Minireviews Open Access
Copyright ©The Author(s) 2025. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Exp Med. Sep 20, 2025; 15(3): 106743
Published online Sep 20, 2025. doi: 10.5493/wjem.v15.i3.106743
Potential role of nanopharmacology in reducing neuroinflammation associated with hypertension and metabolic disorders
Virna Margarita Martín Giménez, Instituto de Investigaciones en Ciencias Químicas, Facultad de Ciencias Químicas y Tecnológicas, Universidad Católica de Cuyo, San Juan 5400, Argentina
Sebastián García Menéndez, Department of Pharmacology, Instituto de Medicina y Biología Experimental de Cuyo, Consejo Nacional de Investigación Científica y Tecnológica, Mendoza 5500, Argentina
Raúl Lelio Sanz, Department of Pathology, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza 5500, Argentina
Máximo Schiavone, Department of Medicina, Universidad Austral, Ciudad Autónoma de Buenos Aires C1010AAZ, Argentina
Leon Ferder, Department of Pharmacology, Universidad Maimónides, Ciudad Autónoma de Buenos Aires C1405, Argentina
Felipe Inserra, Department of Nephrology, Universidad de Maimónides, Ciudad Autónoma de Buenos Aires C1405, Argentina
Walter Manucha, Department of Pathology, Instituto de Medicina y Biología Experimental de Cuyo, Consejo Nacional de Investigación Científica y Tecnológica Universidad Nacional de Cuyo, Laboratorio de Farmacología Experimental Básica y Traslacional, Facultad de Ciencias Médicas, Mendoza 5500, Argentina
ORCID number: Walter Manucha (0000-0002-2279-7626).
Co-first authors: Virna Margarita Martín Giménez and Sebastián García Menéndez.
Author contributions: All authors contributed to the writing, discussion and preparation of this manuscript.
Supported by Agencia Nacional de Promoción de la Investigación, el Desarrollo Tecnológico y la Innovación, No. PICT 2020 Serie A 4000.
Conflict-of-interest statement: The authors declare that they have no conflict of interest.
Open Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Walter Manucha, PhD, Professor, Department of Pathology, Instituto de Medicina y Biología Experimental de Cuyo, Consejo Nacional de Investigación Científica y Tecnológica Universidad Nacional de Cuyo, Laboratorio de Farmacología Experimental Básica y Traslacional, Facultad de Ciencias Médicas, Farmacología, Mendoza 5500, Argentina. wmanucha@yahoo.com.ar
Received: March 11, 2025
Revised: April 17, 2025
Accepted: May 26, 2025
Published online: September 20, 2025
Processing time: 154 Days and 20.3 Hours

Abstract

Hypertension disrupts cerebral blood flow, leading to endothelial dysfunction, breakdown of the blood-brain barrier (BBB), and inflammatory cell infiltration. This cascade triggers glial cell activation, increases oxidative stress, and causes pro-inflammatory cytokine release, creating a neurotoxic environment. In this context, we explore the intricate connection between hypertension, neuroinflammation, and neurodegeneration, as well as how hypertension interacts with other metabolic disorders, such as obesity and diabetes, to further worsen neuroinflammation. Additionally, we discuss the role of the renin-angiotensin-aldosterone system, the impact of the microbiome, and the potential contribution of chronic infections in exacerbating neuroinflammation. It is essential to emphasize the potential of nanotechnology to transform therapeutic approaches. Nanoparticle-based drug delivery systems can enhance the bioavailability and selectivity of antihypertensive drugs, antioxidants, and neuroprotective compounds, enabling targeted delivery across the BBB. By combining effective blood pressure management with nanotechnology-enabled therapies that modulate inflammation, oxidative stress, and protein aggregation, we can explore new avenues for preventing and treating hypertension and metabolic disorder-associated neurodegenerative conditions. Ultimately, hypertension significantly contributes to neuroinflammation and neurodegeneration by promoting neuronal cell death, primarily through impaired cerebral blood flow and disruption of the BBB. The interaction of hypertension with metabolic disorders exacerbates these effects. However, advancements in our understanding and new technologies reveal promising nanopharmacological approaches for targeted drug delivery to the brain, thereby improving treatment outcomes, enhancing adherence, and reducing side effects.

Key Words: Neuroinflammation; Nanotechnology/nanopharmacology; Blood-brain barrier; Oxidative stress; Neurodegeneration; Hypertension; Metabolic disorders

Core Tip: This mini-review unveils the intricate nexus between hypertension, metabolic disorders, and neurodegeneration, spotlighting neuroinflammation as a pivotal mediator. Compromised cerebral blood flow and blood-brain barrier (BBB) disruption initiate a cascade of oxidative stress and pro-inflammatory cytokine release, culminating in neuronal injury. We advocate for the transformative potential of nanopharmacology. Nanoparticle-mediated drug delivery offers a strategic avenue to enhance therapeutic bioavailability and selectivity, enabling targeted intervention across the BBB. By synergizing effective blood pressure management with nanotechnology-driven modulation of neuroinflammation, we pioneer novel strategies to mitigate neurodegenerative pathologies. This paradigm shift holds promise for revolutionizing therapeutic approaches, paving the way for improved patient outcomes in hypertension-associated neurological disorders.



