypertension. Hypertension. 2020;76:795–807.
Kerkhofs D, van Hagen BT, Milanova IV, Schell KJ, van Essen H, Wijnands E, et al. Pharmacological depletion of microglia and perivascular macrophages prevents vascular cognitive impairment in Ang II-induced hypertension. Theranostics. 2020;10:9512–27.
Zhou H, Gao F, Yang X, Lin T, Li Z, Wang Q, et al. Endothelial BACE1 impairs cerebral small vessels via tight junctions and eNOS. Circ Res. 2022;130:1321–41.
Al-Sarraf H, Philip L. Effect of hypertension on the integrity of blood brain and blood CSF barriers, cerebral blood flow and CSF secretion in the rat. Brain Res. 2003;975:179–88.
Lu X, Moeini M, Li B, Thorin E, Lesage F. Hypertension accelerates cerebral tissue PO(2) disruption in Alzheimer’s disease. Neurosci Lett. 2020;715:134626.
Tarantini S, Tucsek Z, Valcarcel-Ares MN, Toth P, Gautam T, Giles CB, et al. Circulating IGF-1 deficiency exacerbates hypertension-induced microvascular rarefaction in the mouse hippocampus and retrosplenial cortex: implications for cerebromicrovascular and brain aging. Age (Dordr). 2016;38:273–89.
Toth P, Tucsek Z, Tarantini S, Sosnowska D, Gautam T, Mitschelen M, et al. IGF-1 deficiency impairs cerebral myogenic autoregulation in hypertensive mice. J Cereb Blood Flow Metab. 2014;34:1887–97.
Tarantini S, Valcarcel-Ares NM, Yabluchanskiy A, Springo Z, Fulop GA, Ashpole N, et al. Insulin-like growth factor 1 deficiency exacerbates hypertension-induced cerebral microhemorrhages in mice, mimicking the aging phenotype. Aging Cell. 2017;16:469–79.
Miller LR, Bickel MA, Tarantini S, Runion ME, Matacchiera Z, Vance ML, et al. IGF1R deficiency in vascular smooth muscle cells impairs myogenic autoregulation and cognition in mice. Front Aging Neurosci. 2024;16:1320808.
Zhang L, Zheng H, Luo J, Li L, Pan X, Jiang T, et al. Inhibition of endothelial nitric oxide synthase reverses the effect of exercise on improving cognitive function in hypertensive rats. Hypertens Res. 2018;41:414–25.
Zhu N, Guo ZF, Kazama K, Yi B, Tongmuang N, Yao H, et al. Epigenetic regulation of vascular smooth muscle cell phenotypic switch and neointimal formation by PRMT5. Cardiovasc Res. 2023;119:2244–55.
Swiatlowska P, Sit B, Feng Z, Marhuenda E, Xanthis I, Zingaro S, et al. Pressure and stiffness sensing together regulate vascular smooth muscle cell phenotype switching. Sci Adv. 2022;8:eabm3471.
Gonzalez-Fernandez E, Fan L, Wang S, Liu Y, Gao W, Thomas KN, et al. The adducin saga: pleiotropic genomic targets for precision medicine in human hypertension-vascular, renal, and cognitive diseases. Physiol Genomics. 2022;54:58–70.
Sharma B, Singh N. Pharmacological inhibition of inducible nitric oxide synthase (iNOS) and nicotinamide adenine dinucleotide phosphate (NADPH) oxidase, convalesce behavior and biochemistry of hypertension induced vascular dementia in rats. Pharmacol Biochem Behav. 2013;103:821–30.
Mravec B, Horvathova L, Padova A. Brain under stress and Alzheimer’s disease. Cell Mol Neurobiol. 2018;38:73–84.
Nation DA, Hong S, Jak AJ, Delano-Wood L, Mills PJ, Bondi MW, et al. Stress, exercise, and Alzheimer’s disease: a neurovascular pathway. Med Hypotheses. 2011;76:847–54.
Tayebati SK, Tomassoni D, Amenta F. Spontaneously hypertensive rat as a model of vascular brain disorder: microanatomy, neurochemistry and behavior. J Neurol Sci. 2012;322:241–9.
Tomassoni D, Avola R, Di Tullio MA, Sabbatini M, Vitaioli L, Amenta F. Increased expression of glial fibrillary acidic protein in the brain of spontaneously hypertensive rats. Clin Exp Hypertens. 2004;26:335–50.
Nyul-Toth A, Tarantini S, Kiss T, Toth P, Galvan V, Tarantini A, et al. Increases in hypertension-induced cerebral microhemorrhages exacerbate gait dysfunction in a mouse model of Alzheimer’s disease. Geroscience. 2020;42:1685–98.
Passos GF, Kilday K, Gillen DL, Cribbs DH, Vasilevko V. Experimental hypertension increases spontaneous intracerebral hemorrhages in a mouse model of cerebral amyloidosis. J Cereb Blood Flow Metab. 2016;36:399–404.
Schrader JM, Stanisavljevic A, Xu F, Van Nostrand WE. Distinct brain proteomic signatures in cerebral small vessel disease rat models of hypertension and cerebral amyloid angiopathy. J Neuropathol Exp Neurol. 2022;81:731–45.
Bueche CZ, Hawkes C, Garz C, Vielhaber S, Attems J, Knight RT, et al. Hypertension drives parenchymal beta-amyloid accumulation in the brain parenchyma. Ann Clin Transl Neurol. 2014;1:124–9.
Schreiber S, Drukarch B, Garz C, Niklass S, Stanaszek L, Kropf S, et al. Interplay between age, cerebral small vessel disease, parenchymal amyloid-beta, and tau pathology: longitudinal studies in hypertensive stroke-prone rats. J Alzheimers Dis. 2014;42:S205–15.
Bueche CZ, Garz C, Stanaszek L, Niklass S, Kropf S, Bittner D, et al. Impact of N-acetylcysteine on cerebral amyloid-beta plaques and kidney damage in spontaneously hypertensive stroke-prone rats. J Alzheimers Dis. 2014;42:S305–13.
Bailey EL, McBride MW, Beattie W, McClure JD, Graham D, Dominiczak AF, et al. Differential gene expression in multiple neurological, inflammatory and connective tissue pathways in a spontaneous model of human small vessel stroke. Neuropathol Appl Neurobiol. 2014;40:855–72.
Carnevale L, Perrotta M, Mastroiacovo F, Perrotta S, Migliaccio A, Fardella V, et al. Advanced magnetic resonance imaging to define the microvascular injury driven by neuroinflammation in the brain of a mouse model of hypertension. Hypertension. 2024;81:636–47.
Faraco G, Park L, Anrather J, Iadecola C. Brain perivascular macrophages: characterization and functional roles in health and disease. J Mol Med (Berl). 2017;95:1143–52.
Csiszar A, Tucsek Z, Toth P, Sosnowska D, Gautam T, Koller A, et al. Synergistic effects of hypertension and aging on cognitive function and hippocampal expression of genes involved in beta-amyloid generation and Alzheimer’s disease. Am J Physiol Heart Circ Physiol. 2013;305:H1120–30.
Faraco G, Park L, Zhou P, Luo W, Paul SM, Anrather J, et al. Hypertension enhances Abeta-induced neurovascular dysfunction, promotes beta-secretase activity, and leads to amyloidogenic processing of APP. J Cereb Blood Flow Metab. 2016;36:241–52.
