Next Article in Journal
Effects of Individualised High Positive End-Expiratory Pressure and Crystalloid Administration on Postoperative Pulmonary Function in Patients Undergoing Robotic-Assisted Radical Prostatectomy: A Prospective Randomised Single-Blinded Pilot Study
Previous Article in Journal
The Impact of Asymptomatic Human Immunodeficiency Virus-Positive Disease Status on Inpatient Complications Following Spine Surgery: A Propensity Score-Matched Analysis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Pathogenesis of Chronic Kidney Disease Is Closely Bound up with Alzheimer’s Disease, Especially via the Renin-Angiotensin System

1
The First Affiliated Hospital of Jinan University, Guangzhou 510000, China
2
Central Laboratory, The Fifth Affiliated Hospital of Jinan University, Heyuan 517000, China
3
Institute of Nephrology, Jinan University, Guangzhou 510000, China
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2023, 12(4), 1459; https://doi.org/10.3390/jcm12041459
Submission received: 12 December 2022 / Revised: 1 February 2023 / Accepted: 7 February 2023 / Published: 12 February 2023
(This article belongs to the Section Nephrology & Urology)

Abstract

:
Chronic kidney disease (CKD) is a clinical syndrome secondary to the definitive change in function and structure of the kidney, which is characterized by its irreversibility and slow and progressive evolution. Alzheimer’s disease (AD) is characterized by the extracellular accumulation of misfolded β-amyloid (Aβ) proteins into senile plaques and the formation of neurofibrillary tangles (NFTs) containing hyperphosphorylated tau. In the aging population, CKD and AD are growing problems. CKD patients are prone to cognitive decline and AD. However, the connection between CKD and AD is still unclear. In this review, we take the lead in showing that the development of the pathophysiology of CKD may also cause or exacerbate AD, especially the renin-angiotensin system (RAS). In vivo studies had already shown that the increased expression of angiotensin-converting enzyme (ACE) produces a positive effect in aggravating AD, but ACE inhibitors (ACEIs) have protective effects against AD. Among the possible association of risk factors in CKD and AD, we mainly discuss the RAS in the systemic circulation and the brain.

1. Introduction

Alzheimer’s disease (AD), a neurodegenerative disease that occurs in old age and pre-senile age, is characterized by progressive cognitive impairment and behavioral impairment [1,2]. Epidemiological survey has estimated that AD comprised 60–80% of ~50 million dementia individuals around the world in 2018, and the number is projected to triple by 2050 [3]. The common risk factors for the development of AD are increased age [4], family history [5], degeneration or vascular dysfunction [6], obesity [7], hypotension or hypertension [8], diabetes [9], hyperlipidemia [10], and the existence of epsilon 4 allele of the apolipoprotein E gene (ApoE4) [4]. Kidney disease is recently proposed as a modifiable risk factor for AD. Studies showed that kidney disease patients expressed a higher level of amyloid precursor protein (APP), a key protein for protein-bound receptor sorting (SorLA). It acts as a central regulator of APP trafficking and processing and is expressed concurrently in neurons (cerebellum, hippocampus, and cortex), renal cells, and gene polymorphism, which is associated with late-onset AD [11,12,13]. Due to the limited effective pharmacotherapies of AD [14,15], it is more and more important to focus on studies based on the prevention against major and modifiable risk factors.
Chronic kidney disease (CKD) is defined in adult patients as a glomerular filtration rate (GFR) of less than 60 mL/min/1.73 m2, or a GFR of greater than 60 mL/min/1.73 m2 but with evidence of equal damage to kidney structures, and an onset time lasting for three months or more [16]. CKD is becoming more and more prevalent (10–13% of the population), irreversible, progressive, and associated with higher cardiovascular risk, stimulating the development of cognitive decline and the progression of AD [17,18,19,20]. Zhang et al. [21] reported that patients with AD in 50,550 CKD individuals were up to 10.8% and the rate has shown an upward tendency year by year. To date, the pathophysiology of AD and the role of CKD in AD progression are not completely clear. It is known that RAS significantly matters in the pathology of CKD [22,23]. Additionally, recent studies have shown that the RAS in the brain potentially contributes to dementia, and inhibitors of this system have been shown to be important, which demonstrated that RAS plays an important role in AD [24]. However, only a few studies have demonstrated the association between CKD and AD from the perspective of the RAS. Therefore, we mainly described the pathophysiology of AD and the role of CKD in AD progression from the aspect of RAS.
In this review, we briefly summarized the progression of the pathophysiology of CKD and AD. Secondly, we summarized the function of the RAS in the systemic circulation and brain. Thirdly, we demonstrated that the progression of the pathophysiology of CKD may be involved in the occurrence and the development of AD, especially from the perspective of the RAS, uremic toxins, erythropoietin (EPO), and extracellular vehicles (EVs). Finally, we discussed whether the pathogenic mechanism of the RAS in AD is associated with the pathogenic mechanism of the RAS in CKD. Understanding the potential mechanism between them may provide the potential possibility for RAS-related drugs to be used to prevent or treat AD.

2. Pathophysiology of Chronic Kidney Disease

Since 1990, the global incidence and prevalence of CKD and the number of associated deaths have increased by 89%, 87%, and 98%, respectively [25]. CKD is caused by abnormal development or injury, inflammation/immune mechanisms, or a toxic insult. Several pathways can perpetuate kidney damage by inducing the hyperfiltration and hypertrophy of remaining nephrons; and by promoting oxidative stress, inflammation, the accumulation of uremic toxins and EPO, vascular calcification, and RAS activation. The perturbation of the balance between pro-and anti-oxidant mechanisms results in metabolic dysregulation and the overproduction of reactive oxygen species (ROS) and/or oxidative end products of lipids, DNA, and proteins, thus leading to oxidative damage in cells, tissues, and organs [16,17,26,27]. The irreversible loss of normal renal tissue function and interstitial fibrosis causes pericytes to transdifferentiate into myofibroblasts, followed by massive extracellular matrix deposition and fibrosis and decreased EPO gene transcription [28,29,30]. The decline in renal function in CKD blocks the excretion of organic compounds, leading to the accumulation of uremic toxins in the body followed by chronic inflammation, endothelial dysfunction, damage to mitochondria, and oxidative stress [30,31,32,33,34,35,36,37,38,39]. The increase in proinflammatory cytokine production, oxidative stress, acidosis, and infections caused by persistent low-grade inflammation results in a dysregulated microvascular response to intrarenal regulators, leading to tubular damage, nephron shedding, and CKD onset [40,41,42,43,44,45,46,47,48]. The imbalance in circulation enhancers and vascular calcification inhibitors in CKD induces vascular calcification and osteogenic differentiation in vascular smooth muscle cells, as well as activating mediators of vascular calcification [47,49,50,51,52]. Glomerulosclerosis decreases downstream peritubular capillary blood flow, causing glomeruli in these areas to secrete excess renin; this further increases circulating angiotensin II (Ang II) levels, which then increases systemic vascular resistance and blood pressure and promotes sodium reabsorption in the proximal tubule and (via aldosterone) the collecting duct [53,54,55,56,57,58]. EVs could play an important role in the development of CKD [36,59]; patients with advanced CKD have elevated levels of platelet-, neutrophil-, erythrocyte-, and endothelial cell-derived EVs [60,61,62]. Platelet-derived EVs are procoagulant and prothrombotic [60,62], and an increased number of endothelial cell-derived EVs was shown to be correlated with endothelial dysfunction, atherosclerosis, and arterial stiffness [63]. The abovementioned processes can lead to alterations in podocytes and changes in glomerular structure and architecture, which can result in the sclerosis of nephrons, further deterioration of renal function, and fibrosis [64].

3. Alzheimer’s Disease Pathogenesis

As life expectancy increases and the global population ages, the prevalence of AD is expected to continue increasing (especially in developing countries), resulting in high disease and economic burdens. The number of people with dementia is projected to reach 152 million by 2050, with the largest increases in low- and middle-income countries [65,66]. Community surveys in China over the past few decades have found a marked increase in AD prevalence [67,68].
The most common cause of AD is the extracellular accumulation of misfolded Aβ proteins into senile plaques and the formation of NFTs containing hyperphosphorylated tau proteins [66,69,70]. Aβ proteins can cause various synaptic defects that have synaptogenic effects and can lead to neuronal death. Meanwhile, Aβ ligands can cause the dysregulation of Ca2+ homeostasis, mitochondrial damage, oxidative stress, altered axonal transport, and glial activation, further leading to synaptic damage and neurotoxicity. Aβ accumulation, in which tau phosphorylation plays a critical role, leads to NFT formation, cell loss, vascular damage, and dementia [71,72,73,74]. Tau oligomers at synapses are toxic to neurons and can cause synaptic damage before NFT formation, triggering neurodegeneration. At the same time, tau hyperphosphorylation causes conformational changes that enable the protein to sequester normal microtubule-associated tau; the resulting aggregates of paired helical filaments can cause synaptic dysfunction [71,72,75]. Both mechanisms may be linked to symptoms of AD including progressive memory loss, cognitive decline, and learning difficulties [76,77]. EVs contribute to the pathology of AD by spreading Aβ and tau, thereby promoting AD progression. Additionally, EVs are potential carriers of pathogenic AD-related proteins that impair neuronal function [78,79,80,81]. RAS activation in the brain; alterations in neurons, microglia, and astrocytes; neuroinflammation; vascular changes, and aging are all risk factors for AD (Figure 1).

4. Renin-Angiotensin System

CKD is closely associated with increased RAS activation [54,56,57,58]. In glomerulosclerosis, there is a decrease in downstream peritubular capillary blood flow [48], which causes glomeruli in these areas to secrete excess renin. This further increases the level of circulating Ang II, which not only has a direct vasoconstrictive effect of increasing systemic vascular resistance and blood pressure but also promotes sodium reabsorption in the proximal tubule and (through aldosterone) the collecting duct [23,53]. With fewer functional glomeruli in CKD patients, the GFR and perfusion pressure must be increased by higher systemic arterial pressure. The overactivation of the sympathetic nervous system in CKD stimulates renin production by renal juxtaglomerular cells, further aggravating kidney damage. In summary, the factors induced by CKD cause excessive activation of the RAS, leading to the upregulation of Ang II receptor type 1 (AT1R) [55]. The specific mechanisms involving the RAS are discussed in detail below.

4.1. RAS in the Systemic Circulation

The RAS is an important humoral regulatory system in the human body. The RAS functions in the circulation as well as in the blood vessel wall, heart, kidney, adrenal gland, and other tissues and it plays an important role in the normal development of the cardiovascular system, cardiovascular homeostasis, the maintenance of electrolyte and fluid balance, and the regulation of blood pressure [82,83].
Angiotensinogen (AGT) and renin are the core elements of the classical RAS pathway. AGT is synthesized in the liver, and renin is an aspartyl protease released by the juxtaglomerular cells of the kidney that cleaves AGT at the amino terminal. This generates the inactive decapeptide angiotensin I (Ang I), which is activated by ACE and hydrolyzed at the C-terminal His–Leu to yield the active octapeptide Ang II, the main effector of the RAS pathway. Ang II is cleaved at the N-terminal Asp residue into the heptapeptide angiotensin III (Ang III) by glutamyl aminopeptidase A(AP-A) but it also converted into Ang (1–7) by carboxypeptidase P-mediated cleavage at the Phe residue, by the monopeptidase ACE2, or the ACE cleavage of Phe–His from Ang (1–9). Ang III can be converted to the hexapeptide Ang IV by membrane alanyl aminopeptidase N, which cleaves the N-terminal Arg of Ang III. Ang IV can be further converted to Ang (3–7) by carboxypeptidase P and prolyl oligopeptides that cleave the Pro–Phe bond. Ang I is biologically inactive. Ang II and Ang III are full agonists at AT1R and AT2R and exert opposite effects. In contrast, Ang IV binds with low affinity to AT1R and AT2R but binds with high affinity and selectivity to AT4R [67,84,85,86,87,88,89,90,91,92].
Ang II has a variety of effects after binding to different Ang II receptors. The binding of Ang II and AT1R can induce a range of physiologic and pathologic effects such as inducing central sympathetic outflow, vasoconstriction, renal sodium reabsorption, and release of aldosterone and arginine vasopressin (AVP) from the adrenal and pituitary glands. Conversely, the binding of Ang II and AT2R can result in vasodilation, apoptosis, cellular proliferation, sympathetic inhibition, decreased sodium reabsorption, and the inhibition of AVP release [84,93,94]. AT1R has two subtypes but their distinct physiologic actions are not fully known. Ang III was shown to decrease sodium reabsorption and exert a cardioprotective effect after binding to AT2R [95]; and Ang IV can affect cognitive function, reduce neuronal apoptosis, and promote inflammation [96,97,98]. Ang 1–7 binds primarily to Mas receptors, counteracting many of the deleterious effects of Ang II [99].