INTRODUCTION

High blood pressure is one of the main cardiovascular risk factors. It affects a significant percentage of the world's population, increasing the risk of cerebrovascular events, cognitive impairment, and neurodegenerative diseases[1]. While the systemic consequences of hypertension on the heart, kidneys, and large blood vessels are well documented, it has become clear that it also exerts a profound impact on the brain[2,3]. Hypertension promotes microvascular alterations, endothelial dysfunction, reduced cerebral blood flow, glial cell activation, and proinflammatory mediator release, culminating in a chronic neuroinflammatory condition[4,5].

It is well known that high blood pressure is often associated with several or all the components of metabolic syndrome (abdominal obesity, hyperlipidemia, and elevated fasting blood glucose levels). The consequence of this association is lack of cardiovascular-renal-metabolic health[6]. Together with high blood pressure, these factors generate progressive, inflammatory-based changes in various tissues, including neuroinflammation[7].

Neuroinflammation is the persistent activation of astrocytes and microglia and the release of cytokines, chemokines, and reactive oxygen species (ROS), which creates a hostile environment for neurons[8,9]. This chronic inflammatory state facilitates the accumulation of pathological proteins, such as beta-amyloid or hyperphosphorylated tau, accelerates neuronal loss, and ultimately contributes to neurodegenerative disease pathogenesis, including Alzheimer's disease (AD), Parkinson's disease (PD), and other cognitive disorders[10,11].

Reducing blood pressure attenuates neuroinflammation. In fact, some anti-hypertension drugs, like renin-angiotensin system blockers and calcium antagonists, positively affect neural inflammation[12-14].

New treatments used to control metabolic syndrome and diabetes mellitus, like sodium glucose cotransporter 2 inhibitors and glucagon-like peptide-1 agonists, mildly reduce blood pressure. Chronic use of these treatments has attenuated neuroinflammation, dementia, and AD[15-18].

In addition to conventional antihypertensive and new metabolic drug interventions, recent research has begun to explore nanodrugs and nanoparticle delivery systems to improve the bioavailability, selectivity, and efficacy of compounds with neuroprotective and anti-inflammatory potential effects[19,20]. These approaches could help overcome biological barriers, such as the blood-brain barrier (BBB), and offer new therapeutic alternatives to mitigate neuroinflammation and neuronal damage associated with hypertension and some metabolic derangements.

This mini-review aims to integrate evidence from the last 5 years on the relationship between hypertension, neuroinflammation, and neurodegeneration. We will explore the vascular and molecular mechanisms that link high blood pressure with brain damage, address the key role of glia and inflammatory signaling pathways, and discuss emerging therapeutic implications, including the potential application of nanopharmaceuticals. In doing so, we will generate a comprehensive view to guide us toward new strategies for preventing and treating neurological deterioration associated with the previously described conditions.

IMPACT OF HYPERTENSION ON CEREBRAL VASCULATURE

Hypertension-induced vascular damage constitutes a fundamental starting point in the pathophysiological cascade that leads to neuroinflammation and neurodegeneration. Chronic hypertension alters arteriolar caliber, thickens vascular walls, and disrupts blood flow autoregulation. These changes facilitate endothelial dysfunction, increasing BBB permeability and allowing immune cell and inflammatory mediator infiltration into the brain parenchyma[21].

A notable example is the study that used mouse models deficient in the Collagen type XVIIIa1 gene to simulate early cerebral small vessel disease microvascular lesions[22]. These animals exhibited microvascular damage, extracellular matrix remodeling, neuroinflammation, and early glial activation, suggesting that hypertension contributes to a damaging environment for neurons from an early stage. Glial activation is accompanied by cytokine, chemokine, and growth factor release that perpetuates the inflammatory state[8].

The retina is a neurosensory tissue with vasculature like the brain, acting as an accessible model to understand the effects of hypertension. Studies in ocular hypertension provide evidence analogous to that in the brain. Yu et al[23] found that increased intraocular pressure induced retinal neurodegeneration through activation of p38/mitogen-activated protein kinases (MAPK) pathways, promoting ganglion cell death and neuroinflammation. Similarly, Shi et al[9] revealed that pathologically high intraocular pressure leads to reactive glial proliferation, impaired blood-retinal barrier, and activation of the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX2)/endothelin-1/extracellular regulated kinase 1/2 (ERK1/2) pathway, contributing to inflammation and neuronal damage.