Liu J, Liu S, Matsumoto Y, Murakami S, Sugakawa Y, Kami A, et al. Angiotensin type 1a receptor deficiency decreases amyloid beta-protein generation and ameliorates brain amyloid pathology. Sci Rep. 2015;5:12059.
Gentile MT, Poulet R, Di Pardo A, Cifelli G, Maffei A, Vecchione C, et al. Beta-amyloid deposition in brain is enhanced in mouse models of arterial hypertension. Neurobiol Aging. 2009;30:222–8.
Naessens DMP, Coolen BF, de Vos J, VanBavel E, Strijkers GJ, Bakker E. Altered brain fluid management in a rat model of arterial hypertension. Fluids Barriers CNS. 2020;17:41.
Bedussi B, Naessens DMP, de Vos J, Olde Engberink R, Wilhelmus MMM, Richard E, et al. Enhanced interstitial fluid drainage in the hippocampus of spontaneously hypertensive rats. Sci Rep. 2017;7:744.
Wang Y, Zhang R, Tao C, Xu Z, Chen W, Wang C, et al. Blood-brain barrier disruption and perivascular beta-amyloid accumulation in the brain of aged rats with spontaneous hypertension: evaluation with dynamic contrast-enhanced magnetic resonance imaging. Korean J Radio. 2018;19:498–507.
Carnevale D, Mascio G, D’Andrea I, Fardella V, Bell RD, Branchi I, et al. Hypertension induces brain beta-amyloid accumulation, cognitive impairment, and memory deterioration through activation of receptor for advanced glycation end products in brain vasculature. Hypertension. 2012;60:188–97.
Kruyer A, Soplop N, Strickland S, Norris EH. Chronic hypertension leads to neurodegeneration in the TgSwDI mouse model of Alzheimer’s disease. Hypertension. 2015;66:175–82.
Lai AY, Joo IL, Trivedi AU, Dorr A, Hill ME, Stefanovic B, et al. Cerebrovascular damage after midlife transient hypertension in non-transgenic and Alzheimer’s disease rats. Brain Res. 2021;1758:147369.
Stozicka Z, Zilka N, Novak P, Kovacech B, Bugos O, Novak M. Genetic background modifies neurodegeneration and neuroinflammation driven by misfolded human tau protein in rat model of tauopathy: implication for immunomodulatory approach to Alzheimer’s disease. J Neuroinflammation. 2010;7:64.
Zilka N, Stozicka Z, Cente M, Kazmerova Z, Kovacech B, Novak M. Immunomodulation of memory-impairing protein tau in Alzheimer’s disease. Neurodegener Dis. 2012;10:242–5.
Morton L, Arndt P, Garza AP, Henneicke S, Mattern H, Gonzalez M, et al. Spatio-temporal dynamics of microglia phenotype in human and murine cSVD: impact of acute and chronic hypertensive states. Acta Neuropathol Commun. 2023;11:204.
Carnevale D, Mascio G, Ajmone-Cat MA, D’Andrea I, Cifelli G, Madonna M, et al. Role of neuroinflammation in hypertension-induced brain amyloid pathology. Neurobiol Aging. 2012;33:205 e19–29.
Gao F, Jing Y, Zang P, Hu X, Gu C, Wu R, et al. Vascular cognitive impairment caused by cerebral small vessel disease is associated with the TLR4 in the hippocampus. J Alzheimers Dis. 2019;70:563–72.
Tayebati SK, Tomassoni D, Amenta F. Neuroinflammatory markers in spontaneously hypertensive rat brain: an immunohistochemical study. CNS Neurol Disord Drug Targets. 2016;15:995–1000.
Evans LE, Taylor JL, Smith CJ, Pritchard HAT, Greenstein AS, Allan SM. Cardiovascular comorbidities, inflammation, and cerebral small vessel disease. Cardiovasc Res. 2021;117:2575–88.
Wang JZ, Zhu WD, Xu ZX, Du WT, Zhang HY, Sun XW, et al. Pin1, endothelial nitric oxide synthase, and amyloid-beta form a feedback signaling loop involved in the pathogenesis of Alzheimer’s disease, hypertension, and cerebral amyloid angiopathy. Med Hypotheses. 2014;82:145–50.
Cifuentes D, Poittevin M, Bonnin P, Ngkelo A, Kubis N, Merkulova-Rainon T, et al. Inactivation of nitric oxide synthesis exacerbates the development of Alzheimer disease pathology in APPPS1 mice (amyloid precursor protein/presenilin-1). Hypertension. 2017;70:613–23.
Cosarderelioglu C, Nidadavolu LS, George CJ, Marx-Rattner R, Powell L, Xue QL, et al. Higher angiotensin II type 1 receptor levels and activity in the postmortem brains of older persons with Alzheimer’s dementia. J Gerontol A Biol Sci Med Sci. 2022;77:664–72.
Min LJ, Mogi M, Shudou M, Jing F, Tsukuda K, Ohshima K, et al. Peroxisome proliferator-activated receptor-gamma activation with angiotensin II type 1 receptor blockade is pivotal for the prevention of blood-brain barrier impairment and cognitive decline in type 2 diabetic mice. Hypertension. 2012;59:1079–88.
Bloch S, Obari D, Girouard H. Angiotensin and neurovascular coupling: beyond hypertension. Microcirculation. 2015;22:159–67.
Girouard H, Park L, Anrather J, Zhou P, Iadecola C. Angiotensin II attenuates endothelium-dependent responses in the cerebral microcirculation through NOX-2-derived radicals. Arterioscler Thromb Vasc Biol. 2006;26:826–32.
Wiesmann M, Roelofs M, van der Lugt R, Heerschap A, Kiliaan AJ, Claassen JA. Angiotensin II, hypertension and angiotensin II receptor antagonism: roles in the behavioural and brain pathology of a mouse model of Alzheimer’s disease. J Cereb Blood Flow Metab. 2017;37:2396–413.
Xue Y, Tang J, Zhang M, He Y, Fu J, Ding F. Durative sleep fragmentation with or without hypertension suppress rapid eye movement sleep and generate cerebrovascular dysfunction. Neurobiol Dis. 2023;184:106222.
Capone C, Faraco G, Park L, Cao X, Davisson RL, Iadecola C. The cerebrovascular dysfunction induced by slow pressor doses of angiotensin II precedes the development of hypertension. Am J Physiol Heart Circ Physiol. 2011;300:H397–407.
Capone C, Faraco G, Peterson JR, Coleman C, Anrather J, Milner TA, et al. Central cardiovascular circuits contribute to the neurovascular dysfunction in angiotensin II hypertension. J Neurosci. 2012;32:4878–86.
Faraco G, Sugiyama Y, Lane D, Garcia-Bonilla L, Chang H, Santisteban MM, et al. Perivascular macrophages mediate the neurovascular and cognitive dysfunction associated with hypertension. J Clin Invest. 2016;126:4674–89.
Iadecola C, Gottesman RF. Neurovascular and cognitive dysfunction in hypertension. Circ Res. 2019;124:1025–44.
Chen X, Gao R, Song Y, Xu T, Jin L, Zhang W, et al. Astrocytic AT1R deficiency ameliorates Abeta-induced cognitive deficits and synaptotoxicity through beta-arrestin2 signaling. Prog Neurobiol. 2023;228:102489.