4.2. RAS in the Brain

Unlike the systemic RAS, a local RAS is present in the heart, kidney, lung, liver, and retina [100,101]. However, there is little information available on the expression and regulation of the RAS in the brain [102]. The effects of the RASs in the central nervous system (CNS) were originally considered to be due to the activity of the circulating RAS components acting through the circumventricular organs on neurons to regulate blood pressure and sodium and water homeostasis in the brain [103]. However, using a variety of methodologic approaches including confocal laser microscopy, in situ hybridization, laser microdissection, and PCR, it was demonstrated that the brain has a local RAS independent of the peripheral RAS.
RAS components such as renin, angiotensinogen, ACE, Ang I, Ang II, and RAS-specific second messengers are expressed in the brain [68,104,105,106]. The most highly expressed component in neurons is the renin receptor ATPase H+ transporting accessory protein 2 (Atp6ap2). AGT is expressed in astrocytes, and AGT receptor (AGTR) expression overlaps with that of the Mas receptor (MasR) and Atp6ap2. The main AGTR subtype is expressed in microglia, which also express ACE2 and MasR. AGT, ACE, AGTR, Atp6ap2, and MasR are expressed in oligodendrocytes. There is increasing evidence that AT1R and AT2R are widely distributed in the CNS. Additionally, AGT constitutively produces many neuroactive peptides in astrocytes.
Renin cleaves AGT into Ang I in neurons and astrocytes. ACE converts Ang I into Ang II, which binds to AT1R and AT2R expressed by neurons, astrocytes, oligodendrocytes, and microglia in various brain regions [107]. ACE2 converts Ang II to Ang (1–7), which then binds to MasR. The activation of the Ang II/AT1R axis initiates a cascade of events that promotes oxidative stress, apoptosis, and neuroinflammation in several brain disorders. Angiotensin-II receptor blockers (ARBs), AT2R, and the activation of the ACE2/Ang (1–7)/MasR axis provide strong neuroprotection [108,109]. Ang II is converted to Ang III by aminopeptidase A/B, and then to Ang IV by aminopeptidase B, which can act on AT4R [110,111].

5. Association between CKD and AD

5.1. RAS Linking CKD and AD

Many components of the RAS system such as Ang II and Ang III are also expressed in the brain [112,113] and act via two mechanisms. The activation of the Ang II/AT1R axis initiates a series of events that promote oxidative stress, apoptosis, and neuroinflammation in several brain disorders. Moreover, ARBs, AT2R, and the activation of the ACE2/Ang (1–7)/MasR axis have neuroprotective effects (Figure 2) [111,113]. Glomerulosclerosis and the decrease in effective (perceived) blood flow in CKD patients result in excess renin release, an increase in circulating Ang II levels, and AT1R upregulation [6]. Ang II increases systemic vascular resistance and blood pressure and promotes sodium reabsorption in the proximal tubule and (via aldosterone) the collecting duct [114,115]. In a retrospective clinical cohort study, Douglas Barthold et al. [116] showed that in combination with statin and RAS-acting antihypertensives (AHTs), particularly ARB therapy, they may be more effective at reducing the risk of AD and related dementias. Michael Ouk et al. [117] reported that among ApoE4 non-carriers with AD, ARB use was related to the greater preservation of memory and attention/psychomotor processing speed, particularly compared with ACEI that do not cross the blood-brain-barrier (RR = 1.200, p = 0.003). More and more studies have suggested that RAS activation may be also involved in the development of AD, which is detailed with specific pathogenesis in the following sections (Table 1).

5.2. RAS and Aβ and Tau in AD

The overactivation of the RAS in the brain—especially Ang II-mediated signaling via AT1R—is associated with AD [135]. In one study, the cerebroventricular infusion of Ang II into aged normal rats increased both tau pathology and APP, leading to a rise in Aβ levels [136]. The link between ACE and Aβ and tau has been confirmed by evaluating the expression and distribution of RAS components in human postmortem brain tissue. ACE2 prevents pathogenic Aβ plaque formation. ACE2 enzyme activity was shown to be reduced by ~50% in the mid-frontal cortex of patients with AD compared with age-matched controls. Additionally, the ratio of ACE2 to ACE1 is decreased in AD patients [119]. In an animal model of sporadic AD, Ang (1–7) expression in the brain increased significantly with disease progression, and an inverse correlation was observed between the Ang (1–7) level and tau hyperphosphorylation. It is thought that components of the RAS aggravate AD by promoting extracellular Aβ deposition and enhancing the intracellular accumulation of pathologic tau protein [6,137]. However, the specific mechanism by which RAS activation links CKD and AD remains unknown.

5.3. RAS and Microglia

Inflammation, which encompasses a variety of immune reactions, is a prominent feature of AD. Microglia are the immune cells that mediate neuroinflammation [18], acting as brain-resident macrophages [138,139]. Microglia have two distinct phenotypes—namely, proinflammatory/classically activated (M1) and anti-inflammatory/alternatively activated (M2) phenotypes according to physiologic or pathologic conditions. The brain RAS plays an important role in microglia polarization. By binding to Ang II, AT1 stimulation triggers NADPH oxidase activation, leading to ROS generation and inducing a shift in microglia phenotype from M2 to M1 [140]. M1 microglia secrete proinflammatory cytokines that exacerbate cognitive dysfunction in the cortex, hippocampus, and basal ganglia by promoting ROS production and neuronal death [141]. The upregulation of AT1R in M1 microglia is associated with inflammation and cognitive impairment through a Toll-like receptor 4 (TLR4)-dependent mechanism [142]; thus, the AT1R-mediated activation of proinflammatory M1 microglia exacerbates inflammation, eventually causing AD [143]. Moreover, the activation of AT2R shifts microglia toward an M2 phenotype. AT2R and MasR activation decrease inducible nitric oxide synthase (iNOS) and the levels of inflammatory cytokines such as C-X-C motif chemokine ligand 12 (CXCL12), interleukin 1β (IL-1β), and IL-6 produced by M1 microglia while increasing anti-inflammatory markers such as IL-10 and IL-4 [143,144,145,146,147]. Thus, AT2R and MasR promote the activation of an M2 anti-inflammatory phenotype, which is a potential mechanism by which neuronal dysfunction and inflammation can be alleviated and cognition impairment can be reversed [143].

5.4. RAS and the Blood–Brain Barrier (BBB)

The BBB is a highly specialized endothelial cell membrane that regulates the entry of plasma-derived components, red blood cells, and leukocytes into the CNS and prevents the entry of potentially neurotoxic molecules [148,149,150]. In a clinical study, Michael Ouk et al. [117] reported that among APOE ε4 non-carriers with AD, ARB use was related to the greater preservation of memory and attention/psychomotor processing speed, particularly compared with ACEIs that do not cross the blood-brain-barrier (RR = 1.200, p = 0.003). Experiments in 5×FAD mice, which recapitulate the main features, and are a widely used model, of AD, showed that the BBB was disrupted in CKD by the action of Ang II [20,151,152], which also induced inflammatory and thrombotic phenotypes in the cerebral microcirculation. The binding of Ang II with AT1R results in leakage of the BBB and the entry of circulating toxins into the brain. Microglia are essential for CNS homeostasis and their overactivation leads to increases in NOS levels and the production of ROS and proinflammatory cytokines including tumor necrosis factor α (TNF-α), IL-1β, and IL-6 [153]. This causes neuronal injury and enhances BBB permeability via the activation of AT1R expressed by brain endothelial cells, resulting in a positive feedback effect.
In conclusion, the occurrence and development of AD are closely related to RAS activation, but it is unknown whether RAS activation in AD and CKD are related, and by which mechanism if so.

6. Uremic Toxins

Uremic toxins accumulate in the body in CKD, leading to neurotoxicity, BBB damage, ischemia/microvascular changes, neuroinflammation, and oxidative stress [154,155]. Patients with CKD have a similar risk of developing AD to the general population [2,156]. An accumulation of uremic toxins in the brain has been reported in patients with uremic syndrome: 5- to 20-fold (or more) increases in the levels of guanidine compounds have been detected in different brain areas [68,157]. Guanidine-based compounds are the main cause of cognitive deficits [157,158]; following kidney transplantation, the levels of these substances were found to decrease with a concomitant alleviation of cognitive deficits. The accumulation of guanidine compounds in the brain causes direct damage to neurons and astrocytes triggered by elevation in the levels of cytokines and interleukins, neuroinflammation, oxidative stress, and increased neuronal apoptosis [159,160,161]. Uremic toxins cause BBB damage [162,163], leading to vascular damage, the influx of endogenous and exogenous toxic chemicals and inflammatory substances into the brain, oxidative stress, neurotoxicity, and cognitive impairment [156].

7. Erythropoietin

Tubulointerstitial fibrosis in CKD patients leads to the loss of EPO secretion [6,18]. Both EPO and its receptor (EPOR) have been detected in the brain [164]. EPO/EPOR signaling is required for regular brain development and is essential for preventing neuron apoptosis, oxidative stress, and inflammation. The protective effects of EPO were first studied at the cellular level using hippocampal neurons and PC12 cells [130,165] and they involve the suppression of oxidative stress, the blockade of apoptosis, and tau phosphorylation induced by Aβ toxicity [131,132]. EPO was shown to alleviate Aβ-induced memory impairment and cognitive deficits by restoring vesicle release probability in Sprague–Dawley rats [132]. The mechanisms by which EPO protects against inflammation involve microglia activation, phosphatidylserine exposure, and protein kinase B (PKB) activity or the prevention of the release of proinflammatory cytokines including IL-6, TNF, and monocyte chemoattractant protein-1 (MCP-1). Thus, EPO is neuroprotective and may prevent or slow the progression of AD. In a large-scale clinical trial, EPO improved cognitive function and slowed progressive neuron atrophy in the brain [166,167,168]. These results suggest that the reduced secretion of EPO promotes cognitive impairment in patients with CKD.

8. Extracellular Vehicles

EVs have important physiologic functions and may contribute to the development and progression of inflammatory, vascular, malignant, infectious, and neurodegenerative diseases. In end-stage renal disease, circulating EVs were found to impair endothelial-dependent vasorelaxation, which was associated with a decrease in endothelial nitric oxide release and endothelial function. In a mouse model of indoxyl sulfate-induced CKD, intravenous administration of EVs from indoxyl sulfate-treated endothelial cells significantly reduced endothelial regeneration [80]. Additionally, animal and cell-based studies have shown that indoxyl sulfate homocysteine enhanced EV release in vitro and in vivo, leading to inflammation, apoptosis, cellular senescence, proliferation, calcification, and neointimal hyperplasia [80,169,170,171]. EVs from the brain of patients with AD contain elevated levels of Aβ oligomers and act as vehicles for the neuron-to-neuron transfer of these toxic species. Blocking the formation, secretion, or uptake of EVs was found to reduce both the spread of oligomers and associated toxicity [78,172]. Additionally, the overexpression of the AD-associated gene bridging integrator 1 (BIN1) stimulated the release of tau via EVs in vitro and aggravated tau pathology in PS19 mice [173]. Furthermore, the injection of physiologic levels of free-form tau into mice greatly diminished the propagation of tau protein compared with the injection of physiologic levels of EV-associated tau, suggesting that EVs are vehicles for the transfer of pathologic tau [174]. As mentioned above, EVs play a role in the pathogenesis of both CKD and AD; however, it is unknown whether CKD affects the pathogenesis of AD through cellular crosstalk mediated by EVs. This can be evaluated in future studies by injecting exosomes extracted from a CKD mouse model into the brain of normal or AD mice.