These retinal models reflect processes like those observed in the brain, reinforcing that hypertension triggers an inflammatory cascade capable of damaging nervous tissue, regardless of region. Thus, the retina can be considered a model to study the early detection of vascular and neuroinflammatory changes in the brain.

MOLECULAR AND CELLULAR MECHANISMS OF NEUROINFLAMMATION

Neuroinflammation associated with hypertensive and metabolic diseases is a complex phenomenon involving multiple molecular pathways. Oxidative stress plays a key role, with NOX2 inducing ROS generation[24]. These reactive species trigger lipid, protein, and genomic damage and activate proinflammatory signaling pathways, including p38/MAPK and ERK1/2[25].

Recent studies have elucidated how these pathways contribute to glial cell inflammation. Yang et al[8] highlighted a role for the cellular FLICE-like inhibitory protein in regulating astroglial inflammation in an experimental model of glaucoma, indicating that ocular hypertension relies on intracellular mediators to orchestrate the inflammatory response in glia.

However, hypertension does not act in isolation; it is integrated into an adverse cardiometabolic environment where obesity, diabetes, and dyslipidemia converge. Patel and Edison[1] analyzed how hyperinsulinemia and insulin resistance potentiate vascular dysfunction by impairing nitric oxide signaling, exacerbating renin-angiotensin-aldosterone system (RAAS) overactivation. This environment sensitizes glial cells to inflammatory triggers and enhances BBB permeability, amplifying central nervous system (CNS) vulnerability and favoring neuroinflammatory cascades that drive cognitive decline[1].

In parallel, elevated levels of free fatty acids (FFAs), commonly present in obesity and metabolic syndromes, activate toll-like receptors (TLRs), particularly TLR4, on microglia and endothelial cells. This leads to upregulation of nuclear factor-kappa B signaling and downstream inflammatory mediators, further aggravating the hypertensive milieu[26]. FFAs also increase mitochondrial dysfunction and contribute to dysregulated phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) signaling, reducing neuroprotective pathway activation and exacerbating oxidative neuronal injury[27].

Emerging evidence also underscores a role for gut dysbiosis in this process. Disruptions in gut microbiota composition reduce neuroprotective short-chain fatty acid production, while increasing circulating endotoxins like lipopolysaccharides (LPS)[28]. These microbial-derived products can translocate into the bloodstream, particularly under hypertensive conditions that impair gut barrier integrity. Once in circulation, they activate systemic inflammation and penetrate the brain through a compromised BBB, enhancing glial reactivity and cytokine production. This "gut–brain axis" represents a potent amplifier of neuroinflammation, especially when intertwined with hypertension-induced vascular vulnerability[29].

On the other hand, Kountouras et al[2] addressed the impact of Helicobacter pylori and metabolic syndrome on mast cell activation and neurodegenerative pathophysiology, showing that hypertension is part of a broader network of systemic inflammatory factors.

RAAS is another critical axis; Tiwari et al[3] showed that angiotensin-converting enzyme 2 modulation in hypertensive models promotes neurogenesis and attenuated the inflammatory impact in the hippocampus through Wnt/β-catenin signaling. Likewise, Gouveia et al[4] found that intranasal administration of irbesartan, an angiotensin II receptor antagonist, reverses cognitive impairment and neuroinflammation in mice with LPS-induced inflammation, activating the PI3K/AKT pathway.

In addition to vascular and metabolic factors, there are other interaction routes. Li et al[30] suggested that chronic oral infection by periodontal pathogens can trigger an oral-brain axis, favoring neuroinflammation and increasing the risk of AD. Hypertension can potentially alter protective barriers, making the CNS more vulnerable to infectious insults and enhancing their harmful effect. The molecular and cellular mechanisms of neuroinflammation in the hypertension context are summarized in Table 1.