Hendrickx JO, Calus E, De Deyn PP, Van Dam D, De Meyer GRY Short-term pharmacological induction of arterial stiffness and hypertension with angiotensin II does not affect learning and memory and cerebral amyloid load in two murine models of Alzheimer’s disease. Int J Mol Sci. 2022;23
Danielyan L, Klein R, Hanson LR, Buadze M, Schwab M, Gleiter CH, et al. Protective effects of intranasal losartan in the APP/PS1 transgenic mouse model of Alzheimer disease. Rejuvenation Res. 2010;13:195–201.
Drews HJ, Yenkoyan K, Lourhmati A, Buadze M, Kabisch D, Verleysdonk S, et al. Intranasal losartan decreases perivascular beta amyloid, inflammation, and the decline of neurogenesis in hypertensive rats. Neurotherapeutics. 2019;16:725–40.
Coatl-Cuaya H, Tendilla-Beltran H, de Jesus-Vasquez LM, Garces-Ramirez L, Gomez-Villalobos MJ, Flores G. Losartan enhances cognitive and structural neuroplasticity impairments in spontaneously hypertensive rats. J Chem Neuroanat. 2022;120:102061.
Campos GV, de Souza AMA, Ji H, West CA, Wu X, Lee DL, et al. The angiotensin type 1 receptor antagonist losartan prevents ovariectomy-induced cognitive dysfunction and anxiety-like behavior in Long Evans rats. Cell Mol Neurobiol. 2020;40:407–20.
Takeda S, Sato N, Takeuchi D, Kurinami H, Shinohara M, Niisato K, et al. Angiotensin receptor blocker prevented beta-amyloid-induced cognitive impairment associated with recovery of neurovascular coupling. Hypertension. 2009;54:1345–52.
Nakagawa T, Hasegawa Y, Uekawa K, Senju S, Nakagata N, Matsui K, et al. Transient mild cerebral ischemia significantly deteriorated cognitive impairment in a mouse model of Alzheimer’s disease via angiotensin AT1 receptor. Am J Hypertens. 2017;30:141–50.
Sharma B, Singh N. Experimental hypertension induced vascular dementia: pharmacological, biochemical and behavioral recuperation by angiotensin receptor blocker and acetylcholinesterase inhibitor. Pharmacol Biochem Behav. 2012;102:101–8.
Shindo T, Takasaki K, Uchida K, Onimura R, Kubota K, Uchida N, et al. Ameliorative effects of telmisartan on the inflammatory response and impaired spatial memory in a rat model of Alzheimer’s disease incorporating additional cerebrovascular disease factors. Biol Pharm Bull. 2012;35:2141–7.
Liu J, Liu S, Tanabe C, Maeda T, Zou K, Komano H. Differential effects of angiotensin II receptor blockers on Abeta generation. Neurosci Lett. 2014;567:51–6.
Abdelkader NF, Abd El-Latif AM, Khattab MM. Telmisartan/17beta-estradiol mitigated cognitive deficit in an ovariectomized rat model of Alzheimer’s disease: Modulation of ACE1/ACE2 and AT1/AT2 ratio. Life Sci. 2020;245:117388.
Sohn YI, Lee NJ, Chung A, Saavedra JM, Scott Turner R, Pak DT, et al. Antihypertensive drug Valsartan promotes dendritic spine density by altering AMPA receptor trafficking. Biochem Biophys Res Commun. 2013;439:464–70.
Bhat SA, Goel R, Shukla S, Shukla R, Hanif K. Angiotensin receptor blockade by inhibiting glial activation promotes hippocampal neurogenesis via activation of Wnt/beta-catenin signaling in hypertension. Mol Neurobiol. 2018;55:5282–98.
Gouveia F, Fonseca C, Silva A, Camins A, Teresa Cruz M, Ettcheto M, et al. Intranasal irbesartan reverts cognitive decline and activates the PI3K/AKT pathway in an LPS-induced neuroinflammation mice model. Int Immunopharmacol. 2024;128:111471.
Steckelings UM, Paulis L, Namsolleck P, Unger T. AT2 receptor agonists: hypertension and beyond. Curr Opin Nephrol Hypertens. 2012;21:142–6.
Horiuchi M, Mogi M. Role of angiotensin II receptor subtype activation in cognitive function and ischaemic brain damage. Br J Pharm. 2011;163:1122–30.
Iwanami J, Mogi M, Tsukuda K, Wang XL, Nakaoka H, Kan-no H, et al. Direct angiotensin II type 2 receptor stimulation by compound 21 prevents vascular dementia. J Am Soc Hypertens. 2015;9:250–6.
Jing F, Mogi M, Sakata A, Iwanami J, Tsukuda K, Ohshima K, et al. Direct stimulation of angiotensin II type 2 receptor enhances spatial memory. J Cereb Blood Flow Metab. 2012;32:248–55.
Ahmed HA, Ishrat T, Pillai B, Fouda AY, Sayed MA, Eldahshan W, et al. RAS modulation prevents progressive cognitive impairment after experimental stroke: a randomized, blinded preclinical trial. J Neuroinflammation. 2018;15:229.
Dai SY, Peng W, Zhang YP, Li JD, Shen Y, Sun XF. Brain endogenous angiotensin II receptor type 2 (AT2-R) protects against DOCA/salt-induced hypertension in female rats. J Neuroinflammation. 2015;12:47.
Sakata A, Mogi M, Iwanami J, Tsukuda K, Min LJ, Fujita T, et al. Sex-different effect of angiotensin II type 2 receptor on ischemic brain injury and cognitive function. Brain Res. 2009;1300:14–23.
Eldahshan W, Sayed MA, Awad ME, Ahmed HA, Gillis E, Althomali W, et al. Stimulation of angiotensin II receptor 2 preserves cognitive function and is associated with an enhanced cerebral vascular density after stroke. Vasc Pharm. 2021;141:106904.
Alshammari A, Pillai B, Kamat P, Jones TW, Bosomtwi A, Khan MB, et al. Angiotensin II type 2 receptor agonism alleviates progressive post-stroke cognitive impairment in aged spontaneously hypertensive rats. Transl Stroke Res. 2025;16:584–99.
Royea J, Lacalle-Aurioles M, Trigiani LJ, Fermigier A, Hamel E. AT2R’s (angiotensin II type 2 receptor’s) role in cognitive and cerebrovascular deficits in a mouse model of Alzheimer disease. Hypertension. 2020;75:1464–74.
Fernando RN, Albiston AL, Chai SY. The insulin-regulated aminopeptidase IRAP is colocalised with GLUT4 in the mouse hippocampus-potential role in modulation of glucose uptake in neurones?. Eur J Neurosci. 2008;28:588–98.
Ramirez-Exposito MJ, Martinez-Martos JM, Canton-Habas V, Del Pilar Carrera-Gonzalez M. Putative involvement of endocrine disruptors in the Alzheimer’s disease via the insulin-regulated aminopeptidase/GLUT4 pathway. Curr Neuropharmacol. 2021;19:939–56.
Zhou Z, Orchard SG, Nelson MR, Fravel MA, Ernst ME. Angiotensin receptor blockers and cognition: a scoping review. Curr Hypertens Rep. 2024;26:1–19.