9. Endothelin

Endothelin (ET) plays a major role in the development of proteinuria, fibrosis, and CKD progression. ET-1 is involved in cell proliferation, hypertrophy, inflammation, and extracellular matrix accumulation, which attributes to the progression of CKD. With CKD progressions such as insulin resistance, dyslipidemia, ROS formation, and nitric oxide deficiency, the production of ET-1 will increase [175,176]. Hiddo J. L. Heerspink et al. [177] demonstrated that the selective ET-A receptor antagonist atrasentan could reduce albuminuria and reduce the risk of kidney failure in CKD patients [177]. Endothelin-converting enzyme inhibitors (ECEIs) have been reported to delay CKD progression by regulating autophagy, the NLRP3 inflammasome, and endoplasmic reticulum stress [178]. In addition, as a potent vasoconstrictor, ET-1 contributes to cerebrovascular dysfunction and neuroinflammation, which is associated with the progression of AD and related dementias [179,180]. Gulati et al. [181] showed that ET-B receptors agonist IRL-1620 reduced oxidative stress and improved learning and memory in an aged APP/PS1 transgenic mouse model of AD. Impaired clearance of Aβ has been considered one of the pathophysiologies of AD, while endothelin-converting enzyme (ECE)-1 and ECE-2 are known enzymes that could degrade Aβ in vivo. Studies have shown that alterations in ECE activity may be deemed as a cause for increased intraneuronal Aβ in AD [182,183]. To sum up, ECEI could delay the progression of CKD, while ECE could promote Aβ to improve AD. Whether we can explore the relationship between CKD and AD from the balance between ECE and ECEI, maybe a future research goal.

10. Conclusions and Future Perspectives

The available evidence suggests that CKD and AD are pathologically related through the RAS, uremic toxins, and EPO, which contribute to the occurrence and development of CKD and may aggravate the development of AD. In CKD, excess renin is released and increases circulating Ang II levels, resulting in AT1R upregulation and enhancing systemic vascular resistance, increasing blood pressure, and promoting sodium reabsorption in the proximal tubule and (through aldosterone) the collecting duct. In AD animal models, the cerebroventricular infusion of Ang II into aged normal rats increased both tau pathology and APP levels, leading to an increase in Aβ accumulation. It was also shown that Ang (1–7) expression in the brain increased with disease progression and that there was an inverse correlation between Ang (1–7) level and tau hyperphosphorylation. In AD model mice, Ang II not only impaired BBB function in the cerebral microcirculation but also induced inflammatory and thrombotic phenotypes. The binding of Ang II with AT1R damaged the BBB, leading to its leakage and the entry of circulating toxins into the brain. Additionally, AT2R and MasR promoted an M2 anti-inflammatory phenotype in microglia, which is a potential mechanism for alleviating neuronal dysfunction and inflammation and ultimately, for reversing cognition impairment. Based on the current evidence, we propose that the combination of Ang II and AT1R causes BBB leakage and activates microglia to secrete inflammatory factors that lead to apoptosis, neuronal injury, and neurodegeneration, resulting in the aggravation of AD; the activation of the AT2R/MasR axis produces the opposite physiological effect.
It remains unclear whether RAS imbalance in CKD is a cause of AD and vice versa. The following open questions warrant investigation in future studies: (1) Do CKD patients with AD have more severe imbalances in the RAS than those without AD? (2) What are the most significantly altered components of the RAS in CKD patients with AD, and are these components mainly proinflammatory (ACE/AT1R) or anti-inflammatory (ACE2/AT2R/MasR)? (3) Can the use of ACEI/ARB drugs prevent or delay the occurrence of AD? Answering these questions may provide insights that can guide the development of novel treatments for both diseases.

Author Contributions

Conceptualization, K.M. and Z.-R.Z.; writing—original draft preparation, K.M. and Z.-R.Z.; writing—review and editing, K.M. and Y.M.; supervision, Y.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the National Natural Science Foundation of China, (82270756); the Natural Science Foundation of Guangdong, China (2018A030313527); the Basic and Applied Basic Research Foundation of Guangdong Province, China (2019A1515010176); the Science and Technology Project of Guangzhou, China (202102010133); the Science and Technology Project of Shenzhen, China (JCYJ20190808095615389); and the National Natural Science Foundation of China (82000686).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

CKDchronic kidney disease
ADAlzheimer’s disease
β-amyloid
NFTNeurofibrillary tangle
RASRenin-angiotensin system
ACE 1, 2Angiotensin-converting enzyme 1, 2
ACEIACE inhibitor
ApoE4Epsilon 4 allele of the apolipoprotein E gene
APPAmyloid precursor protein
SorLASorting protein-related receptor
GFRGlomerular filtration rate
EPOErythropoietin
EVExtracellular vehicle
ROSReactive oxygen species
Ang I, II, III, IVAngiotensin I, II, III, IV
AT1/2/4RAng II receptor type 1, 2, 4
AGTAngiotensinogen
AVPArginine vasopressin
CNSCentral nervous system
Atp6ap2ATPase H+ transporting accessory protein 2
AGTRAGT receptor
MasRMas receptor
ARBAngiotensin-II-receptor blocker
TLR4Toll-like receptor 4
iNOSInducible nitric oxide synthase
CXCL12C-X-C motif chemokine ligand 12
IL-1β, 4, 6, 10Interleukin 1β, 4, 6, 10
BBBBlood-brain barrier
TNF-αTumor necrosis factor α
EPOREPO receptor
PKBProtein kinase B
MCP-1Monocyte chemoattractant protein-1