Table 1 Interplay between hypertension, molecular pathways, and neuroinflammation.
Stage
Pathway/factor
Role/mechanism
Crosstalk
Neuroinflammatory outcome
HypertensionInitiates vascular injury, oxidative stress, and endothelial dysfunctionTriggers activation of RAAS, NADPH oxidase, and other stress-response pathwaysCompromised blood-brain barrier, vascular leakage
Vascular damageRAASAngiotensin II vasoconstriction, oxidative stress, proinflammatory signalingActivates NADPH oxidase, p38/MAPK, suppresses ACE2Endothelial dysfunction, proinflammatory environment
Oxidative stressNADPH oxidaseProduces ROS in response to angiotensin II and inflammationROS activates p38/MAPK, ERK1/2; contributes to Wnt/β-catenin and PI3K/AKT dysregulationPromotes inflammation, cell damage
Inflammatory signal transductionp38/MAPKInduces cytokine release (e.g., interleukin-6, tumor necrosis factor-alpha), microglial activationInteracts with ERK1/2 and Wnt/β-catenin; enhanced by ROSNeuroinflammation, synaptic dysfunction
ERK1/2Regulates gene expression under oxidative and inflammatory stressCrosstalk with p38/MAPK and PI3K/AKTGlial activation, neurotoxicity
PI3K/AKTNormally protective (anti-apoptotic); dysregulated under chronic stressInteracts with ERK1/2, influenced by oxidative stressLoss of neuroprotection, increased cell vulnerability
Anti-inflammatory counteractionACE2Converts angiotensin II to angiotensin-(1-7), countering RAAS effectsOpposes RAAS; downregulated in hypertensionLoss of balance sustained inflammation
Cellular remodelingWnt/β-cateninControls cell survival, synaptic plasticity; dysregulated in neuroinflammationActivated by MAPK/ROS; contributes to chronic glial activationSynaptic loss, glial scarring
Cumulative effectIntegrated pathwaysChronic activation of RAAS, MAPK, NADPH oxidase, Wnt, and ERK1/2Synergistic feedback loops worsen endothelial and neuronal stressSustained neuroinflammation, cognitive decline, neurodegeneration
CONVERGING PATHWAYS IN INFLAMMATION AND NEURODEGENERATION:

Long-term inflammation creates a neurotoxic environment that is permissible for neurodegeneration. Chronic activation of astrocytes and microglia, continuous release of proinflammatory cytokines, such as interleukin-1β and tumor necrosis factor-alpha, and oxidative stress contribute to synaptic dysfunction, neuronal loss, and accumulation of aberrant proteins such as hyperphosphorylated tau. These alterations are standard pathological features in various neurodegenerative diseases[31].

El-Desouky et al[10] highlighted the relevance of intervening on the inflammatory axis and protein aggregation. Compound RF26 is a new phosphodiesterase type 5 (PDE5) inhibitor that improves memory and reduces tau aggregation in a tauopathy model. These findings suggest that modulating inflammatory and vascular pathways can attenuate or reverse some aspects of neurodegenerative deterioration[10].

At the clinical level, hypertension is one of several factors that converge in neurodegeneration. Khalil et al[11] addressed cardiovascular dysautonomia in PD, demonstrating that vascular and autonomic alterations may contribute to cognitive decline. In AD, Patel and Edison[1] highlight the relationship between hypertension, obesity, diabetes, and amyloid protein accumulation, reinforcing the concept that cardiometabolic conditions generate an inflammatory microenvironment that precipitates or accelerates neurodegeneration.

Interaction with pathogens or the microbiome adds complexity[30][2]. These factors can ignite or perpetuate inflammation in a brain already vulnerable from hypertension.

THERAPEUTIC INTERVENTIONS AND FUTURE PERSPECTIVES

In addition to blood pressure control, therapeutic approaches should be aimed at interrupting the inflammatory and oxidative cascades that mediate brain damage. In this regard, nanotechnology-based drug delivery systems[20] have emerged as a promising strategy to improve the efficacy and selectivity of antihypertensive and neuroprotective therapies[32-36].

In this sense, inhibition of prooxidative and proinflammatory pathways, such as in the combination of NADPH and N-acetylcysteine, attenuates neurodegeneration and neuroinflammation[23]. Additionally, including phytochemicals or antioxidant molecules in nanoparticles could improve their bioavailability and effectively target damaged nervous tissue[19,37,38].

Another possible alternative is modulation of the RAAS and associated pathways. Nanoparticles can improve the delivery of RAAS inhibitors, increasing their stability and ability to cross the BBB[27]. For example, nanoparticles targeting smooth muscle cells in the pulmonary vasculature have alleviated hypoxic vasoconstriction[39]. Similarly, encapsulating anti-hypertensive compounds in nanometric systems like polyethylene glycol-poly lactic acid-co-glycolic acid (PEG-PLGA) nanocarriers can enhance their efficacy and reduce doses and side effects[40]. Indeed, pharmacokinetic results for PEG-PLGA nanoparticles loaded with isoliensinine have shown promising results, demonstrating enhanced stability, sustained release, and improved bioavailability compared to free isoliensinine in an in vivo model of hypertension (angiotensin II-induced mice). This nanoparticle system significantly increased drug cellular uptake and targeted delivery, particularly in vascular smooth muscle cells, leading to greater efficacy in reducing angiotensin II-induced hypertension[40]. While there was no report of toxicity and the delivery system shows potential in clinical applications, further studies are necessary to confirm long-term safety, scalability, and efficacy across diverse patient populations.

This approach is also being evaluated for its utility in inhibiting protein aggregation and related metabolic pathways. In addition to compounds such as RF26 (a novel PDE5 inhibitor)[10], co-administration of nanoparticles with anti-inflammatory drugs, antioxidants, or vitamin D analogues can block key inflammatory and metabolic pathways[33-36,41] to prevent neurodegeneration[42].