Gouveia F, Camins A, Ettcheto M, Bicker J, Falcao A, Cruz MT, et al. Targeting brain Renin-Angiotensin System for the prevention and treatment of Alzheimer’s disease: past, present and future. Ageing Res Rev. 2022;77:101612.
Glodzik L, Santisteban MM. Blood-brain barrier crossing renin-angiotensin system drugs: considerations for dementia and cognitive decline. Hypertension. 2021;78:644–6.
Jiang T, Zhang YD, Zhou JS, Zhu XC, Tian YY, Zhao HD, et al. Angiotensin-(1-7) is reduced and inversely correlates with tau hyperphosphorylation in animal models of Alzheimer’s disease. Mol Neurobiol. 2016;53:2489–97.
Evans CE, Miners JS, Piva G, Willis CL, Heard DM, Kidd EJ, et al. ACE2 activation protects against cognitive decline and reduces amyloid pathology in the Tg2576 mouse model of Alzheimer’s disease. Acta Neuropathol. 2020;139:485–502.
Cao C, Hasegawa Y, Hayashi K, Takemoto Y, Kim-Mitsuyama S. Chronic angiotensin 1-7 infusion prevents angiotensin-II-induced cognitive dysfunction and skeletal muscle injury in a mouse model of Alzheimer’s disease. J Alzheimers Dis. 2019;69:297–309.
Hay M, Polt R, Heien ML, Vanderah TW, Largent-Milnes TM, Rodgers K, et al. A Novel angiotensin-(1-7) glycosylated Mas receptor agonist for treating vascular cognitive impairment and inflammation-related memory dysfunction. J Pharm Exp Ther. 2019;369:9–25.
Stoyell-Conti FF, Chabbra A, Puthentharayil J, Rigatto K, Speth RC. Chronic administration of pharmacological doses of angiotensin 1-7 and iodoangiotensin 1-7 has minimal effects on blood pressure, heart rate, and cognitive function of spontaneously hypertensive rats. Physiol Rep. 2021;9:e14812.
Higaki A, Mogi M, Iwanami J, Min LJ, Bai HY, Shan BS, et al. Beneficial effect of Mas receptor deficiency on vascular cognitive impairment in the presence of angiotensin II type 2 receptor. J Am Heart Assoc. 2018;7
Tran S, Kuruppu S, Rajapakse NW. Chronic renin-angiotensin system activation induced neuroinflammation: common mechanisms underlying hypertension and dementia?. J Alzheimers Dis. 2022;85:943–55.
Sharma B, Singh N. Defensive effect of natrium diethyldithiocarbamate trihydrate (NDDCT) and lisinopril in DOCA-salt hypertension-induced vascular dementia in rats. Psychopharmacology (Berl). 2012;223:307–17.
Collu R, Giunti E, Daley S, Chen M, Xia W. Angiotensin-converting enzyme inhibitors and statins therapies-induced changes in omics profiles in humans and transgenic tau mice. Biomed Pharmacother. 2023;168:115756.
Koronyo-Hamaoui M, Shah K, Koronyo Y, Bernstein E, Giani JF, Janjulia T, et al. ACE overexpression in myelomonocytic cells: effect on a mouse model of Alzheimer’s disease. Curr Hypertens Rep. 2014;16:444.
Veiras LC, Cao D, Saito S, Peng Z, Bernstein EA, Shen JZY, et al. Overexpression of ACE in myeloid cells increases immune effectiveness and leads to a new way of considering inflammation in acute and chronic diseases. Curr Hypertens Rep. 2020;22:4.
Cao D, Veiras L, Ahmed F, Shibata T, Bernstein EA, Okwan-Duodu D, et al. The non-cardiovascular actions of ACE. Peptides. 2022;152:170769.
Wang X, Zhu Y, Wang S, Wang Z, Sun H, He Y, et al. Effects of eplerenone on cerebral aldosterone levels and brain lesions in spontaneously hypertensive rats. Clin Exp Hypertens. 2020;42:531–8.
Shamsi TN, Parveen R, Fatima S. Characterization, biomedical and agricultural applications of protease inhibitors: a review. Int J Biol Macromol. 2016;91:1120–33.
Nalivaeva NN, Zhuravin IA, Turner AJ. Neprilysin expression and functions in development, ageing and disease. Mech Ageing Dev. 2020;192:111363.
Iwata N, Tsubuki S, Takaki Y, Shirotani K, Lu B, Gerard NP, et al. Metabolic regulation of brain Abeta by neprilysin. Science. 2001;292:1550–2.
Zhang H, Liu D, Wang Y, Huang H, Zhao Y, Zhou H. Meta-analysis of expression and function of neprilysin in Alzheimer’s disease. Neurosci Lett. 2017;657:69–76.
Langenickel TH, Tsubouchi C, Ayalasomayajula S, Pal P, Valentin MA, Hinder M, et al. The effect of LCZ696 (sacubitril/valsartan) on amyloid-beta concentrations in cerebrospinal fluid in healthy subjects. Br J Clin Pharm. 2016;81:878–90.
Brum WS, Docherty KF, Ashton NJ, Zetterberg H, Hansson O, McMurray JJV, et al. Effect of neprilysin inhibition on Alzheimer disease plasma biomarkers: a secondary analysis of a randomized clinical trial. JAMA Neurol. 2024;81:197–200.
Dewan P, Shen L, Pedro Ferreira J, Jhund PS, Anand IS, Chandra A, et al. Effect of sacubitril/valsartan on cognitive function in patients with heart failure with preserved ejection fraction: a prespecified analysis of PARAGON-HF. Circulation. 2024;150:272–82.
Bartsch T, Wulff P. The hippocampus in aging and disease: from plasticity to vulnerability. Neuroscience. 2015;309:1–16.
Bink DI, Ritz K, Aronica E, van der Weerd L, Daemen MJ. Mouse models to study the effect of cardiovascular risk factors on brain structure and cognition. J Cereb Blood Flow Metab. 2013;33:1666–84.
Diemer NH, Valente E, Bruhn T, Berg M, Jorgensen MB, Johansen FF. Glutamate receptor transmission and ischemic nerve cell damage: evidence for involvement of excitotoxic mechanisms. Prog Brain Res. 1993;96:105–23.
Johnson AC, Li Z, Orfila JE, Herson PS, Cipolla MJ. Hippocampal network dysfunction as a mechanism of early-onset dementia after preeclampsia and eclampsia. Prog Neurobiol. 2021;199:101938.
Lee J, Kim HJ. Normal aging induces changes in the brain and neurodegeneration progress: review of the structural, biochemical, metabolic, cellular, and molecular changes. Front Aging Neurosci. 2022;14:931536.
Toth P, Tarantini S, Springo Z, Tucsek Z, Gautam T, Giles CB, et al. Aging exacerbates hypertension-induced cerebral microhemorrhages in mice: role of resveratrol treatment in vasoprotection. Aging Cell. 2015;14:400–8.
Bennett HC, Zhang Q, Wu YT, Manjila SB, Chon U, Shin D, et al. Aging drives cerebrovascular network remodeling and functional changes in the mouse brain. Nat Commun. 2024;15:6398.
Sure VN, Sakamuri S, Sperling JA, Evans WR, Merdzo I, Mostany R, et al. A novel high-throughput assay for respiration in isolated brain microvessels reveals impaired mitochondrial function in the aged mice. Geroscience. 2018;40:365–75.