References

  1. Mayeux, R.; Stern, Y. Epidemiology of Alzheimer Disease. Cold Spring Harb. Perspect. Med. 2012, 2, a006239. [Google Scholar] [CrossRef] [PubMed]
  2. Fogari, R.; Zoppi, A. Effect of Antihypertensive Agents on Quality of Life in the Elderly. Drugs Aging 2004, 21, 377–393. [Google Scholar] [CrossRef] [PubMed]
  3. DeTure, M.A.; Dickson, D.W. The Neuropathological Diagnosis of Alzheimer’s Disease. Mol. Neurodegener. 2019, 14, 32. [Google Scholar] [CrossRef]
  4. Kopp, J.B. Global Glomerulosclerosis in Primary Nephrotic Syndrome: Including Age as a Variable to Predict Renal Outcomes. Kidney Int. 2018, 93, 1043–1044. [Google Scholar] [CrossRef]
  5. Huang, W.; Qiu, C.; von Strauss, E.; Winblad, B.; Fratiglioni, L. Apoe Genotype, Family History of Dementia, and Alzheimer Disease Risk: A 6-Year Follow-up Study. Arch. Neurol. 2004, 61, 1930–1934. [Google Scholar] [CrossRef] [PubMed]
  6. Li, J.; Wang, Y.J.; Zhang, M.; Xu, Z.Q.; Gao, C.Y.; Fang, C.Q.; Yan, J.C.; Zhou, H.D.; Group Chongqing Ageing Study. Vascular Risk Factors Promote Conversion from Mild Cognitive Impairment to Alzheimer Disease. Neurology 2011, 76, 1485–1491. [Google Scholar] [CrossRef]
  7. Kivipelto, M.; Ngandu, T.; Fratiglioni, L.; Viitanen, M.; Kåreholt, I.; Winblad, B.; Helkala, E.L.; Tuomilehto, J.; Soininen, H.; Nissinen, A. Obesity and Vascular Risk Factors at Midlife and the Risk of Dementia and Alzheimer Disease. Arch. Neurol. 2005, 62, 1556–1560. [Google Scholar] [CrossRef]
  8. Forbes, J.M.; Cooper, M.E. Mechanisms of Diabetic Complications. Physiol. Rev. 2013, 93, 137–188. [Google Scholar] [CrossRef]
  9. Stanciu, G.D.; Bild, V.; Ababei, D.C.; Rusu, R.N.; Cobzaru, A.; Paduraru, L.; Bulea, D. Link between Diabetes and Alzheimer’s Disease Due to the Shared Amyloid Aggregation and Deposition Involving Both Neurodegenerative Changes and Neurovascular Damages. J. Clin. Med. 2020, 9, 1713. [Google Scholar] [CrossRef]
  10. Wan, J.; Wang, S.; Haynes, K.; Denburg, M.R.; Shin, D.B.; Gelfand, J.M. Risk of Moderate to Advanced Kidney Disease in Patients with Psoriasis: Population Based Cohort Study. BMJ 2013, 347, f5961. [Google Scholar] [CrossRef]
  11. Willnow, T.E.; Andersen, O.M. Sorting Receptor Sorla—A Trafficking Path to Avoid Alzheimer Disease. J. Cell Sci. 2013, 126, 2751–2760. [Google Scholar] [CrossRef]
  12. Scherzer, C.R.; Offe, K.; Gearing, M.; Rees, H.D.; Fang, G.; Heilman, C.J.; Schaller, C.; Bujo, H.; Levey, A.I.; Lah, J.J. Loss of Apolipoprotein E Receptor Lr11 in Alzheimer Disease. Arch. Neurol. 2004, 61, 1200–1205. [Google Scholar] [CrossRef]
  13. Nielsen, M.S.; Gustafsen, C.; Madsen, P.; Nyengaard, J.R.; Hermey, G.; Bakke, O.; Mari, M.; Schu, P.; Pohlmann, R.; Dennes, A.; et al. Sorting by the Cytoplasmic Domain of the Amyloid Precursor Protein Binding Receptor Sorla. Mol. Cell. Biol. 2007, 27, 6842–6851. [Google Scholar] [CrossRef]
  14. Ştefănescu, R.; Stanciu, G.D.; Luca, A.; Caba, I.C.; Tamba, B.I.; Mihai, C.T. Contributions of Mass Spectrometry to the Identification of Low Molecular Weight Molecules Able to Reduce the Toxicity of Amyloid-Beta Peptide to Cell Cultures and Transgenic Mouse Models of Alzheimer’s Disease. Molecules 2019, 24, 1167. [Google Scholar] [CrossRef]
  15. Stanciu, G.D.; Luca, A.; Rusu, R.N.; Bild, V.; Chiriac, S.I.B.; Solcan, C.; Bild, W.; Ababei, D.C. Alzheimer’s Disease Pharmacotherapy in Relation to Cholinergic System Involvement. Biomolecules 2019, 10, 40. [Google Scholar] [CrossRef]
  16. Jha, V.; Garcia-Garcia, G.; Iseki, K.; Li, Z.; Naicker, S.; Plattner, B.; Saran, R.; Wang, A.Y.M.; Yang, C.W. Chronic Kidney Disease: Global Dimension and Perspectives. Lancet 2013, 382, 260–272. [Google Scholar] [CrossRef]
  17. Krishnan, A.V.; Kiernan, M.C. Neurological Complications of Chronic Kidney Disease. Nat. Rev. Neurol. 2009, 5, 542–551. [Google Scholar] [CrossRef]
  18. Zhang, C.Y.; He, F.F.; Su, H.; Zhang, C.; Meng, X.F. Association between Chronic Kidney Disease and Alzheimer’s Disease: An Update. Metab. Brain Dis. 2020, 35, 883–894. [Google Scholar] [CrossRef]
  19. Stanciu, G.D.; Ababei, D.C.; Bild, V.; Bild, W.; Paduraru, L.; Gutu, M.M.; Tamba, B.I. Renal Contributions in the Pathophysiology and Neuropathological Substrates Shared by Chronic Kidney Disease and Alzheimer’s Disease. Brain Sci. 2020, 10, 563. [Google Scholar] [CrossRef]
  20. Nakagawa, T.; Hasegawa, Y.; Uekawa, K.; Kim-Mitsuyama, S. Chronic Kidney Disease Accelerates Cognitive Impairment in a Mouse Model of Alzheimer’s Disease, through Angiotensin Ii. Exp. Gerontol. 2017, 87, 108–112. [Google Scholar] [CrossRef]
  21. Xue, L.; Lou, Y.; Feng, X.; Wang, C.; Ran, Z.; Zhang, X. Prevalence of Chronic Kidney Disease and Associated Factors among the Chinese Population in Taian, China. BMC Nephrol. 2014, 15, 205. [Google Scholar] [CrossRef] [PubMed]
  22. Loutradis, C.; Price, A.; Ferro, C.J.; Sarafidis, P. Renin-Angiotensin System Blockade in Patients with Chronic Kidney Disease: Benefits, Problems in Everyday Clinical Use, and Open Questions for Advanced Renal Dysfunction. J. Hum. Hypertens. 2021, 35, 499–509. [Google Scholar] [CrossRef]
  23. Walther, C.P.; Winkelmayer, W.C.; Richardson, P.A.; Virani, S.S.; Navaneethan, S.D. Renin-Angiotensin System Blocker Discontinuation and Adverse Outcomes in Chronic Kidney Disease. Nephrol. Dial. Transplant. 2021, 36, 1893–1899. [Google Scholar] [CrossRef] [PubMed]
  24. Natale, G.; Calabrese, V.; Marino, G.; Campanelli, F.; Urciuolo, F.; de Iure, A.; Ghiglieri, V.; Calabresi, P.; Bossola, M.; Picconi, B. Effects of Uremic Toxins on Hippocampal Synaptic Transmission: Implication for Neurodegeneration in Chronic Kidney Disease. Cell Death Discov. 2021, 7, 295. [Google Scholar] [CrossRef] [PubMed]
  25. Xie, Y.; Bowe, B.; Mokdad, A.H.; Xian, H.; Yan, Y.; Li, T.; Maddukuri, G.; Tsai, C.Y.; Floyd, T.; Al-Aly, Z. Analysis of the Global Burden of Disease Study Highlights the Global, Regional, and National Trends of Chronic Kidney Disease Epidemiology from 1990 to 2016. Kidney Int. 2018, 94, 567–581. [Google Scholar] [CrossRef] [PubMed]
  26. Singh, D.K.; Winocour, P.; Farrington, K. Oxidative Stress in Early Diabetic Nephropathy: Fueling the Fire. Nat. Rev. Endocrinol. 2011, 7, 176–184. [Google Scholar] [CrossRef]
  27. Kim, H.J.; Vaziri, N.D. Contribution of Impaired Nrf2-Keap1 Pathway to Oxidative Stress and Inflammation in Chronic Renal Failure. Am. J. Physiol. Renal. Physiol. 2010, 298, F662–F671. [Google Scholar] [CrossRef]
  28. Noonan, M.L.; Clinkenbeard, E.L.; Ni, P.; Swallow, E.A.; Tippen, S.P.; Agoro, R.; Allen, M.R.; White, K.E. Erythropoietin and a Hypoxia-Inducible Factor Prolyl Hydroxylase Inhibitor (Hif-Phdi) Lowers Fgf23 in a Model of Chronic Kidney Disease (Ckd). Physiol. Rep. 2020, 8, e14434. [Google Scholar] [CrossRef]
  29. Noonan, M.L.; Ni, P.; Agoro, R.; Sacks, S.A.; Swallow, E.A.; Wheeler, J.A.; Clinkenbeard, E.L.; Capitano, M.L.; Prideaux, M.; Atkins, G.J.; et al. The Hif-Phi Bay 85-3934 (Molidustat) Improves Anemia and Is Associated with Reduced Levels of Circulating Fgf23 in a Ckd Mouse Model. J. Bone Miner. Res. 2021, 36, 1117–1130. [Google Scholar] [CrossRef]
  30. Pieniazek, A.; Bernasinska-Slomczewska, J.; Gwozdzinski, L. Uremic Toxins and Their Relation with Oxidative Stress Induced in Patients with Ckd. Int. J. Mol. Sci. 2021, 22, 6196. [Google Scholar] [CrossRef]
  31. Shaykh, M.; Pegoraro, A.A.; Mo, W.; Arruda, J.A.L.; Dunea, G.; Singh, A.K. Carbamylated Proteins Activate Glomerular Mesangial Cells and Stimulate Collagen Deposition. J. Lab. Clin. Med. 1999, 133, 302–308. [Google Scholar] [CrossRef]
  32. Pieniazek, A.; Brzeszczynska, J.; Kruszynska, I.; Gwozdzinski, K. Investigation of Albumin Properties in Patients with Chronic Renal Failure. Free Radic. Res. 2009, 43, 1008–1018. [Google Scholar] [CrossRef]
  33. Jaisson, S.; Pietrement, C.; Gillery, P. Carbamylation-Derived Products: Bioactive Compounds and Potential Biomarkers in Chronic Renal Failure and Atherosclerosis. Clin. Chem. 2011, 57, 1499–1505. [Google Scholar] [CrossRef]
  34. Koppe, L.; Nyam, E.; Vivot, K.; Fox, J.E.M.; Dai, X.Q.; Nguyen, B.N.; Trudel, D.; Attané, C.; Moullé, V.S.; MacDonald, P.E.; et al. Urea Impairs Beta Cell Glycolysis and Insulin Secretion in Chronic Kidney Disease. J. Clin. Investig. 2016, 126, 3598–3612. [Google Scholar] [CrossRef]
  35. van Gelder, M.K.; Middel, I.R.; Vernooij, R.W.; Bots, M.L.; Verhaar, M.C.; Masereeuw, R.; Grooteman, M.P.; Nubé, M.J.; van den Dorpel, M.A.; Blankestijn, P.J.; et al. Protein-Bound Uremic Toxins in Hemodialysis Patients Relate to Residual Kidney Function, Are Not Influenced by Convective Transport, and Do Not Relate to Outcome. Toxins 2020, 12, 234. [Google Scholar] [CrossRef]
  36. Yaker, L.; Kamel, S.; Ausseil, J.; Boullier, A. Effects of Chronic Kidney Disease and Uremic Toxins on Extracellular Vesicle Biology. Toxins 2020, 12, 811. [Google Scholar] [CrossRef]
  37. Chao, C.T.; Lin, S.H. Uremic Toxins and Frailty in Patients with Chronic Kidney Disease: A Molecular Insight. Int. J. Mol. Sci. 2021, 22, 6270. [Google Scholar] [CrossRef]
  38. Lim, Y.J.; Sidor, N.A.; Tonial, N.C.; Che, A.; Urquhart, B.L. Uremic Toxins in the Progression of Chronic Kidney Disease and Cardiovascular Disease: Mechanisms and Therapeutic Targets. Toxins 2021, 13, 142. [Google Scholar] [CrossRef]
  39. Wojtaszek, E.; Oldakowska-Jedynak, U.; Kwiatkowska, M.; Glogowski, T.; Malyszko, J. Uremic Toxins, Oxidative Stress, Atherosclerosis in Chronic Kidney Disease, and Kidney Transplantation. Oxid. Med. Cell. Longev. 2021, 2021, 6651367. [Google Scholar] [CrossRef]
  40. Tamaki, N. Ischemia and Inflammation on Chronic Kidney Disease. J. Nucl. Cardiol. 2019, 26, 441–442. [Google Scholar] [CrossRef]