Therapeutic and phytotherapeutic combinations are interesting approaches. Nanodrugs that release natural compounds (phytochemicals, antioxidants) or hormones, such as melatonin or anandamide (AEA), could offer synergistic effects. For example, AEA encapsulated in polymeric micelles has shown beneficial renal effects in hypertension[35], improving the pharmacokinetic profile of AEA, targeting to renal cells and prolonged anti-hypertensive effects in spontaneously hypertensive rats. The nanomicelles enhanced AEA bioavailability and induced sustained natriuretic and diuretic responses compared to free AEA, which lost efficacy over time. Nevertheless, the limited sample size and the absence of long-term safety data present key limitations, emphasizing the need for further studies to validate clinical relevance and optimize organ-specific delivery strategies. Likewise, thymoquinone or indole-3-carbinol could potentially enhance AEA’s anti-inflammatory and antioxidant actions[43,44].

These interventions suggest the need for a multifactorial approach. Nanotechnology offers tools to achieve targeted release, improve the permeability of biological barriers, and reduce systemic drug toxicity. Despite the promising therapeutic potential of pharmaceutical nanoformulations for hypertension and neuroinflammation, several barriers hinder their clinical adoption. Key challenges include concerns about long-term toxicity, limited scalability of manufacturing processes, and complex regulatory pathways due to the novelty and variability of nanocarriers. Additionally, the lack of extensive clinical trials—many still in preclinical or early-phase stages—restricts comprehensive safety and efficacy data. To address these issues, rigorous biocompatibility testing, standardized manufacturing protocols, and adaptive regulatory frameworks are essential. Incorporating personalized dosing strategies based on patient-specific pharmacokinetics and biomarker profiles may further enhance treatment outcomes and minimize adverse effects, paving the way for safer, more targeted nano-based therapies.

CONCLUSION

Taken together, it is clear that hypertension damages the peripheral cardiovascular system and exerts a direct and multifaceted impact on the central nervous system. Elevated blood pressure causes microvascular alterations, endothelial dysfunction, and breakdown of protective barriers, which sets a favorable scenario for glial activation, oxidative stress, and proinflammatory cytokine release. This chronic inflammatory environment promotes neuronal degeneration, facilitating the onset and progression of neurodegenerative diseases.

Multiple molecular pathways participate in this cascade, including p38/MAPK, ERK1/2, NOX2, RAAS, and Wnt/β-catenin, in addition to contributions by metabolic, infectious, and microbiome factors. Nanotechnology has emerged as a promising tool to improve the efficacy of therapies, facilitate the targeted delivery of anti-inflammatory, antioxidant, or neuroprotective drugs, and potentially reverse or slow hypertension-associated neurodegeneration.

Emerging therapeutic strategies suggest that tight blood pressure control combined with anti-inflammatory, antioxidant, metabolic pathway modulating and nanopharmaceutical-based interventions may open new routes towards preventing and treating cognitive decline. Future research should focus on validating these approaches in clinical trials, optimizing nanometric formulations and developing biomarkers to guide personalized therapies to improve the quality of life of affected individuals.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Medicine, research and experimental