Kemper TL, Blatt GJ, Killiany RJ, Moss MB. Neuropathology of progressive cognitive decline in chronically hypertensive rhesus monkeys. Acta Neuropathol. 2001;101:145–53.
Kaiser D, Weise G, Moller K, Scheibe J, Posel C, Baasch S, et al. Spontaneous white matter damage, cognitive decline and neuroinflammation in middle-aged hypertensive rats: an animal model of early-stage cerebral small vessel disease. Acta Neuropathol Commun. 2014;2:169.
Ritz MF, Grond-Ginsbach C, Engelter S, Lyrer P. Gene expression suggests spontaneously hypertensive rats may have altered metabolism and reduced hypoxic tolerance. Curr Neurovasc Res. 2012;9:10–9.
Mortensen KN, Sanggaard S, Mestre H, Lee H, Kostrikov S, Xavier ALR, et al. Impaired glymphatic transport in spontaneously hypertensive rats. J Neurosci. 2019;39:6365–77.
Monte B, Constantinou S, Koundal S, Lee H, Dai F, Gursky Z, et al. Characterization of perivascular space pathology in a rat model of cerebral small vessel disease by in vivo magnetic resonance imaging. J Cereb Blood Flow Metab. 2022;42:1813–26.
Koundal S, Liu X, Sanggaard S, Mortensen K, Wardlaw J, Nedergaard M, et al. Brain morphometry and longitudinal relaxation time of spontaneously hypertensive rats (SHRs) in early and intermediate stages of hypertension investigated by 3D VFA-SPGR MRI. Neuroscience. 2019;404:14–26.
Venkat P, Chopp M, Chen J. Models and mechanisms of vascular dementia. Exp Neurol. 2015;272:97–108.
Ueno M, Sakamoto H, Tomimoto H, Akiguchi I, Onodera M, Huang CL, et al. Blood-brain barrier is impaired in the hippocampus of young adult spontaneously hypertensive rats. Acta Neuropathol. 2004;107:532–8.
Xu X, Xiao C, Yi M, Yang J, Liao M, Zhou K, et al. Cerebral perfusion characteristics and dynamic brain structural changes in stroke-prone renovascular hypertensive rats: a preclinical model for cerebral small vessel disease. Transl Stroke Res. 2024, (e-pub ahead of print 20240305;https://doi.org/10.1007/s12975-024-01239-8).
Roberts R, Huckstepp RTR. Innate sleep apnea in spontaneously hypertensive rats is associated with microvascular rarefaction and neuronal loss in the preBotzinger complex. Stroke. 2023;54:3141–52.
Helman AM, Murphy MP. Vascular cognitive impairment: Modeling a critical neurologic disease in vitro and in vivo. Biochim Biophys Acta. 2016;1862:975–82.
Choi JY, Cui Y, Kim BG. Interaction between hypertension and cerebral hypoperfusion in the development of cognitive dysfunction and white matter pathology in rats. Neuroscience. 2015;303:115–25.
Valerio Romanini C, Dias Fiuza Ferreira E, Correia Bacarin C, Verussa MH, Weffort de Oliveira RM, Milani H. Neurohistological and behavioral changes following the four-vessel occlusion/internal carotid artery model of chronic cerebral hypoperfusion: comparison between normotensive and spontaneously hypertensive rats. Behav Brain Res. 2013;252:214–21.
Yang Y, Kimura-Ohba S, Thompson JF, Salayandia VM, Cosse M, Raz L, et al. Vascular tight junction disruption and angiogenesis in spontaneously hypertensive rat with neuroinflammatory white matter injury. Neurobiol Dis. 2018;114:95–110.
Kitamura A, Saito S, Maki T, Oishi N, Ayaki T, Hattori Y, et al. Gradual cerebral hypoperfusion in spontaneously hypertensive rats induces slowly evolving white matter abnormalities and impairs working memory. J Cereb Blood Flow Metab. 2016;36:1592–602.
Raz L, Bhaskar K, Weaver J, Marini S, Zhang Q, Thompson JF, et al. Hypoxia promotes tau hyperphosphorylation with associated neuropathology in vascular dysfunction. Neurobiol Dis. 2019;126:124–36.
Guo S, Deng W, Xing C, Zhou Y, Ning M, Lo EH. Effects of aging, hypertension and diabetes on the mouse brain and heart vasculomes. Neurobiol Dis. 2019;126:117–23.
Hainsworth AH, Allan SM, Boltze J, Cunningham C, Farris C, Head E, et al. Translational models for vascular cognitive impairment: a review including larger species. BMC Med. 2017;15:16.
Wiesmann M, Zerbi V, Jansen D, Lutjohann D, Veltien A, Heerschap A, et al. Hypertension, cerebrovascular impairment, and cognitive decline in aged AbetaPP/PS1 mice. Theranostics. 2017;7:1277–89.
Cifuentes D, Poittevin M, Dere E, Broqueres-You D, Bonnin P, Benessiano J, et al. Hypertension accelerates the progression of Alzheimer-like pathology in a mouse model of the disease. Hypertension. 2015;65:218–24.
de Montgolfier O, Pincon A, Pouliot P, Gillis MA, Bishop J, Sled JG, et al. High systolic blood pressure induces cerebral microvascular endothelial dysfunction, neurovascular unit damage, and cognitive decline in mice. Hypertension. 2019;73:217–28.
Helms HC, Abbott NJ, Burek M, Cecchelli R, Couraud PO, Deli MA, et al. In vitro models of the blood-brain barrier: An overview of commonly used brain endothelial cell culture models and guidelines for their use. J Cereb Blood Flow Metab. 2016;36:862–90.
Ko E, Kamm RD. Neurovascular models for organ-on-a-chips. vitro models. 2022;1:125–7.
Suo Z, Su G, Placzek A, Kundtz A, Humphrey J, Crawford F, et al. A beta vasoactivity in vivo. Ann N. Y Acad Sci. 2000;903:156–63.
Paris D, Humphrey J, Quadros A, Patel N, Crescentini R, Crawford F, et al. Vasoactive effects of A beta in isolated human cerebrovessels and in a transgenic mouse model of Alzheimer’s disease: role of inflammation. Neurol Res. 2003;25:642–51.
Haase N, Herse F, Spallek B, Haase H, Morano I, Qadri F, et al. Amyloid-beta peptides activate alpha1-adrenergic cardiovascular receptors. Hypertension. 2013;62:966–72.
Tayler HM, Palmer JC, Thomas TL, Kehoe PG, Paton JF, Love S. Cerebral Abeta(40) and systemic hypertension. J Cereb Blood Flow Metab. 2018;38:1993–2005.
Yu JT, Wang ND, Ma T, Jiang H, Guan J, Tan L. Roles of beta-adrenergic receptors in Alzheimer’s disease: implications for novel therapeutics. Brain Res Bull. 2011;84:111–7.
Palmer JC, Tayler HM, Dyer L, Kehoe PG, Paton JFR, Love S. Zibotentan, an endothelin A receptor antagonist, prevents amyloid-beta-induced hypertension and maintains cerebral perfusion. J Alzheimers Dis. 2020;73:1185–99.