  41. Pereira-Balbino, K.; de-Paula-Jorge, M.; Queiroz-Ribeiro, A.; Stampini-Duarte-Martino, H. Modulation of Intestinal Microbiota, Control of Nitrogen Products and Inflammation by Pre/Probiotics in Chronic Kidney Disease: A Systematic Review. Nutr. Hosp. 2018, 35, 722–730. [Google Scholar]
  42. Mihai, S.; Codrici, E.; Popescu, I.D.; Enciu, A.M.; Albulescu, L.; Necula, L.G.; Mambet, C.; Anton, G.; Tanase, C. Inflammation-Related Mechanisms in Chronic Kidney Disease Prediction, Progression, and Outcome. J. Immunol. Res. 2018, 2018, 2180373. [Google Scholar] [CrossRef] [PubMed]
  43. Diaz-Ricart, M.; Torramade-Moix, S.; Pascual, G.; Palomo, M.; Moreno-Castaño, A.B.; Martinez-Sanchez, J.; Vera, M.; Cases, A.; Escolar, G. Endothelial Damage, Inflammation and Immunity in Chronic Kidney Disease. Toxins 2020, 12, 361. [Google Scholar] [CrossRef] [PubMed]
  44. Ebert, T.; Pawelzik, S.C.; Witasp, A.; Arefin, S.; Hobson, S.; Kublickiene, K.; Shiels, P.G.; Bäck, M.; Stenvinkel, P. Inflammation and Premature Ageing in Chronic Kidney Disease. Toxins 2020, 12, 227. [Google Scholar] [CrossRef]
  45. Mazzaferro, S.; De Martini, N.; Rotondi, S.; Tartaglione, L.; Ureña-Torres, P.; Bover, J.; Pasquali, M.; Era-Edta Working Group on CKD-MBD. Bone, Inflammation and Chronic Kidney Disease. Clin. Chim. Acta 2020, 506, 236–240. [Google Scholar] [CrossRef]
  46. Brennan, E.; Kantharidis, P.; Cooper, M.E.; Godson, C. Pro-Resolving Lipid Mediators: Regulators of Inflammation, Metabolism and Kidney Function. Nat. Rev. Nephrol. 2021, 17, 725–739. [Google Scholar] [CrossRef]
  47. Düsing, P.; Zietzer, A.; Goody, P.R.; Hosen, M.R.; Kurts, C.; Nickenig, G.; Jansen, F. Vascular Pathologies in Chronic Kidney Disease: Pathophysiological Mechanisms and Novel Therapeutic Approaches. J. Mol. Med. 2021, 99, 335–348. [Google Scholar] [CrossRef]
  48. Panizo, S.; Martínez-Arias, L.; Alonso-Montes, C.; Cannata, P.; Martín-Carro, B.; Fernández-Martín, J.L.; Naves-Díaz, M.; Carrillo-López, N.; Cannata-Andía, J.B. Fibrosis in Chronic Kidney Disease: Pathogenesis and Consequences. Int. J. Mol. Sci. 2021, 22, 408. [Google Scholar] [CrossRef]
  49. Kakani, E.; Elyamny, M.; Ayach, T.; El-Husseini, A. Pathogenesis and Management of Vascular Calcification in Ckd and Dialysis Patients. Semin. Dial. 2019, 32, 553–561. [Google Scholar] [CrossRef]
  50. Mace, M.L.; Gravesen, E.; Nordholm, A.; Egstrand, S.; Morevati, M.; Nielsen, C.; Kjaer, A.; Behets, G.; d′Haese, P.; Olgaard, K.; et al. Chronic Kidney Disease-Induced Vascular Calcification Impairs Bone Metabolism. J. Bone Miner. Res. 2021, 36, 510–522. [Google Scholar] [CrossRef]
  51. Singh, A.; Tandon, S.; Tandon, C. An Update on Vascular Calcification and Potential Therapeutics. Mol. Biol. Rep. 2021, 48, 887–896. [Google Scholar] [CrossRef]
  52. Zhang, Y.X.; Tang, R.N.; Wang, L.T.; Liu, B.C. Role of Crosstalk between Endothelial Cells and Smooth Muscle Cells in Vascular Calcification in Chronic Kidney Disease. Cell Prolif. 2021, 54, e12980. [Google Scholar] [CrossRef]
  53. Xie, X.; Liu, Y.; Perkovic, V.; Li, X.; Ninomiya, T.; Hou, W.; Zhao, N.; Liu, L.; Lv, J.; Zhang, H.; et al. Renin-Angiotensin System Inhibitors and Kidney and Cardiovascular Outcomes in Patients with Ckd: A Bayesian Network Meta-Analysis of Randomized Clinical Trials. Am. J. Kidney Dis. 2016, 67, 728–741. [Google Scholar] [CrossRef]
  54. Zhou, L.; Liu, Y. Wnt/Beta-Catenin Signaling and Renin-Angiotensin System in Chronic Kidney Disease. Curr. Opin. Nephrol. Hypertens. 2016, 25, 100–106. [Google Scholar] [CrossRef]
  55. Saldanha da Silva, A.A.; Rodrigues Prestes, T.R.; Lauar, A.O.; Finotti, B.B.; Simoes e Silva, A.C. Renin Angiotensin System and Cytokines in Chronic Kidney Disease: Clinical and Experimental Evidence. Protein Pept. Lett. 2017, 24, 799–808. [Google Scholar] [CrossRef]
  56. Nistor, I.; De Sutter, J.; Drechsler, C.; Goldsmith, D.; Soler, M.J.; Tomson, C.; Wiecek, A.; Donciu, M.D.; Bolignano, D.; Van Biesen, W.; et al. Effect of Renin-Angiotensin-Aldosterone System Blockade in Adults with Diabetes Mellitus and Advanced Chronic Kidney Disease Not on Dialysis: A Systematic Review and Meta-Analysis. Nephrol. Dial. Transplant. 2018, 33, 12–22. [Google Scholar] [CrossRef]
  57. Leon, S.J.; Tangri, N. The Use of Renin-Angiotensin System Inhibitors in Patients with Chronic Kidney Disease. Can. J. Cardiol. 2019, 35, 1220–1227. [Google Scholar] [CrossRef]
  58. Laffer, C.L.; Elijovich, F.; Sahinoz, M.; Pitzer, A.; Kirabo, A. New Insights into the Renin-Angiotensin System in Chronic Kidney Disease. Circ. Res. 2020, 127, 607–609. [Google Scholar] [CrossRef]
  59. Karpman, D.; Ståhl, A.L.; Arvidsson, I. Extracellular Vesicles in Renal Disease. Nat. Rev. Nephrol. 2017, 13, 545–562. [Google Scholar] [CrossRef]
  60. Ando, M.; Iwata, A.; Ozeki, Y.; Tsuchiya, K.; Akiba, T.; Nihei, H. Circulating Platelet-Derived Microparticles with Procoagulant Activity May Be a Potential Cause of Thrombosis in Uremic Patients. Kidney Int. 2002, 62, 1757–1763. [Google Scholar] [CrossRef]
  61. Faure, V.; Dou, L.; Sabatier, F.; Cerini, C.; Sampol, J.; Berland, Y.; Brunet, P.; Dignat-George, F. Elevation of Circulating Endothelial Microparticles in Patients with Chronic Renal Failure. J. Thromb. Haemost. 2006, 4, 566–573. [Google Scholar] [CrossRef] [PubMed]
  62. Burton, J.O.; Hamali, H.A.; Singh, R.; Abbasian, N.; Parsons, R.; Patel, A.K.; Goodall, A.H.; Brunskill, N.J. Elevated Levels of Procoagulant Plasma Microvesicles in Dialysis Patients. PLoS ONE 2013, 8, e72663. [Google Scholar] [CrossRef] [PubMed]
  63. Dursun, I.; Poyrazoglu, H.M.; Gunduz, Z.; Ulger, H.; Yýkýlmaz, A.; Dusunsel, R.; Patýroglu, T.; Gurgoze, M. The Relationship between Circulating Endothelial Microparticles and Arterial Stiffness and Atherosclerosis in Children with Chronic Kidney Disease. Nephrol. Dial. Transplant. 2009, 24, 2511–2518. [Google Scholar] [CrossRef] [PubMed]
  64. Charles, C.; Ferris, A.H. Chronic Kidney Disease. Prim. Care 2020, 47, 585–595. [Google Scholar] [CrossRef]
  65. Lane, C.A.; Hardy, J.; Schott, J.M. Alzheimer’s Disease. Eur. J. Neurol. 2018, 25, 59–70. [Google Scholar] [CrossRef]
  66. Scheltens, P.; De Strooper, B.; Kivipelto, M.; Holstege, H.; Chételat, G.; Teunissen, C.E.; Cummings, J.; van der Flier, W.M. Alzheimer’s Disease. Lancet 2021, 397, 1577–1590. [Google Scholar] [CrossRef]
  67. Ou, Z.; Jiang, T.; Gao, Q.; Tian, Y.Y.; Zhou, J.S.; Wu, L.; Shi, J.Q.; Zhang, Y.D. Mitochondrial-Dependent Mechanisms Are Involved in Angiotensin Ii-Induced Apoptosis in Dopaminergic Neurons. J. Renin-Angiotensin-Aldosterone Syst. 2016, 17, 1470320316672349. [Google Scholar] [CrossRef]
  68. He, S.; Wang, L.H.; Liu, Y.; Li, Y.Q.; Chen, H.T.; Xu, J.H.; Peng, W.; Lin, G.W.; Wei, P.P.; Li, B.; et al. Single-Cell Transcriptome Profiling of an Adult Human Cell Atlas of 15 Major Organs. Genome Biol. 2020, 21, 294. [Google Scholar] [CrossRef]
  69. Khan, S.; Barve, K.H.; Kumar, M.S. Recent Advancements in Pathogenesis, Diagnostics and Treatment of Alzheimer’s Disease. Curr. Neuropharmacol. 2020, 18, 1106–1125. [Google Scholar] [CrossRef]
  70. Vaz, M.; Silvestre, S. Alzheimer’s Disease: Recent Treatment Strategies. Eur. J. Pharmacol. 2020, 887, 173554. [Google Scholar] [CrossRef]
  71. Xin, S.H.; Tan, L.; Cao, X.; Yu, J.T.; Tan, L. Clearance of Amyloid Beta and Tau in Alzheimer’s Disease: From Mechanisms to Therapy. Neurotox. Res. 2018, 34, 733–748. [Google Scholar] [CrossRef]
  72. Tiwari, S.; Atluri, V.; Kaushik, A.; Yndart, A.; Nair, M. Alzheimer’s Disease: Pathogenesis, Diagnostics, and Therapeutics. Int. J. Nanomed. 2019, 14, 5541–5554. [Google Scholar] [CrossRef]
  73. Zhang, H.; Zheng, Y. Beta Amyloid Hypothesis in Alzheimer’s Disease: Pathogenesis, Prevention, and Management. Zhongguo Yi Xue Ke Xue Yuan Xue Bao 2019, 41, 702–708. [Google Scholar]
  74. Han, C.; Yang, Y.; Guan, Q.; Zhang, X.; Shen, H.; Sheng, Y.; Wang, J.; Zhou, X.; Li, W.; Guo, L.; et al. New Mechanism of Nerve Injury in Alzheimer’s Disease: Beta-Amyloid-Induced Neuronal Pyroptosis. J. Cell. Mol. Med. 2020, 24, 8078–8090. [Google Scholar] [CrossRef]
  75. Li, L.; Shi, R.; Gu, J.; Tung, Y.C.; Zhou, Y.; Zhou, D.; Wu, R.; Chu, D.; Jin, N.; Deng, K.; et al. Alzheimer’s Disease Brain Contains Tau Fractions with Differential Prion-Like Activities. Acta Neuropathol. Commun. 2021, 9, 28. [Google Scholar] [CrossRef]
  76. Breijyeh, Z.; Karaman, R. Comprehensive Review on Alzheimer’s Disease: Causes and Treatment. Molecules 2020, 25, 5789. [Google Scholar] [CrossRef]
  77. Ashrafian, H.; Zadeh, E.H.; Khan, R.H. Review on Alzheimer’s Disease: Inhibition of Amyloid Beta and Tau Tangle Formation. Int. J. Biol. Macromol. 2021, 167, 382–394. [Google Scholar] [CrossRef]
  78. Hill, A.F. Extracellular Vesicles and Neurodegenerative Diseases. J. Neurosci. 2019, 39, 9269–9273. [Google Scholar] [CrossRef]
  79. Jiang, L.; Dong, H.; Cao, H.; Ji, X.; Luan, S.; Liu, J. Exosomes in Pathogenesis, Diagnosis, and Treatment of Alzheimer’s Disease. Med. Sci. Monit. 2019, 25, 3329–3335. [Google Scholar] [CrossRef]
  80. Martins, T.S.; Trindade, D.; Vaz, M.; Campelo, I.; Almeida, M.; Trigo, G.; da Cruz e Silva, O.A.; Henriques, A.G. Diagnostic and Therapeutic Potential of Exosomes in Alzheimer’s Disease. J. Neurochem. 2021, 156, 162–181. [Google Scholar] [CrossRef]
  81. Zhang, T.; Ma, S.; Lv, J.; Wang, X.; Afewerky, H.K.; Li, H.; Lu, Y. The Emerging Role of Exosomes in Alzheimer’s Disease. Ageing Res. Rev. 2021, 68, 101321. [Google Scholar] [CrossRef] [PubMed]
  82. Tian, X.Y.; Ma, S.; Huang, Y.; Wong, W.T. A Revisit on the Renin-Angiotensin System in Cardiovascular Biology. Trends Cardiovasc. Med. 2016, 26, 229–231. [Google Scholar] [CrossRef] [PubMed]
  83. Claflin, K.E.; Sandgren, J.A.; Lambertz, A.M.; Weidemann, B.J.; Littlejohn, N.K.; Burnett, C.M.; Pearson, N.A.; Morgan, D.A.; Gibson-Corley, K.N.; Rahmouni, K.; et al. Angiotensin At1a Receptors on Leptin Receptor-Expressing Cells Control Resting Metabolism. J. Clin. Investig. 2017, 127, 1414–1424. [Google Scholar] [CrossRef] [PubMed]