Country of origin: Argentina

Peer-review report’s classification

Scientific Quality: Grade C

Novelty: Grade D

Creativity or Innovation: Grade D

Scientific Significance: Grade D

P-Reviewer: Wu W S-Editor: Luo ML L-Editor: Filipodia P-Editor: Zhao YQ

References
1.  Patel V, Edison P. Cardiometabolic risk factors and neurodegeneration: a review of the mechanisms underlying diabetes, obesity and hypertension in Alzheimer's disease. J Neurol Neurosurg Psychiatry. 2024;95:581-589.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 30]  [Article Influence: 30.0]  [Reference Citation Analysis (0)]
2.  Kountouras J, Boziki M, Kazakos E, Theotokis P, Kesidou E, Nella M, Bakirtzis C, Karafoulidou E, Vardaka E, Mouratidou MC, Kyrailidi F, Tzitiridou-Chatzopoulou M, Orovou E, Giartza-Taxidou E, Deretzi G, Grigoriadis N, Doulberis M. Impact of Helicobacter pylori and metabolic syndrome on mast cell activation-related pathophysiology and neurodegeneration. Neurochem Int. 2024;175:105724.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 9]  [Reference Citation Analysis (0)]
3.  Tiwari P, Mueed S, Abdulkareem AO, Hanif K. Activation of angiotensin converting enzyme 2 promotes hippocampal neurogenesis via activation of Wnt/β-catenin signaling in hypertension. Mol Cell Neurosci. 2024;130:103953.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 7]  [Reference Citation Analysis (0)]
4.  Gouveia F, Fonseca C, Silva A, Camins A, Teresa Cruz M, Ettcheto M, Fortuna A. Intranasal irbesartan reverts cognitive decline and activates the PI3K/AKT pathway in an LPS-induced neuroinflammation mice model. Int Immunopharmacol. 2024;128:111471.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 7]  [Reference Citation Analysis (0)]
5.  Rombaut A, Brautaset R, Williams PA, Tribble JR. Intravitreal injection of the Galectin-3 inhibitor TD139 provides neuroprotection in a rat model of ocular hypertensive glaucoma. Mol Brain. 2024;17:84.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
6.  Ndumele CE, Rangaswami J, Chow SL, Neeland IJ, Tuttle KR, Khan SS, Coresh J, Mathew RO, Baker-Smith CM, Carnethon MR, Despres JP, Ho JE, Joseph JJ, Kernan WN, Khera A, Kosiborod MN, Lekavich CL, Lewis EF, Lo KB, Ozkan B, Palaniappan LP, Patel SS, Pencina MJ, Powell-Wiley TM, Sperling LS, Virani SS, Wright JT, Rajgopal Singh R, Elkind MSV; American Heart Association. Cardiovascular-Kidney-Metabolic Health: A Presidential Advisory From the American Heart Association. Circulation. 2023;148:1606-1635.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 108]  [Cited by in RCA: 460]  [Article Influence: 230.0]  [Reference Citation Analysis (0)]
7.  Ali NH, Al-Kuraishy HM, Al-Gareeb AI, Alexiou A, Papadakis M, Bahaa MM, Alibrahim F, Batiha GE. New insight on the potential detrimental effect of metabolic syndrome on the Alzheimer disease neuropathology: Mechanistic role. J Cell Mol Med. 2024;28:e70118.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 6]  [Reference Citation Analysis (0)]
8.  Yang X, Zeng Q, İnam MG, İnam O, Lin CS, Tezel G. cFLIP in the molecular regulation of astroglia-driven neuroinflammation in experimental glaucoma. J Neuroinflammation. 2024;21:145.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
9.  Shi X, Li P, Herb M, Liu H, Wang M, Wang X, Feng Y, van Beers T, Xia N, Li H, Prokosch V. Pathological high intraocular pressure induces glial cell reactive proliferation contributing to neuroinflammation of the blood-retinal barrier via the NOX2/ET-1 axis-controlled ERK1/2 pathway. J Neuroinflammation. 2024;21:105.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 9]  [Reference Citation Analysis (0)]
10.  El-Desouky S, Abdel-Halim M, Fathalla RK, Abadi AH, Piazza GA, Salama M, El-Khodery SA, Youssef MA, Elfarrash S. A novel phosphodiesterase 5 inhibitor, RF26, improves memory impairment and ameliorates tau aggregation and neuroinflammation in the P301S tauopathy mouse model of Alzheimer's disease. Exp Neurol. 2025;384:115058.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
11.  Khalil I, Sayad R, Kedwany AM, Sayed HH, Caprara ALF, Rissardo JP. Cardiovascular dysautonomia and cognitive impairment in Parkinson's disease (Review). Med Int (Lond). 2024;4:70.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 7]  [Reference Citation Analysis (0)]
12.  Nist KM, Bard H, McBride B, Capriglione AL, Moreira JD, Farb DH, Wainford RD. Losartan attenuates sex-dependent hypertension, neuroinflammation, and cognitive impairment in the aging male sprague-dawley rat. Geroscience.  2024.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
13.  Zhang W, Jiao B, Yu S, Zhang K, Sun J, Liu B, Zhang X. Spinal AT1R contributes to neuroinflammation and neuropathic pain via NOX2-dependent redox signaling in microglia. Free Radic Biol Med. 2025;227:143-156.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 3]  [Cited by in RCA: 4]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