Yamada K, Uchida S, Takahashi S, Takayama M, Nagata Y, Suzuki N, et al. Effect of a centrally active angiotensin-converting enzyme inhibitor, perindopril, on cognitive performance in a mouse model of Alzheimer’s disease. Brain Res. 2010;1352:176–86.
Torika N, Asraf K, Roasso E, Danon A, Fleisher-Berkovich S. Angiotensin converting enzyme inhibitors ameliorate brain inflammation associated with microglial activation: possible implications for Alzheimer’s disease. J Neuroimmune Pharm. 2016;11:774–85.
Trigiani LJ, Royea J, Lacalle-Aurioles M, Tong XK, Hamel E. Pleiotropic benefits of the angiotensin receptor blocker candesartan in a mouse model of Alzheimer disease. Hypertension. 2018;72:1217–26.
Tsukuda K, Mogi M, Iwanami J, Min LJ, Sakata A, Jing F, et al. Cognitive deficit in amyloid-beta-injected mice was improved by pretreatment with a low dose of telmisartan partly because of peroxisome proliferator-activated receptor-gamma activation. Hypertension. 2009;54:782–7.
Fu XX, Wei B, Cao HM, Duan R, Deng Y, Lian HW, et al. Telmisartan alleviates Alzheimer’s disease-related neuropathologies and cognitive impairments. J Alzheimers Dis. 2023;94:919–33.
Kishi T, Hirooka Y, Sunagawa K. Telmisartan protects against cognitive decline via up-regulation of brain-derived neurotrophic factor/tropomyosin-related kinase B in hippocampus of hypertensive rats. J Cardiol. 2012;60:489–94.
Kurata T, Lukic V, Kozuki M, Wada D, Miyazaki K, Morimoto N, et al. Telmisartan reduces progressive accumulation of cellular amyloid beta and phosphorylated tau with inflammatory responses in aged spontaneously hypertensive stroke resistant rat. J Stroke Cerebrovasc Dis. 2014;23:2580–90.
Liu W, Yamashita T, Kurata T, Kono S, Hishikawa N, Deguchi K, et al. Protective effect of telmisartan on neurovascular unit and inflammasome in stroke-resistant spontaneously hypertensive rats. Neurol Res. 2015;37:491–501.
Zhai Y, Yamashita T, Kurata T, Fukui Y, Sato K, Kono S, et al. Strong reduction of low-density lipoprotein receptor/apolipoprotein E expressions by telmisartan in cerebral cortex and hippocampus of stroke resistant spontaneously hypertensive rats. J Stroke Cerebrovasc Dis. 2014;23:2350–61.
Yamada K, Horita T, Takayama M, Takahashi S, Takaba K, Nagata Y, et al. Effect of a centrally active angiotensin converting enzyme inhibitor, perindopril, on cognitive performance in chronic cerebral hypo-perfusion rats. Brain Res. 2011;1421:110–20.
Kono S, Kurata T, Sato K, Omote Y, Hishikawa N, Yamashita T, et al. Neurovascular protection by telmisartan via reducing neuroinflammation in stroke-resistant spontaneously hypertensive rat brain after ischemic stroke. J Stroke Cerebrovasc Dis. 2015;24:537–47.
Kurata T, Lukic V, Kozuki M, Wada D, Miyazaki K, Morimoto N, et al. Long-term effect of telmisartan on Alzheimer’s amyloid genesis in SHR-SR after tMCAO. Transl Stroke Res. 2015;6:107–15.
Gao Y, Li W, Liu Y, Wang Y, Zhang J, Li M, et al. Effect of telmisartan on preventing learning and memory deficits via peroxisome proliferator-activated receptor-gamma in vascular dementia spontaneously hypertensive rats. J Stroke Cerebrovasc Dis. 2018;27:277–85.
Rodriguez-Ortiz CJ, Thorwald MA, Rodriguez R, Mejias-Ortega M, Kieu Z, Maitra N, et al. Angiotensin receptor blockade with olmesartan alleviates brain pathology in obese OLETF rats. Clin Exp Pharm Physiol. 2023;50:228–37.
Goel R, Bhat SA, Hanif K, Nath C, Shukla R. Perindopril attenuates lipopolysaccharide-induced amyloidogenesis and memory impairment by suppression of oxidative stress and RAGE activation. ACS Chem Neurosci. 2016;7:206–17.
Yoo S, Stremlau M, Pinto A, Woo H, Curtis O, van Praag H. Effects of combined anti-hypertensive and statin treatment on memory, fear extinction, adult neurogenesis, and angiogenesis in adult and middle-aged mice. Cells. 2021;10:1778.
Mamo JCL, Lam V, Giles C, Coulson SH, Fimognari N, Mooranian A, et al. Antihypertensive agents do not prevent blood-brain barrier dysfunction and cognitive deficits in dietary-induced obese mice. Int J Obes (Lond). 2017;41:926–34.
Liu X, Wang Z, Xia Y, Yu G, Zeng K, Luo H, et al. Losartan-induced hypotension leads to tau hyperphosphorylation and memory deficit. J Alzheimers Dis. 2014;40:419–27.
Liu S, Ando F, Fujita Y, Liu J, Maeda T, Shen X, et al. A clinical dose of angiotensin-converting enzyme (ACE) inhibitor and heterozygous ACE deletion exacerbate Alzheimer’s disease pathology in mice. J Biol Chem. 2019;294:9760–70.
Wang J, Zhao Z, Lin E, Zhao W, Qian X, Freire D, et al. Unintended effects of cardiovascular drugs on the pathogenesis of Alzheimer’s disease. PLoS One. 2013;8:e65232.
Hemming ML, Selkoe DJ, Farris W. Effects of prolonged angiotensin-converting enzyme inhibitor treatment on amyloid beta-protein metabolism in mouse models of Alzheimer disease. Neurobiol Dis. 2007;26:273–81.
Paris D, Bachmeier C, Patel N, Quadros A, Volmar CH, Laporte V, et al. Selective antihypertensive dihydropyridines lower Aβ accumulation by targeting both the production and the clearance of Aβ across the blood-brain barrier. Mol Med. 2011;17:149–62.
Amenta F, Tomassoni D. Treatment with nicardipine protects brain in an animal model of hypertension-induced damage. Clin Exp Hypertens. 2004;26:351–61.
Kerkhofs D, Helgers R, Hermes D, Steinbusch HPJ, Van Essen H, Leenders P, et al. Amlodipine limits microglia activation and cognitive dysfunction in aged hypertensive mice. J Hypertens. 2023;41:1159–67.
Sabbatini M, Tomassoni D, Amenta F. Hypertensive brain damage: comparative evaluation of protective effect of treatment with dihydropyridine derivatives in spontaneously hypertensive rats. Mech Ageing Dev. 2001;122:2085–105.
Morin A, Mouzon B, Ferguson S, Paris D, Browning M, Stewart W, et al. Nilvadipine suppresses inflammation via inhibition of P-SYK and restores spatial memory deficits in a mouse model of repetitive mild TBI. Acta Neuropathol Commun. 2020;8:166.
Sakurai-Yamashita Y, Harada N, Niwa M. Lercanidipine rescues hippocampus pyramidal neurons from mild ischemia-induced delayed neuronal death in SHRSP. Cell Mol Neurobiol. 2011;31:561–7.