  84. Gao, J.; Zhang, H.; Le, K.D.; Chao, J.; Gao, L. Activation of Central Angiotensin Type 2 Receptors Suppresses Norepinephrine Excretion and Blood Pressure in Conscious Rats. Am. J. Hypertens. 2011, 24, 724–730. [Google Scholar] [CrossRef] [PubMed]
  85. Chao, J.; Gao, J.; Parbhu, K.J.K.; Gao, L. Angiotensin Type 2 Receptors in the Intermediolateral Cell Column of the Spinal Cord: Negative Regulation of Sympathetic Nerve Activity and Blood Pressure. Int. J. Cardiol. 2013, 168, 4046–4055. [Google Scholar] [CrossRef]
  86. Gao, J.; Zucker, I.H.; Gao, L. Activation of Central Angiotensin Type 2 Receptors by Compound 21 Improves Arterial Baroreflex Sensitivity in Rats with Heart Failure. Am. J. Hypertens. 2014, 27, 1248–1256. [Google Scholar] [CrossRef]
  87. Harfouch, E.; Daoud, S. Allelic Variation in Hla-Drb1* Loci in Syrian Pemphigus Vulgaris Patients. Int. J. Dermatol. 2014, 53, 1460–1463. [Google Scholar] [CrossRef]
  88. Kemp, B.A.; Howell, N.L.; Gildea, J.J.; Keller, S.R.; Padia, S.H.; Carey, R.M. At2 Receptor Activation Induces Natriuresis and Lowers Blood Pressure. Circ. Res. 2014, 115, 388–399. [Google Scholar] [CrossRef]
  89. De Kloet, A.D.; Wang, L.; Ludin, J.A.; Smith, J.A.; Pioquinto, D.J.; Hiller, H.; Steckelings, U.M.; Scheuer, D.A.; Sumners, C.; Krause, E.G. Reporter Mouse Strain Provides a Novel Look at Angiotensin Type-2 Receptor Distribution in the Central Nervous System. Brain Struct. Funct. 2016, 221, 891–912. [Google Scholar] [CrossRef]
  90. Ji, Y.; Wang, Z.; Li, Z.; Zhang, A.; Jin, Y.; Chen, H.; Le, X. Angiotensin Ii Enhances Proliferation and Inflammation through At1/Pkc/Nf-Kappab Signaling Pathway in Hepatocellular Carcinoma Cells. Cell. Physiol. Biochem. 2016, 39, 13–32. [Google Scholar] [CrossRef]
  91. Kemp, B.A.; Howell, N.L.; Keller, S.R.; Gildea, J.J.; Padia, S.H.; Carey, R.M. At2 Receptor Activation Prevents Sodium Retention and Reduces Blood Pressure in Angiotensin Ii-Dependent Hypertension. Circ. Res. 2016, 119, 532–543. [Google Scholar] [CrossRef]
  92. Goel, R.; Bhat, S.A.; Hanif, K.; Nath, C.; Shukla, R. Angiotensin Ii Receptor Blockers Attenuate Lipopolysaccharide-Induced Memory Impairment by Modulation of Nf-Kappab-Mediated Bdnf/Creb Expression and Apoptosis in Spontaneously Hypertensive Rats. Mol. Neurobiol. 2018, 55, 1725–1739. [Google Scholar] [CrossRef]
  93. Gao, J.; Marc, Y.; Iturrioz, X.; Leroux, V.; Balavoine, F.; Llorens-Cortes, C. A New Strategy for Treating Hypertension by Blocking the Activity of the Brain Renin-Angiotensin System with Aminopeptidase a Inhibitors. Clin. Sci. 2014, 127, 135–148. [Google Scholar] [CrossRef]
  94. Sparks, M.A.; Crowley, S.D.; Gurley, S.B.; Mirotsou, M.; Coffman, T.M. Classical Renin-Angiotensin System in Kidney Physiology. Compr. Physiol. 2014, 4, 1201–1228. [Google Scholar]
  95. Park, B.M.; Gao, S.; Cha, S.A.; Park, B.H.; Kim, S.H. Cardioprotective Effects of Angiotensin Iii against Ischemic Injury Via the At2 Receptor and Katp Channels. Physiol. Rep. 2013, 1, e00151. [Google Scholar] [CrossRef]
  96. Yeatman, H.R.; Albiston, A.L.; Burns, P.; Chai, S.Y. Forebrain Neurone-Specific Deletion of Insulin-Regulated Aminopeptidase Causes Age Related Deficits in Memory. Neurobiol. Learn. Mem. 2016, 136, 174–182. [Google Scholar] [CrossRef]
  97. Paris, J.J.; Eans, S.O.; Mizrachi, E.; Reilley, K.J.; Ganno, M.L.; McLaughlin, J.P. Central Administration of Angiotensin Iv Rapidly Enhances Novel Object Recognition among Mice. Neuropharmacology 2013, 70, 247–253. [Google Scholar] [CrossRef]
  98. Hennrikus, M.; Gonzalez, A.A.; Prieto, M.C. The Prorenin Receptor in the Cardiovascular System and Beyond. Am. J. Physiol. Heart Circ. Physiol. 2018, 314, H139–H145. [Google Scholar] [CrossRef]
  99. Nunes-Silva, A.; Rocha, G.C.; Magalhaes, D.M.; Vaz, L.N.; de Faria, M.H.S.; Simoes e Silva, A.C. Physical Exercise and Ace2-Angiotensin-(1-7)-Mas Receptor Axis of the Renin Angiotensin System. Protein Pept. Lett. 2017, 24, 809–816. [Google Scholar] [CrossRef] [PubMed]
  100. Top 10 Health Technology Hazards for 2013 Are Named. OR Manag. 2013, 29, 16–19.
  101. Ola, M.S.; Alhomida, A.S.; Ferrario, C.M.; Ahmad, S. Role of Tissue Renin-Angiotensin System and the Chymase/Angiotensin-(1-12) Axis in the Pathogenesis of Diabetic Retinopathy. Curr. Med. Chem. 2017, 24, 3104–3114. [Google Scholar] [CrossRef] [PubMed]
  102. Stornetta, R.L.; Hawelu-Johnson, C.L.; Guyenet, P.G.; Lynch, K.R. Astrocytes Synthesize Angiotensinogen in Brain. Science 1988, 242, 1444–1446. [Google Scholar] [CrossRef]
  103. Labandeira-Garcia, J.L.; Rodríguez-Perez, A.I.; Garrido-Gil, P.; Rodriguez-Pallares, J.; Lanciego, J.L.; Guerra, M.J. Brain Renin-Angiotensin System and Microglial Polarization: Implications for Aging and Neurodegeneration. Front. Aging Neurosci. 2017, 9, 129. [Google Scholar] [CrossRef]
  104. Zeisel, A.; Muñoz-Manchado, A.B.; Codeluppi, S.; Lönnerberg, P.; La Manno, G.; Juréus, A.; Marques, S.; Munguba, H.; He, L.; Betsholtz, C.; et al. Brain Structure. Cell Types in the Mouse Cortex and Hippocampus Revealed by Single-Cell Rna-Seq. Science 2015, 347, 1138–1142. [Google Scholar] [CrossRef]
  105. Agarwal, D.; Sandor, C.; Volpato, V.; Caffrey, T.M.; Monzón-Sandoval, J.; Bowden, R.; Alegre-Abarrategui, J.; Wade-Martins, R.; Webber, C. A Single-Cell Atlas of the Human Substantia Nigra Reveals Cell-Specific Pathways Associated with Neurological Disorders. Nat. Commun. 2020, 11, 4183. [Google Scholar] [CrossRef]
  106. Kanton, S.; Boyle, M.J.; He, Z.; Santel, M.; Weigert, A.; Sanchís-Calleja, F.; Guijarro, P.; Sidow, L.; Fleck, J.S.; Han, D.; et al. Organoid Single-Cell Genomic Atlas Uncovers Human-Specific Features of Brain Development. Nature 2019, 574, 418–422. [Google Scholar] [CrossRef]
  107. Rivas-Santisteban, R.; Lillo, J.; Muñoz, A.; Rodríguez-Pérez, A.I.; Labandeira-García, J.L.; Navarro, G.; Franco, R. Novel Interactions Involving the Mas Receptor Show Potential of the Renin-Angiotensin System in the Regulation of Microglia Activation: Altered Expression in Parkinsonism and Dyskinesia. Neurotherapeutics 2021, 18, 998–1016. [Google Scholar] [CrossRef]
  108. Rivas-Santisteban, R.; Rodriguez-Perez, A.I.; Muñoz, A.; Reyes-Resina, I.; Labandeira-García, J.L.; Navarro, G.; Franco, R. Angiotensin At1 and At2 Receptor Heteromer Expression in the Hemilesioned Rat Model of Parkinson’s Disease That Increases with Levodopa-Induced Dyskinesia. J. Neuroinflammation 2020, 17, 243. [Google Scholar] [CrossRef]
  109. Loera-Valencia, R.; Eroli, F.; Garcia-Ptacek, S.; Maioli, S. Brain Renin-Angiotensin System as Novel and Potential Therapeutic Target for Alzheimer’s Disease. Int. J. Mol. Sci. 2021, 22, 10139. [Google Scholar] [CrossRef]
  110. Cassader, M.; Ruiu, G.; Leoncavallo, A.R.; Pagano, G. Changes in Carbohydrate and Lipid Metabolism in Chronic Renal Insufficiency. Clin. Ter. 1987, 122, 271–279. [Google Scholar]
  111. Royea, J.; Hamel, E. Brain Angiotensin Ii and Angiotensin Iv Receptors as Potential Alzheimer’s Disease Therapeutic Targets. Geroscience 2020, 42, 1237–1256. [Google Scholar] [CrossRef] [PubMed]
  112. Mowry, F.E.; Peaden, S.C.; Stern, J.E.; Biancardi, V.C. Tlr4 and At1r Mediate Blood-Brain Barrier Disruption, Neuroinflammation, and Autonomic Dysfunction in Spontaneously Hypertensive Rats. Pharmacol. Res. 2021, 174, 105877. [Google Scholar] [CrossRef] [PubMed]
  113. Mohammed, M.; Berdasco, C.; Lazartigues, E. Brain Angiotensin Converting Enzyme-2 in Central Cardiovascular Regulation. Clin. Sci. 2020, 134, 2535–2547. [Google Scholar] [CrossRef]
  114. Maning, J.; Negussie, S.; Clark, M.A.; Lymperopoulos, A. Biased Agonism/Antagonism at the Angii-At1 Receptor: Implications for Adrenal Aldosterone Production and Cardiovascular Therapy. Pharmacol. Res. 2017, 125, 14–20. [Google Scholar] [CrossRef] [PubMed]
  115. Dang, Z.; Su, S.; Jin, G.; Nan, X.; Ma, L.; Li, Z.; Lu, D.; Ge, R. Tsantan Sumtang Attenuated Chronic Hypoxia-Induced Right Ventricular Structure Remodeling and Fibrosis by Equilibrating Local Ace-Angii-At1r/Ace2-Ang1-7-Mas Axis in Rat. J. Ethnopharmacol. 2020, 250, 112470. [Google Scholar] [CrossRef]
  116. Barthold, D.; Joyce, G.; Brinton, R.D.; Wharton, W.; Kehoe, P.G.; Zissimopoulos, J. Association of Combination Statin and Antihypertensive Therapy with Reduced Alzheimer’s Disease and Related Dementia Risk. PLoS ONE 2020, 15, e0229541. [Google Scholar] [CrossRef]
  117. Ouk, M.; Wu, C.Y.; Rabin, J.S.; Jackson, A.; Edwards, J.D.; Ramirez, J.; Masellis, M.; Swartz, R.H.; Herrmann, N.; Lanctot, K.L.; et al. The Use of Angiotensin-Converting Enzyme Inhibitors vs. Angiotensin Receptor Blockers and Cognitive Decline in Alzheimer’s Disease: The Importance of Blood-Brain Barrier Penetration and Apoe Epsilon4 Carrier Status. Alzheimers Res. Ther. 2021, 13, 43. [Google Scholar] [CrossRef]
  118. Miners, J.S.; Ashby, E.; Van Helmond, Z.; Chalmers, K.A.; Palmer, L.E.; Love, S.; Kehoe, P.G. Angiotensin-Converting Enzyme (Ace) Levels and Activity in Alzheimer’s Disease, and Relationship of Perivascular Ace-1 to Cerebral Amyloid Angiopathy. Neuropathol. Appl. Neurobiol. 2008, 34, 181–193. [Google Scholar] [CrossRef]
  119. Kehoe, P.G.; Wong, S.; Al Mulhim, N.; Palmer, L.E.; Miners, J.S. Angiotensin-Converting Enzyme 2 Is Reduced in Alzheimer’s Disease in Association with Increasing Amyloid-Beta and Tau Pathology. Alzheimers Res. Ther. 2016, 8, 50. [Google Scholar] [CrossRef]
  120. Barnes, N.M.; Cheng, C.H.; Costall, B.; Naylor, R.J.; Williams, T.J.; Wischik, C.M. Angiotensin Converting Enzyme Density Is Increased in Temporal Cortex from Patients with Alzheimer’s Disease. Eur. J. Pharmacol. 1991, 200, 289–292. [Google Scholar] [CrossRef]