14.  Kerkhofs D, Helgers R, Hermes D, Steinbusch HPJ, Van Essen H, Leenders P, Prickaerts J, Staals J, Biessen EA, Van Oostenbrugge RJ, Foulquier S. Amlodipine limits microglia activation and cognitive dysfunction in aged hypertensive mice. J Hypertens. 2023;41:1159-1167.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 7]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
15.  Youn YJ, Kim S, Jeong HJ, Ah YM, Yu YM. Sodium-glucose cotransporter-2 inhibitors and their potential role in dementia onset and cognitive function in patients with diabetes mellitus: a systematic review and meta-analysis. Front Neuroendocrinol. 2024;73:101131.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 24]  [Reference Citation Analysis (0)]
16.  Hung CH, Lu LY. New Insights into the Role of SGLT-2 Inhibitors in the Prevention of Dementia. Neurol Int. 2024;16:1717-1730.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 7]  [Reference Citation Analysis (0)]
17.  Du H, Meng X, Yao Y, Xu J. The mechanism and efficacy of GLP-1 receptor agonists in the treatment of Alzheimer's disease. Front Endocrinol (Lausanne). 2022;13:1033479.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 2]  [Cited by in RCA: 55]  [Article Influence: 18.3]  [Reference Citation Analysis (0)]
18.  Lin MH, Cheng PC, Hsiao PJ, Chen SC, Hung CH, Kuo CH, Huang SK, Clair Chiou HY. The GLP-1 receptor agonist exenatide ameliorates neuroinflammation, locomotor activity, and anxiety-like behavior in mice with diet-induced obesity through the modulation of microglial M2 polarization and downregulation of SR-A4. Int Immunopharmacol. 2023;115:109653.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 22]  [Reference Citation Analysis (0)]
19.  Bakrim S, Aboulaghras S, El Menyiy N, El Omari N, Assaggaf H, Lee LH, Montesano D, Gallo M, Zengin G, AlDhaheri Y, Bouyahya A. Phytochemical Compounds and Nanoparticles as Phytochemical Delivery Systems for Alzheimer's Disease Management. Molecules. 2022;27:9043.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 11]  [Reference Citation Analysis (0)]
20.  Girigoswami K, Arunkumar R, Girigoswami A. Management of hypertension addressing hyperuricaemia: introduction of nano-based approaches. Ann Med. 2024;56:2352022.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
21.  Iadecola C, Davisson RL. Hypertension and cerebrovascular dysfunction. Cell Metab. 2008;7:476-484.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 422]  [Cited by in RCA: 392]  [Article Influence: 23.1]  [Reference Citation Analysis (0)]
22.  Khoshneviszadeh M, Henneicke S, Pirici D, Senthilnathan A, Morton L, Arndt P, Kaushik R, Norman O, Jukkola J, Dunay IR, Seidenbecher C, Heikkinen A, Schreiber S, Dityatev A. Microvascular damage, neuroinflammation and extracellular matrix remodeling in Col18a1 knockout mice as a model for early cerebral small vessel disease. Matrix Biol. 2024;128:39-64.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 10]  [Reference Citation Analysis (0)]
23.  Yu N, Wu X, Zhang C, Qin Q, Gu Y, Ke W, Liu X, Zhang Q, Liu Z, Chen M, Wang K. NADPH and NAC synergistically inhibits chronic ocular hypertension-induced neurodegeneration and neuroinflammation through regulating p38/MAPK pathway and peroxidation. Biomed Pharmacother. 2024;175:116711.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 3]  [Reference Citation Analysis (0)]
24.  Lassègue B, Griendling KK. NADPH oxidases: functions and pathologies in the vasculature. Arterioscler Thromb Vasc Biol. 2010;30:653-661.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 440]  [Cited by in RCA: 462]  [Article Influence: 28.9]  [Reference Citation Analysis (0)]
25.  Infanger DW, Sharma RV, Davisson RL. NADPH oxidases of the brain: distribution, regulation, and function. Antioxid Redox Signal. 2006;8:1583-1596.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 290]  [Cited by in RCA: 292]  [Article Influence: 15.4]  [Reference Citation Analysis (0)]
26.  Kim F, Pham M, Luttrell I, Bannerman DD, Tupper J, Thaler J, Hawn TR, Raines EW, Schwartz MW. Toll-like receptor-4 mediates vascular inflammation and insulin resistance in diet-induced obesity. Circ Res. 2007;100:1589-1596.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 407]  [Cited by in RCA: 426]  [Article Influence: 23.7]  [Reference Citation Analysis (1)]
27.  Kitagishi Y, Matsuda S. Diets involved in PPAR and PI3K/AKT/PTEN pathway may contribute to neuroprotection in a traumatic brain injury. Alzheimers Res Ther. 2013;5:42.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 31]  [Cited by in RCA: 40]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
28.  Marizzoni M, Cattaneo A, Mirabelli P, Festari C, Lopizzo N, Nicolosi V, Mombelli E, Mazzelli M, Luongo D, Naviglio D, Coppola L, Salvatore M, Frisoni GB. Short-Chain Fatty Acids and Lipopolysaccharide as Mediators Between Gut Dysbiosis and Amyloid Pathology in Alzheimer's Disease. J Alzheimers Dis. 2020;78:683-697.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 94]  [Cited by in RCA: 214]  [Article Influence: 53.5]  [Reference Citation Analysis (0)]