Naessens DMP, de Vos J, Richard E, Wilhelmus MMM, Jongenelen CAM, Scholl ER, et al. Effect of long-term antihypertensive treatment on cerebrovascular structure and function in hypertensive rats. Sci Rep. 2023;13:3481.
Dobarro M, Orejana L, Aguirre N, Ramirez MJ. Propranolol reduces cognitive deficits, amyloid beta levels, tau phosphorylation and insulin resistance in response to chronic corticosterone administration. Int J Neuropsychopharmacol. 2013;16:1351–60.
Wang J, Wright HM, Vempati P, Li H, Wangsa J, Dzhuan A, et al. Investigation of nebivolol as a novel therapeutic agent for the treatment of Alzheimer’s disease. J Alzheimers Dis. 2013;33:1147–56.
Evans AK, Ardestani PM, Yi B, Park HH, Lam RK, Shamloo M. Beta-adrenergic receptor antagonism is proinflammatory and exacerbates neuroinflammation in a mouse model of Alzheimer’s Disease. Neurobiol Dis. 2020;146:105089.
Zhang L, Chan LuL, Huang WM, Wai Y, Yew MS. DT. Effects of DL-3-n-butylphthalide on vascular dementia and angiogenesis. Neurochem Res. 2012;37:911–9.
Ho SC, Ho YF, Lai TH, Liu TH, Su SY, Wu RY. Effect of Tianma Gouteng Decoction with subtractive ingredients and its active constituents on memory acquisition. Am J Chin Med. 2008;36:593–602.
Huang SW, Wang W, Zhang MY, Liu QB, Luo SY, Peng Y, et al. The effect of ethyl acetate extract from persimmon leaves on Alzheimer’s disease and its underlying mechanism. Phytomedicine. 2016;23:694–704.
Fei YL, Lv HJ, Li YB, Liu J, Qian YH, Yang WN, et al. Tongxinluo improves cognition by decreasing beta-amyloid in spontaneous hypertensive rats. Brain Res. 2017;1663:151–60.
Watanabe H, Zhao Q, Matsumoto K, Tohda M, Murakami Y, Zhang SH, et al. Pharmacological evidence for antidementia effect of Choto-san (Gouteng-san), a traditional Kampo medicine. Pharmacol Biochem Behav. 2003;75:635–43.
Wei M, Chen L, Liu J, Zhao J, Liu W, Feng F. Protective effects of a Chotosan Fraction and its active components on beta-amyloid-induced neurotoxicity. Neurosci Lett. 2016;617:143–9.
Hussein G, Nakamura M, Zhao Q, Iguchi T, Goto H, Sankawa U, et al. Antihypertensive and neuroprotective effects of astaxanthin in experimental animals. Biol Pharm Bull. 2005;28:47–52.
Liu TH, Tsai TY. Effects of equol on deoxycorticosterone acetate salt-induced hypertension and associated vascular dementia in rats. Food Funct. 2016;7:3444–57.
Wang J, Ho L, Chen L, Zhao Z, Zhao W, Qian X, et al. Valsartan lowers brain beta-amyloid protein levels and improves spatial learning in a mouse model of Alzheimer disease. J Clin Invest. 2007;117:3393–402.
Pauls E, Bayod S, Mateo L, Alcalde V, Juan-Blanco T, Sanchez-Soto M, et al. Identification and drug-induced reversion of molecular signatures of Alzheimer’s disease onset and progression in App(NL-G-F), App(NL-F), and 3xTg-AD mouse models. Genome Med. 2021;13:168.
Siddiqi FH, Menzies FM, Lopez A, Stamatakou E, Karabiyik C, Ureshino R, et al. Felodipine induces autophagy in mouse brains with pharmacokinetics amenable to repurposing. Nat Commun. 2019;10:1817.
Michaels NJ, Lemmon K, Plemel JR, Jensen SK, Mishra MK, Brown D, et al. Aging-exacerbated acute axon and myelin injury is associated with microglia-derived reactive oxygen species and is alleviated by the generic medication indapamide. J Neurosci. 2020;40:8587–600.
Teng T, Ridgley DM, Tsoy A, Sun GY, Askarova S, Lee JC. Azelnidipine attenuates the oxidative and NFkappaB pathways in amyloid-beta-stimulated cerebral endothelial cells. ACS Chem Neurosci. 2019;10:209–15.
Choi DH, Lee J. A mini-review of the NADPH oxidases in vascular dementia: correlation with NOXs and risk factors for VaD. Int J Mol Sci. 2017;18
Olasehinde TA, Oladapo SFAK, Oyeleye IF. SI. Effect of diet supplemented with African Star Apple Fruit Pulp on purinergic, cholinergic and monoaminergic enzymes, TNF-alpha expression and redox imbalance in the brain of hypertensive rats. Nutr Neurosci. 2023;26:496–510.
Taylor JL, Walsh KR, Mosneag IE, Danby TGE, Luka N, Chanda B, et al. Uncoupling of Ca(2+) sparks from BK channels in cerebral arteries underlies hypoperfusion in hypertension-induced vascular dementia. Proc Natl Acad Sci USA. 2023;120:e2307513120.
Terry AV Jr., Hernandez CM, Buccafusco JJ, Gattu M. Deficits in spatial learning and nicotinic-acetylcholine receptors in older, spontaneously hypertensive rats. Neuroscience. 2000;101:357–68.
Tayebati SK, Di Tullio MA, Amenta F. Effect of treatment with the cholinesterase inhibitor rivastigmine on vesicular acetylcholine transporter and choline acetyltransferase in rat brain. Clin Exp Hypertens. 2004;26:363–73.
Tayebati SK, Di Tullio MA, Tomassoni D, Amenta F. Neuroprotective effect of treatment with galantamine and choline alphoscerate on brain microanatomy in spontaneously hypertensive rats. J Neurol Sci. 2009;283:187–94.
Tomassoni D, Avola R, Mignini F, Parnetti L, Amenta F. Effect of treatment with choline alphoscerate on hippocampus microanatomy and glial reaction in spontaneously hypertensive rats. Brain Res. 2006;1120:183–90.
Tomassoni D, Catalani A, Cinque C, Di Tullio MA, Tayebati SK, Cadoni A, et al. Effects of cholinergic enhancing drugs on cholinergic transporters in the brain and peripheral blood lymphocytes of spontaneously hypertensive rats. Curr Alzheimer Res. 2012;9:120–7.
Avolio E, Mahata SK, Mantuano E, Mele M, Alo R, Facciolo RM, et al. Antihypertensive and neuroprotective effects of catestatin in spontaneously hypertensive rats: interaction with GABAergic transmission in amygdala and brainstem. Neuroscience. 2014;270:48–57.
Nakazaki M, Sasaki M, Kataoka-Sasaki Y, Oka S, Suzuki J, Sasaki Y, et al. Intravenous infusion of mesenchymal stem cells improves impaired cognitive function in a cerebral small vessel disease model. Neuroscience. 2019;408:361–77.
Karakas E, Simorowski N, Furukawa H. Subunit arrangement and phenylethanolamine binding in GluN1/GluN2B NMDA receptors. Nature. 2011;475:249–53.
Benicky J, Sanchez-Lemus E, Honda M, Pang T, Orecna M, Wang J, et al. Angiotensin II AT1 receptor blockade ameliorates brain inflammation. Neuropsychopharmacology. 2011;36:857–70.