  121. Soto, M.E.; van Kan, G.A.; Nourhashemi, F.; Gillette-Guyonnet, S.; Cesari, M.; Cantet, C.; Rolland, Y.; Vellas, B. Angiotensin-Converting Enzyme Inhibitors and Alzheimer’s Disease Progression in Older Adults: Results from the Reseau Sur La Maladie D’alzheimer Francais Cohort. J. Am. Geriatr. Soc. 2013, 61, 1482–1488. [Google Scholar] [CrossRef]
  122. Li, N.C.; Lee, A.; Whitmer, R.A.; Kivipelto, M.; Lawler, E.; Kazis, L.E.; Wolozin, B. Use of Angiotensin Receptor Blockers and Risk of Dementia in a Predominantly Male Population: Prospective Cohort Analysis. BMJ 2010, 340, b5465. [Google Scholar] [CrossRef] [PubMed]
  123. Ongali, B.; Nicolakakis, N.; Tong, X.K.; Aboulkassim, T.; Papadopoulos, P.; Rosa-Neto, P.; Lecrux, C.; Imboden, H.; Hamel, E. Angiotensin Ii Type 1 Receptor Blocker Losartan Prevents and Rescues Cerebrovascular, Neuropathological and Cognitive Deficits in an Alzheimer’s Disease Model. Neurobiol. Dis. 2014, 68, 126–136. [Google Scholar] [CrossRef] [PubMed]
  124. Fogari, R.; Mugellini, A.; Zoppi, A.; Derosa, G.; Pasotti, C.; Fogari, E.; Preti, P. Influence of Losartan and Atenolol on Memory Function in Very Elderly Hypertensive Patients. J. Hum. Hypertens. 2003, 17, 781–785. [Google Scholar] [CrossRef] [PubMed]
  125. Fournier, A.; Oprisiu-Fournier, R.; Serot, J.M.; Godefroy, O.; Achard, J.M.; Faure, S.; Mazouz, H.; Temmar, M.; Albu, A.; Bordet, R.; et al. Prevention of Dementia by Antihypertensive Drugs: How At1-Receptor-Blockers and Dihydropyridines Better Prevent Dementia in Hypertensive Patients Than Thiazides and Ace-Inhibitors. Expert Rev. Neurother. 2009, 9, 1413–1431. [Google Scholar] [CrossRef]
  126. Lemma, F.; Bombardieri, T.; Salibra, M.; Trovato, P.; De Pasquale, M.C.; Faraone, V.; Punturiero, R.; Di Mauro, S. Tumor Markers in the Diagnosis of Pancreatic Carcinoma. Minerva Med. 1989, 80, 645–649. [Google Scholar]
  127. Karbowska, M.; Hermanowicz, J.M.; Tankiewicz-Kwedlo, A.; Kalaska, B.; Kaminski, T.W.; Nosek, K.; Wisniewska, R.J.; Pawlak, D. Neurobehavioral Effects of Uremic Toxin-Indoxyl Sulfate in the Rat Model. Sci. Rep. 2020, 10, 9483. [Google Scholar] [CrossRef]
  128. Larsson, S.C.; Traylor, M.; Markus, H.S.; Michaëlsson, K. Serum Parathyroid Hormone, 25-Hydroxyvitamin D, and Risk of Alzheimer’s Disease: A Mendelian Randomization Study. Nutrients 2018, 10, 1243. [Google Scholar] [CrossRef]
  129. Assaraf, M.I.; Diaz, Z.; Liberman, A.; Miller, W.H., Jr.; Arvanitakis, Z.; Li, Y.; Bennett, D.A.; Schipper, H.M. Brain Erythropoietin Receptor Expression in Alzheimer Disease and Mild Cognitive Impairment. J. Neuropathol. Exp. Neurol. 2007, 66, 389–398. [Google Scholar] [CrossRef]
  130. Viviani, B.; Bartesaghi, S.; Corsini, E.; Villa, P.; Ghezzi, P.; Garau, A.; Galli, C.L.; Marinovich, M. Erythropoietin Protects Primary Hippocampal Neurons Increasing the Expression of Brain-Derived Neurotrophic Factor. J. Neurochem. 2005, 93, 412–421. [Google Scholar] [CrossRef]
  131. Ma, R.; Xiong, N.; Huang, C.; Tang, Q.; Hu, B.; Xiang, J.; Li, G. Erythropoietin Protects Pc12 Cells from Beta-Amyloid(25-35)-Induced Apoptosis Via Pi3k/Akt Signaling Pathway. Neuropharmacology 2009, 56, 1027–1034. [Google Scholar] [CrossRef]
  132. Tazangi, P.E.; Moosavi, S.M.S.; Shabani, M.; Haghani, M. Erythropoietin Improves Synaptic Plasticity and Memory Deficits by Decrease of the Neurotransmitter Release Probability in the Rat Model of Alzheimer’s Disease. Pharmacol. Biochem. Behav. 2015, 130, 15–21. [Google Scholar] [CrossRef]
  133. Yuyama, K.; Sun, H.; Sakai, S.; Mitsutake, S.; Okada, M.; Tahara, H.; Furukawa, J.I.; Fujitani, N.; Shinohara, Y.; Igarashi, Y. Decreased Amyloid-Beta Pathologies by Intracerebral Loading of Glycosphingolipid-Enriched Exosomes in Alzheimer Model Mice. J. Biol. Chem. 2014, 289, 24488–24498. [Google Scholar] [CrossRef]
  134. Yuyama, K.; Sun, H.; Mitsutake, S.; Igarashi, Y. Sphingolipid-Modulated Exosome Secretion Promotes Clearance of Amyloid-Beta by Microglia. J. Biol. Chem. 2012, 287, 10977–10989. [Google Scholar] [CrossRef]
  135. Kehoe, P.G.; Miners, S.; Love, S. Angiotensins in Alzheimer’s Disease—Friend or Foe? Trends Neurosci. 2009, 32, 619–628. [Google Scholar] [CrossRef]
  136. Zhu, D.; Shi, J.; Zhang, Y.; Wang, B.; Liu, W.; Chen, Z.; Tong, Q. Central Angiotensin Ii Stimulation Promotes Beta Amyloid Production in Sprague Dawley Rats. PLoS ONE 2011, 6, e16037. [Google Scholar]
  137. Zou, K.; Yamaguchi, H.; Akatsu, H.; Sakamoto, T.; Ko, M.; Mizoguchi, K.; Gong, J.S.; Yu, W.; Yamamoto, T.; Kosaka, K.; et al. Angiotensin-Converting Enzyme Converts Amyloid Beta-Protein 1-42 (Abeta(1-42)) to Abeta(1-40), and Its Inhibition Enhances Brain Abeta Deposition. J. Neurosci. 2007, 27, 8628–8635. [Google Scholar] [CrossRef]
  138. Hu, X.; Liou, A.K.; Leak, R.K.; Xu, M.; An, C.; Suenaga, J.; Shi, Y.; Gao, Y.; Zheng, P.; Chen, J. Neurobiology of Microglial Action in Cns Injuries: Receptor-Mediated Signaling Mechanisms and Functional Roles. Prog. Neurobiol. 2014, 119–120, 60–84. [Google Scholar] [CrossRef]
  139. Prinz, M.; Priller, J. Microglia and Brain Macrophages in the Molecular Age: From Origin to Neuropsychiatric Disease. Nat. Rev. Neurosci. 2014, 15, 300–312. [Google Scholar] [CrossRef]
  140. Cui, C.; Xu, P.; Li, G.; Qiao, Y.; Han, W.; Geng, C.; Liao, D.; Yang, M.; Chen, D.; Jiang, P. Vitamin D Receptor Activation Regulates Microglia Polarization and Oxidative Stress in Spontaneously Hypertensive Rats and Angiotensin Ii-Exposed Microglial Cells: Role of Renin-Angiotensin System. Redox Biol. 2019, 26, 101295. [Google Scholar] [CrossRef]
  141. Jackson, L.; Eldahshan, W.; Fagan, S.C.; Ergul, A. Within the Brain: The Renin Angiotensin System. Int. J. Mol. Sci. 2018, 19, 876. [Google Scholar] [CrossRef] [PubMed]
  142. Biancardi, V.C.; Stranahan, A.M.; Krause, E.G.; de Kloet, A.D.; Stern, J.E. Cross Talk between At1 Receptors and Toll-Like Receptor 4 in Microglia Contributes to Angiotensin Ii-Derived Ros Production in the Hypothalamic Paraventricular Nucleus. Am. J. Physiol. Heart Circ. Physiol. 2016, 310, H404–H415. [Google Scholar] [CrossRef] [PubMed]
  143. Bernstein, K.E.; Koronyo, Y.; Salumbides, B.C.; Sheyn, J.; Pelissier, L.; Lopes, D.H.; Shah, K.H.; Bernstein, E.A.; Fuchs, D.T.; Yu, J.J.Y.; et al. Angiotensin-Converting Enzyme Overexpression in Myelomonocytes Prevents Alzheimer’s-Like Cognitive Decline. J. Clin. Investig. 2014, 124, 1000–1012. [Google Scholar] [CrossRef] [PubMed]
  144. Arroja, M.M.C.; Reid, E.; McCabe, C. Therapeutic Potential of the Renin Angiotensin System in Ischaemic Stroke. Exp. Transl. Stroke Med. 2016, 8, 8. [Google Scholar] [CrossRef]
  145. Regenhardt, R.W.; Desland, F.; Mecca, A.P.; Pioquinto, D.J.; Afzal, A.; Mocco, J.; Sumners, C. Anti-Inflammatory Effects of Angiotensin-(1-7) in Ischemic Stroke. Neuropharmacology 2013, 71, 154–163. [Google Scholar] [CrossRef]
  146. Fouda, A.Y.; Pillai, B.; Dhandapani, K.M.; Ergul, A.; Fagan, S.C. Role of Interleukin-10 in the Neuroprotective Effect of the Angiotensin Type 2 Receptor Agonist, Compound 21, after Ischemia/Reperfusion Injury. Eur. J. Pharmacol. 2017, 799, 128–134. [Google Scholar] [CrossRef]
  147. Liu, M.; Shi, P.; Sumners, C. Direct Anti-Inflammatory Effects of Angiotensin-(1-7) on Microglia. J. Neurochem. 2016, 136, 163–171. [Google Scholar] [CrossRef]
  148. Zenaro, E.; Piacentino, G.; Constantin, G. The Blood-Brain Barrier in Alzheimer’s Disease. Neurobiol. Dis. 2017, 107, 41–56. [Google Scholar] [CrossRef]
  149. Sweeney, M.D.; Zhao, Z.; Montagne, A.; Nelson, A.R.; Zlokovic, B.V. Blood-Brain Barrier: From Physiology to Disease and Back. Physiol. Rev. 2019, 99, 21–78. [Google Scholar] [CrossRef]
  150. Obermeier, B.; Daneman, R.; Ransohoff, R.M. Development, Maintenance and Disruption of the Blood-Brain Barrier. Nat. Med. 2013, 19, 1584–1596. [Google Scholar] [CrossRef]
  151. Maarouf, C.L.; Kokjohn, T.A.; Whiteside, C.M.; Macias, M.P.; Kalback, W.M.; Sabbagh, M.N.; Beach, T.G.; Vassar, R.; Roher, A.E. Molecular Differences and Similarities between Alzheimer’s Disease and the 5xfad Transgenic Mouse Model of Amyloidosis. Biochem. Insights 2013, 6, BCI-S13025. [Google Scholar] [CrossRef]
  152. 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, e003154. [Google Scholar] [CrossRef]
  153. Orihuela, R.; McPherson, C.A.; Harry, G.J. Microglial M1/M2 Polarization and Metabolic States. Br. J. Pharmacol. 2016, 173, 649–665. [Google Scholar] [CrossRef]
  154. Hamed, S.A. Neurologic Conditions and Disorders of Uremic Syndrome of Chronic Kidney Disease: Presentations, Causes, and Treatment Strategies. Expert Rev. Clin. Pharmacol. 2019, 12, 61–90. [Google Scholar] [CrossRef]
  155. Hailpern, S.M.; Melamed, M.L.; Cohen, H.W.; Hostetter, T.H. Moderate Chronic Kidney Disease and Cognitive Function in Adults 20 to 59 Years of Age: Third National Health and Nutrition Examination Survey (Nhanes Iii). J. Am. Soc. Nephrol. 2007, 18, 2205–2213. [Google Scholar] [CrossRef]
  156. McQuillan, R.; Jassal, S.V. Neuropsychiatric Complications of Chronic Kidney Disease. Nat. Rev. Nephrol. 2010, 6, 471–479. [Google Scholar] [CrossRef]
  157. Vannorsdall, T.D.; Jinnah, H.A.; Gordon, B.; Kraut, M.; Schretlen, D.J. Cerebral Ischemia Mediates the Effect of Serum Uric Acid on Cognitive Function. Stroke 2008, 39, 3418–3420. [Google Scholar] [CrossRef]
  158. Tian, M.; Zhu, D.; Xie, W.; Shi, J. Central Angiotensin Ii-Induced Alzheimer-Like Tau Phosphorylation in Normal Rat Brains. FEBS Lett. 2012, 586, 3737–3745. [Google Scholar] [CrossRef] [PubMed]
  159. Jiang, T.; Gao, L.; Shi, J.; Lu, J.; Wang, Y.; Zhang, Y. Angiotensin-(1-7) Modulates Renin-Angiotensin System Associated with Reducing Oxidative Stress and Attenuating Neuronal Apoptosis in the Brain of Hypertensive Rats. Pharmacol. Res. 2013, 67, 84–93. [Google Scholar] [CrossRef]