29.  Yang T, Zubcevic J. Gut-Brain Axis in Regulation of Blood Pressure. Front Physiol. 2017;8:845.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 40]  [Cited by in RCA: 54]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
30.  Li R, Wang J, Xiong W, Luo Y, Feng H, Zhou H, Peng Y, He Y, Ye Q. The oral-brain axis: can periodontal pathogens trigger the onset and progression of Alzheimer's disease? Front Microbiol. 2024;15:1358179.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 10]  [Reference Citation Analysis (0)]
31.  Heppner FL, Ransohoff RM, Becher B. Immune attack: the role of inflammation in Alzheimer disease. Nat Rev Neurosci. 2015;16:358-372.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1284]  [Cited by in RCA: 1584]  [Article Influence: 158.4]  [Reference Citation Analysis (0)]
32.  Hettiarachchi SD, Kwon YM, Omidi Y, Speth RC. Nanoparticle approaches for the renin-angiotensin system. Heliyon. 2023;9:e16951.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 6]  [Reference Citation Analysis (0)]
33.  Martín Giménez VM, Lahore H, Ferder L, Holick MF, Manucha W. The little-explored therapeutic potential of nanoformulations of 1,25-dihydroxyvitamin D(3) and its active analogs in prevalent inflammatory and oxidative disorders. Nanomedicine (Lond). 2021;16:2327-2330.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 3]  [Reference Citation Analysis (0)]
34.  Scuto M, Majzúnová M, Torcitto G, Antonuzzo S, Rampulla F, Di Fatta E, Trovato Salinaro A. Functional Food Nutrients, Redox Resilience Signaling and Neurosteroids for Brain Health. Int J Mol Sci. 2024;25:12155.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 12]  [Reference Citation Analysis (0)]
35.  Martín Giménez VM, Moretton MA, Chiappetta DA, Salgueiro MJ, Fornés MW, Manucha W. Polymeric Nanomicelles Loaded with Anandamide and Their Renal Effects as a Therapeutic Alternative for Hypertension Treatment by Passive Targeting. Pharmaceutics. 2023;15:176.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
36.  Jeon SG, Cha MY, Kim JI, Hwang TW, Kim KA, Kim TH, Song KC, Kim JJ, Moon M. Vitamin D-binding protein-loaded PLGA nanoparticles suppress Alzheimer's disease-related pathology in 5XFAD mice. Nanomedicine. 2019;17:297-307.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 45]  [Cited by in RCA: 70]  [Article Influence: 11.7]  [Reference Citation Analysis (0)]
37.  Abdelazim K, Ghit A, Assal D, Dorra N, Noby N, Khattab SN, El Feky SE, Hussein A. Production and therapeutic use of astaxanthin in the nanotechnology era. Pharmacol Rep. 2023;75:771-790.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 18]  [Cited by in RCA: 16]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
38.  Akanchise T, Angelova A. Ginkgo Biloba and Long COVID: In Vivo and In Vitro Models for the Evaluation of Nanotherapeutic Efficacy. Pharmaceutics. 2023;15:1562.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 17]  [Reference Citation Analysis (0)]
39.  Li M, Shang X, Lou H, Wang Z, Xiang S, Qiu Y, Hu F, Yu F, Yuan H. Active Anchoring Stimuli-Responsive Nano-Craft to Relieve Pulmonary Vasoconstriction by Targeting Smooth Muscle Cell for Hypoxic Pulmonary Hypertension Treatment. Adv Healthc Mater. 2024;13:e2400113.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 3]  [Reference Citation Analysis (0)]
40.  Yao M, Wu M, Yuan M, Wu M, Shen A, Chen Y, Lian D, Liu X, Peng J. Enhancing the therapeutic potential of isoliensinine for hypertension through PEG-PLGA nanoparticle delivery: A comprehensive in vivo and in vitro study. Biomed Pharmacother. 2024;174:116541.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 5]  [Cited by in RCA: 6]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
41.  Holick MF, Mazzei L, García Menéndez S, Martín Giménez VM, Al Anouti F, Manucha W. Genomic or Non-Genomic? A Question about the Pleiotropic Roles of Vitamin D in Inflammatory-Based Diseases. Nutrients. 2023;15:767.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 21]  [Reference Citation Analysis (0)]
42.  Bukhari SA, Sadia H, Chauhdary Z. The neuroprotective potential of Gerbera Jamesonii in a neuronal demyelination rat model through the modulation of interleukins, cyclooxygenase and tumor necrosis factor-α. Inflammopharmacology.  2025.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
43.  Okoh OS, Akintunde JK, Akamo AJ, Akpan U. Thymoquinone inhibits Neuroinflammatory mediators and vasoconstriction injury via NF-κB dependent NeuN/GFAP/Ki-67 in hypertensive Dams and F1 male pups on exposure to a mixture of Bisphenol-A analogues. Toxicol Appl Pharmacol. 2025;494:117162.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
44.  Prado NJ, Ramirez D, Mazzei L, Parra M, Casarotto M, Calvo JP, Cuello Carrión D, Ponce Zumino AZ, Diez ER, Camargo A, Manucha W. Anti-inflammatory, antioxidant, antihypertensive, and antiarrhythmic effect of indole-3-carbinol, a phytochemical derived from cruciferous vegetables. Heliyon. 2022;8:e08989.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 2]  [Cited by in RCA: 16]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]