Xu X, Du L, Jiang J, Yang M, Wang Z, Wang Y, et al. Microglial TREM2 mitigates inflammatory responses and neuronal apoptosis in angiotensin II-induced hypertension in middle-aged mice. Front Aging Neurosci. 2021;13:716917.
Tang Y, Zhang D, Chang Y, Zheng J. Atrial natriuretic peptide associated with cardiovascular diseases inhibits amyloid-beta aggregation via cross-seeding. ACS Chem Neurosci. 2023;14:312–22.
Bretland KA, Lin L, Bretland KM, Smith MA, Fleming SM, Dengler-Crish CM. Irisin treatment lowers levels of phosphorylated tau in the hippocampus of pre-symptomatic female but not male htau mice. Neuropathol Appl Neurobiol. 2021;47:967–78.
Boyarko B, Podvin S, Greenberg B, Momper JD, Huang Y, Gerwick WH, et al. Evaluation of bumetanide as a potential therapeutic agent for Alzheimer’s disease. Front Pharm. 2023;14:1190402.
Sharma P, Gaur N, Jayant S, Sharma BM, Singh B, Kharkwal H, et al. Salubrious effects of ulinastatin and quercetin alone or in combination in endothelial dysfunction and vascular dementia. Pharmacol Rep. 2022;74:481–92.
Singh P, Gupta S, Sharma B. Melatonin receptor and KATP channel modulation in experimental vascular dementia. Physiol Behav. 2015;142:66–78.
Singh P, Gupta S, Sharma B. Antagonism of endothelin (ETA and ETB) receptors during renovascular hypertension-induced vascular dementia improves cognition. Curr Neurovasc Res. 2016;13:219–29.
Singh P, Sharma B. Selective sserotonin-norepinephrine re-uptake inhibition limits renovascular-hypertension induced cognitive impairment, endothelial dysfunction, and oxidative stress injury. Curr Neurovasc Res. 2016;13:135–46.
Pacheco-Quinto J, Herdt A, Eckman CB, Eckman EA. Endothelin-converting enzymes and related metalloproteases in Alzheimer’s disease. J Alzheimers Dis. 2013;33:S101–10.
Panchal M, Rholam M, Brakch N. Abnormalities of peptide metabolism in Alzheimer disease. Curr Neurovasc Res. 2004;1:317–23.
Poorgolizadeh E, Homayouni Moghadam F, Dormiani K, Rezaei N, Nasr-Esfahani MH. Do neprilysin inhibitors walk the line? Heart ameliorative but brain threatening!. Eur J Pharm. 2021;894:173851.
Yadav DK. Potential therapeutic strategies of phytochemicals in neurodegenerative disorders. Curr Top Med Chem. 2021;21:2814–38.
Zisapel N. New perspectives on the role of melatonin in human sleep, circadian rhythms and their regulation. Br J Pharm. 2018;175:3190–9.
Ghafouri-Fard S, Shoorei H, Bahroudi Z, Abak A, Majidpoor J, Taheri M. An update on the role of miR-124 in the pathogenesis of human disorders. Biomed Pharmacother. 2021;135:111198.
Bailey EL, McBride MW, McClure JD, Beattie W, Graham D, Dominiczak AF, et al. Effects of dietary salt on gene and protein expression in brain tissue of a model of sporadic small vessel disease. Clin Sci (Lond). 2018;132:1315–28.
Kleinewietfeld M, Manzel A, Titze J, Kvakan H, Yosef N, Linker RA, et al. Sodium chloride drives autoimmune disease by the induction of pathogenic TH17 cells. Nature. 2013;496:518–22.
Wilck N, Matus MG, Kearney SM, Olesen SW, Forslund K, Bartolomaeus H, et al. Salt-responsive gut commensal modulates T(H)17 axis and disease. Nature. 2017;551:585–9.
Lin B, Hasegawa Y, Takane K, Koibuchi N, Cao C, Kim-Mitsuyama S High-fat-diet intake enhances cerebral amyloid angiopathy and cognitive impairment in a mouse model of Alzheimer’s disease, independently of metabolic disorders. J Am Heart Assoc. 2016;5
Langdon KD, Cordova CA, Granter-Button S, Boyd JD, Peeling J, Murphy TH, et al. Executive dysfunction and blockage of brain microvessels in a rat model of vascular cognitive impairment. J Cereb Blood Flow Metab. 2018;38:1727–40.
Zhou Y, Xie L, Schroder J, Schuster IS, Nakai M, Sun G, et al. Dietary fiber and microbiota metabolite receptors enhance cognition and alleviate disease in the 5xFAD mouse model of Alzheimer’s disease. J Neurosci. 2023;43:6460–75.
Sochocka M, Karska J, Pszczolowska M, Ochnik M, Fulek M, Fulek K, et al. Cognitive decline in early and premature menopause. Int J Mol Sci. 2023;24
Pradhyumnan H, Reddy V, Bassett ZQ, Patel SH, Zhao W, Dave KR, et al. Post-stroke periodic estrogen receptor-beta agonist improves cognition in aged female rats. Neurochem Int. 2023;165:105521.
Drucker DJ. Mechanisms of action and therapeutic application of glucagon-like peptide-1. Cell Metab. 2018;27:740–56.
Zheng J, Xie Y, Ren L, Qi L, Wu L, Pan X, et al. GLP-1 improves the supportive ability of astrocytes to neurons by promoting aerobic glycolysis in Alzheimer’s disease. Mol Metab. 2021;47:101180.
Chung JY, Jeong JH, Song J. Resveratrol modulates the gut-brain axis: Focus on glucagon-like peptide-1, 5-HT, and gut microbiota. Front Aging Neurosci. 2020;12:588044.
Ravindranath V, Dang HM, Goya RG, Mansour H, Nimgaonkar VL, Russell VA, et al. Regional research priorities in brain and nervous system disorders. Nature. 2015;527:S198–206.
Loo G, Puar T, Foo R, Ong TK, Wang TD, Nguyen QN, et al. Unique characteristics of Asians with hypertension: what is known and what can be done?. J Hypertens. 2024;42:1482.
Prevention of Cognitive Impairment by Hypertension Management (PCIHM) Working Group of the Japanese Society of Hypertension
Masaki Mogi1, Hisatomi Arima11, Shintaro Ishida12, Miki Kawazoe11, Takako Fujii11, Yori Inoue11,12, Koichi Yamamoto2, Atsushi Sakima13, Taku Inoue14, Takahiro Komori10, Toshiharu Ninomiya15, Yoichi Nozato2,16, Takuro Kubozono17, Hiroshi Akasaka18, Yume Nohara19, Eiichiro Yamamoto20, Michiaki Nagai21,22, Masafumi Ihara3, Yoichi Takami2, Shuko Takeda6,7, Toru Yamashita4,5
Author information
Authors and Affiliations
Consortia
Corresponding author
Ethics declarations
Conflict of interest
The authors declare no competing interests.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
About this article
Cite this article
Mogi, M., Takami, Y., Ihara, M. et al. Current updates on latest basic research of scientific relevance in hypertensive dementia. Hypertens Res (2025). https://doi.org/10.1038/s41440-025-02417-3
Received:
Revised:
Accepted:
Published:
Version of record:
DOI: https://doi.org/10.1038/s41440-025-02417-3