  160. Watanabe, K.; Sato, E.; Mishima, E.; Watanabe, M.; Abe, T.; Takahashi, N.; Nakayama, M. Effect of Uremic Toxins on Hippocampal Cell Damage: Analysis in Vitro and in Rat Model of Chronic Kidney Disease. Heliyon 2021, 7, e06221. [Google Scholar] [CrossRef]
  161. Haruyama, N.; Fujisaki, K.; Yamato, M.; Eriguchi, M.; Noguchi, H.; Torisu, K.; Tsuruya, K.; Kitazono, T. Improvement in Spatial Memory Dysfunction by Telmisartan through Reduction of Brain Angiotensin Ii and Oxidative Stress in Experimental Uremic Mice. Life Sci. 2014, 113, 55–59. [Google Scholar] [CrossRef] [PubMed]
  162. Burek, M.; Burmester, S.; Salvador, E.; Möller-Ehrlich, K.; Schneider, R.; Roewer, N.; Nagai, M.; Förster, C.Y. Kidney Ischemia/Reperfusion Injury Induces Changes in the Drug Transporter Expression at the Blood-Brain Barrier in Vivo and in Vitro. Front. Physiol. 2020, 11, 569881. [Google Scholar] [CrossRef]
  163. Ohtsuki, S.; Asaba, H.; Takanaga, H.; Deguchi, T.; Hosoya, K.I.; Otagiri, M.; Terasaki, T. Role of Blood-Brain Barrier Organic Anion Transporter 3 (Oat3) in the Efflux of Indoxyl Sulfate, a Uremic Toxin: Its Involvement in Neurotransmitter Metabolite Clearance from the Brain. J. Neurochem. 2002, 83, 57–66. [Google Scholar] [CrossRef] [PubMed]
  164. Wakhloo, D.; Scharkowski, F.; Curto, Y.; Butt, U.J.; Bansal, V.; Steixner-Kumar, A.A.; Wüstefeld, L.; Rajput, A.; Arinrad, S.; Zillmann, M.R.; et al. Functional Hypoxia Drives Neuroplasticity and Neurogenesis Via Brain Erythropoietin. Nat. Commun. 2020, 11, 1313. [Google Scholar] [CrossRef]
  165. Li, G.; Ma, R.; Huang, C.; Tang, Q.; Fu, Q.; Liu, H.; Hu, B.; Xiang, J. Protective Effect of Erythropoietin on Beta-Amyloid-Induced Pc12 Cell Death through Antioxidant Mechanisms. Neurosci. Lett. 2008, 442, 143–147. [Google Scholar] [CrossRef]
  166. Hooshmandi, E.; Motamedi, F.; Moosavi, M.; Katinger, H.; Zakeri, Z.; Zaringhalam, J.; Maghsoudi, A.; Ghasemi, R.; Maghsoudi, N. Cepo-Fc (an Epo Derivative) Protects Hippocampus against Abeta-Induced Memory Deterioration: A Behavioral and Molecular Study in a Rat Model of Abeta Toxicity. Neuroscience 2018, 388, 405–417. [Google Scholar] [CrossRef]
  167. Cevik, B.; Solmaz, V.; Yigitturk, G.; Cavusoğlu, T.; Peker, G.; Erbas, O. Neuroprotective Effects of Erythropoietin on Alzheimer’s Dementia Model in Rats. Adv. Clin. Exp. Med. 2017, 26, 23–29. [Google Scholar] [CrossRef]
  168. Maurice, T.; Mustafa, M.H.; Desrumaux, C.; Keller, E.; Naert, G.; Garcia-Barcelo, M.D.L.C.; Cruz, Y.R.; Rodriguez, J.C.G. Intranasal Formulation of Erythropoietin (Epo) Showed Potent Protective Activity against Amyloid Toxicity in the Abeta25-35 Non-Transgenic Mouse Model of Alzheimer’s Disease. J. Psychopharmacol. 2013, 27, 1044–1057. [Google Scholar] [CrossRef]
  169. Carmona, A.; Guerrero, F.; Buendia, P.; Obrero, T.; Aljama, P.; Carracedo, J. Microvesicles Derived from Indoxyl Sulfate Treated Endothelial Cells Induce Endothelial Progenitor Cells Dysfunction. Front. Physiol. 2017, 8, 666. [Google Scholar] [CrossRef]
  170. Ryu, J.H.; Park, H.; Kim, S.J. The Effects of Indoxyl Sulfate-Induced Endothelial Microparticles on Neointimal Hyperplasia Formation in an Ex Vivo Model. Ann. Surg. Treat. Res. 2017, 93, 11–17. [Google Scholar] [CrossRef]
  171. Alique, M.; Bodega, G.; Corchete, E.; García-Menéndez, E.; de Sequera, P.; Luque, R.; Rodríguez-Padrón, D.; Marqués, M.; Portolés, J.; Carracedo, J.; et al. Microvesicles from Indoxyl Sulfate-Treated Endothelial Cells Induce Vascular Calcification in Vitro. Comput. Struct. Biotechnol. J. 2020, 18, 953–966. [Google Scholar] [CrossRef] [PubMed]
  172. Sinha, M.S.; Ansell-Schultz, A.; Civitelli, L.; Hildesjö, C.; Larsson, M.; Lannfelt, L.; Ingelsson, M.; Hallbeck, M. Alzheimer’s Disease Pathology Propagation by Exosomes Containing Toxic Amyloid-Beta Oligomers. Acta Neuropathol. 2018, 136, 41–56. [Google Scholar] [CrossRef]
  173. McAvoy, K.M.; Sait, H.R.; Marsh, G.; Peterson, M.; Reynolds, T.L.; Gagnon, J.; Geisler, S.; Leach, P.; Roberts, C.; Cahir-McFarland, E.; et al. Cell-Autonomous and Non-Cell Autonomous Effects of Neuronal Bin1 Loss in Vivo. PLoS ONE 2019, 14, e0220125. [Google Scholar] [CrossRef]
  174. Ruan, Z.; Pathak, D.; Venkatesan Kalavai, S.; Yoshii-Kitahara, A.; Muraoka, S.; Bhatt, N.; Takamatsu-Yukawa, K.; Hu, J.; Wang, Y.; Hersh, S.; et al. Alzheimer’s Disease Brain-Derived Extracellular Vesicles Spread Tau Pathology in Interneurons. Brain 2021, 144, 288–309. [Google Scholar] [CrossRef]
  175. Kohan, D.E.; Barton, M. Endothelin and Endothelin Antagonists in Chronic Kidney Disease. Kidney Int. 2014, 86, 896–904. [Google Scholar] [CrossRef]
  176. Eroglu, E.; Kocyigit, I.; Lindholm, B. The Endothelin System as Target for Therapeutic Interventions in Cardiovascular and Renal Disease. Clin. Chim. Acta 2020, 506, 92–106. [Google Scholar] [CrossRef]
  177. Heerspink, H.J.; Xie, D.; Bakris, G.; Correa-Rotter, R.; Hou, F.F.; Kitzman, D.W.; Kohan, D.; Makino, H.; McMurray, J.J.; Perkovic, V.; et al. Early Response in Albuminuria and Long-Term Kidney Protection During Treatment with an Endothelin Receptor Antagonist: A Prespecified Analysis from the Sonar Trial. J. Am. Soc. Nephrol. 2021, 32, 2900–2911. [Google Scholar] [CrossRef]
  178. Hsu, Y.H.; Zheng, C.M.; Chou, C.L.; Chen, Y.J.; Lee, Y.H.; Lin, Y.F.; Chiu, H.W. Therapeutic Effect of Endothelin-Converting Enzyme Inhibitor on Chronic Kidney Disease through the Inhibition of Endoplasmic Reticulum Stress and the Nlrp3 Inflammasome. Biomedicines 2021, 9, 398. [Google Scholar] [CrossRef]
  179. Palmer, J.C.; Barker, R.; Kehoe, P.G.; Love, S. Endothelin-1 Is Elevated in Alzheimer’s Disease and Upregulated by Amyloid-Beta. J. Alzheimers Dis. 2012, 29, 853–861. [Google Scholar] [CrossRef]
  180. Karakaya, E.; Abdul, Y.; Chowdhury, N.; Wellslager, B.; Jamil, S.; Albayram, O.; Yilmaz, Ö.; Ergul, A. Porphyromonas Gingivalis Infection Upregulates the Endothelin (Et) System in Brain Microvascular Endothelial Cells. Can. J. Physiol. Pharmacol. 2022, 100, 679–688. [Google Scholar] [CrossRef]
  181. Gulati, A.; Hornick, M.G.; Briyal, S.; Lavhale, M.S. A Novel Neuroregenerative Approach Using Et(B) Receptor Agonist, Irl-1620, to Treat Cns Disorders. Physiol. Res. 2018, 67 (Suppl. 1), S95–S113. [Google Scholar] [CrossRef]
  182. Pacheco-Quinto, J.; Eckman, E.A. Endothelin-Converting Enzymes Degrade Intracellular Beta-Amyloid Produced within the Endosomal/Lysosomal Pathway and Autophagosomes. J. Biol. Chem. 2013, 288, 5606–5615. [Google Scholar] [CrossRef] [PubMed]
  183. Pacheco-Quinto, J.; Eckman, C.B.; Eckman, E.A. Major Amyloid-Beta-Degrading Enzymes, Endothelin-Converting Enzyme-2 and Neprilysin, Are Expressed by Distinct Populations of Gabaergic Interneurons in Hippocampus and Neocortex. Neurobiol. Aging 2016, 48, 83–92. [Google Scholar] [CrossRef] [PubMed]
Figure 1. CKD and AD pathogenesis are closely related through the RAS.
Figure 1. CKD and AD pathogenesis are closely related through the RAS.
Jcm 12 01459 g001
Figure 2. RAS involvement in AD. Components of the RAS bind with AT1R to cause injury to the BBB, resulting in increased cytokine release, the numbers of M1 microglia, and Aβ accumulation.
Figure 2. RAS involvement in AD. Components of the RAS bind with AT1R to cause injury to the BBB, resulting in increased cytokine release, the numbers of M1 microglia, and Aβ accumulation.
Jcm 12 01459 g002
Table 1. The clinical studies that assess the association between CKD and AD.
Table 1. The clinical studies that assess the association between CKD and AD.
FactorAttributesReference
Renin-Angiotensin SystemACE1ACE1 accumulates in severe AD patients.Miners et al.
[118]
ACE2ACE2 activated, causes lower hippocampal Aβ and restored cognitionKehoe et al.
[119]
ACEIACEI was higher in the temporal cortex of Alzheimer’s patientsBarnes et al.
[120]
ACEIACEI causes a slower rate of cognitive declineSoto et al.
[121]
ARBsARBs significantly reduce the incidence and progression of Alzheimer’s disease
and dementia
Li et al.
[122]
ARBsARBs rescue cerebrovascular and
cognitive function in adults.
Ongali et al.
[123]
ARBsARBs could improve cognitive function, in particular immediate and delayed memory.Fogari et al.
[124]
ACEI and ARBsARBs may have greater cognition protective effects than ACEIFournier et al.
[125]
Uremic toxinsUric AcidSystemic hyperuricemia induces cognitive dysfunctionLemma et al.
[126]
Indoxyl SulfateChronic exposure to Indoxyl Sulfate leads to reduced locomotor activity and spatial memory, as well as increased stress sensitivity, and apathetic behaviorKarbowska et al.
[127]
Parathyroid
hormone
Parathyroid hormone decline brain impairment by
vitamin D
Larsson et al.
[128]
ErythropoietinEPOEpo-EpoR significant cytoprotection by
antioxidant, antiapoptotic, anti-inflammatory, neurotrophic, angiogenic, and synaptogenic activities.
Assaraf et al. [129]
Viviani et al. [130]
Ma et al. [131]
Esmaeili Tazangi et al.
[132]
Extracellular
Vehicles
EVsEVs contribute to peptide clearance from the
extracellular space and reducing
Aβ pathology
Yuyama et al.
[133,134]
Soares Martins et al. [80]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Ma, K.; Zheng, Z.-R.; Meng, Y. Pathogenesis of Chronic Kidney Disease Is Closely Bound up with Alzheimer’s Disease, Especially via the Renin-Angiotensin System. J. Clin. Med. 2023, 12, 1459. https://doi.org/10.3390/jcm12041459

AMA Style

Ma K, Zheng Z-R, Meng Y. Pathogenesis of Chronic Kidney Disease Is Closely Bound up with Alzheimer’s Disease, Especially via the Renin-Angiotensin System. Journal of Clinical Medicine. 2023; 12(4):1459. https://doi.org/10.3390/jcm12041459

Chicago/Turabian Style

Ma, Ke, Zi-Run Zheng, and Yu Meng. 2023. "Pathogenesis of Chronic Kidney Disease Is Closely Bound up with Alzheimer’s Disease, Especially via the Renin-Angiotensin System" Journal of Clinical Medicine 12, no. 4: 1459. https://doi.org/10.3390/jcm12041459

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop