Svoboda | Graniru | BBC Russia | Golosameriki | Facebook
Next Article in Journal
Daphnetin Ameliorates Neuropathic Pain via Regulation of Microglial Responses and Glycerophospholipid Metabolism in the Spinal Cord
Previous Article in Journal
In Vitro/In Vivo Correlation of Two Extended-Release Cilostazol Formulations
Previous Article in Special Issue
Enzymatic Metabolic Switches of Astrocyte Response to Lipotoxicity as Potential Therapeutic Targets for Nervous System Diseases
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Promising Strategies to Reduce the SARS-CoV-2 Amyloid Deposition in the Brain and Prevent COVID-19-Exacerbated Dementia and Alzheimer’s Disease

by
Nikita Navolokin
1,2,
Viktoria Adushkina
2,
Daria Zlatogorskaya
2,
Valeria Telnova
2,
Arina Evsiukova
2,
Elena Vodovozova
3,
Anna Eroshova
4,
Elina Dosadina
4,
Sergey Diduk
4,5 and
Oxana Semyachkina-Glushkovskaya
2,*
1
Department of Pathological Anatomy, Saratov Medical State University, Bolshaya Kazachaya Str. 112, 410012 Saratov, Russia
2
Department of Biology, Saratov State University, Astrakhanskaya 82, 410012 Saratov, Russia
3
Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya 16/10, 117997 Moscow, Russia
4
Department of Biotechnology, Leeners LLC, Nagornyi Proezd 3a, 117105 Moscow, Russia
5
Research Institute of Carcinogenesis of the N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia, Kashirskoe Shosse 24, 115522 Moscow, Russia
*
Author to whom correspondence should be addressed.
Pharmaceuticals 2024, 17(6), 788; https://doi.org/10.3390/ph17060788
Submission received: 29 April 2024 / Revised: 2 June 2024 / Accepted: 11 June 2024 / Published: 16 June 2024
(This article belongs to the Special Issue Multi-target Drug Treatments for Neurodegenerative Disease)

Abstract

:
The COVID-19 pandemic, caused by infection with the SARS-CoV-2 virus, is associated with cognitive impairment and Alzheimer’s disease (AD) progression. Once it enters the brain, the SARS-CoV-2 virus stimulates accumulation of amyloids in the brain that are highly toxic to neural cells. These amyloids may trigger neurological symptoms in COVID-19. The meningeal lymphatic vessels (MLVs) play an important role in removal of toxins and mediate viral drainage from the brain. MLVs are considered a promising target to prevent COVID-19-exacerbated dementia. However, there are limited methods for augmentation of MLV function. This review highlights new discoveries in the field of COVID-19-mediated amyloid accumulation in the brain associated with the neurological symptoms and the development of promising strategies to stimulate clearance of amyloids from the brain through lymphatic and other pathways. These strategies are based on innovative methods of treating brain dysfunction induced by COVID-19 infection, including the use of photobiomodulation, plasmalogens, and medicinal herbs, which offer hope for addressing the challenges posed by the SARS-CoV-2 virus.

Graphical Abstract

1. Introduction

Alzheimer’s disease (AD) is a brain disorder that is accompanied by a gradual decline in memory, thinking and behavior skills. According to world statistics, the global number of people over 65 years old with AD doubles every 5 years [1]. As estimated, the incidence of AD will triple by 2060 unless the effective therapy is found to prevent or slow AD [1,2]. AD is the sixth leading cause of death in the United States [2]. During the period of 2020–2021, the number of deaths from stroke, cardiovascular disease, and human immunodeficiency virus decreased, whereas the number of people who died from AD increased more than 145% [2]. There is growing evidence that such a significant increase in mortality from AD may be associated with the COVID-19 pandemic in 2020 and 2021 [2,3,4,5,6,7,8,9,10,11,12,13,14,15,16,17]. COVID-19 is primarily known as a severe acute respiratory disease caused by the SARS-CoV-2 virus, but it also affects the central nervous system (CNS) and other organs [18,19,20,21,22,23]. Many neurological symptoms, including memory loss, cognitive impairment, fatigue, brain fog, and insomnia are reported in up to 30% of cases and may appear long after the infection (long COVID) [18]. However, despite considerable evidence that there is a relationship between COVID-19 and AD, it remains unclear if the SARS-CoV-2 virus itself causes AD and/or if the SARS-CoV-2 infection exacerbates existing AD. The virus hypothesis for AD has been proposed for decades, and despite supportive evidence, virus etiology of AD is still controversial [7,22,23,24,25,26,27,28]. On the one hand, many clinical observations indicate an increased risk for COVID-19 in people with AD [2,3,4,5,6,7,8,9,10,11,12,13,14,15,16,17]. On the other hand, SARS-CoV-2 infection causes long-lasting neurological sequelae [18,19,20,21,22,23]. The key question, whether COVID-19 can trigger new-onset AD or only accelerate AD progression, is unclear.
It was recently discovered that the SARS-CoV-2 virus proteome includes different sequences of amyloid-forming proteins [4,29,30]. It is assumed that amyloid-forming proteins from the SARS-CoV-2 virus may be involved in the development of neurological symptoms in COVID-19-infected patients [3,4]. The SARS-CoV-2 amyloid could have cytotoxic and neuroinflammatory effects similar to those of amyloids that are responsible for amyloid-related neurodegenerative diseases, including AD. Therefore, COVID-19-exacerbated AD could be a SARS-CoV-2 amyloid disorder. However, there is not enough evidence for this yet. Nevertheless, it has been noted that COVID-19 triggers systemic AA amyloidosis and upregulation of serum amyloid A protein [31]. On the other hand, amyloid beta (Aβ) has antiviral and antibacterial effects [32,33,34,35]. To this end, Aβ may be activated in the COVID-19-infected brain as a protective mechanism that can explain the proposed viral involvement in Aβ aggregation in the brain. However, despite the lack of a clear understanding of whether COVID-19 causes pathological amyloid accumulation in the brain or whether it aggravates already ongoing neurodegenerative processes, the indisputable fact is that the COVID-19 infection is accompanied by amyloid aggravation in the brain associated with the neurological disorders, including symptoms of AD and dementia.
Medicines may improve or slow the progression of AD [36,37,38]. However, there is no promising treatment that cures AD and reduces the accumulation of amyloids in the brain. Clinical investigations have failed to show any effective pharmacological therapy for AD [39,40,41,42]. The latest clinical trials of aducanumab on 3285 patients with AD revealed significant consequences associated with immunotherapy, such as edema, microhemorrhage, nausea dizziness, headache, and confusion [42]. However, Aβ immunotherapy can be improved by modulation of functions of the meningeal lymphatic vessels (MLVs) [43]. MLVs are now thought to be the major route for clearance of amyloids in AD [44,45,46,47,48,49]. Therefore, strategies for augmentation of lymphatic removal of amyloids from the brain can lay the foundation for the development of promising therapeutic approaches for AD [44]. MLV-based therapeutic strategies may be also useful for alleviating infection-induced neurological damage due to facilitation of virus clearance from the brain to the cervical lymph nodes [50].
However, despite the emerging evidence suggesting that MLVs may serve as an efflux route for Aβ from the human and animal brain, confocal analysis of sinus-associated MLVs in patients who died from AD has not found Aβ deposits directly inside of MLVs [51]. This suggests the need to develop alternative strategies beyond MLV modulation to stimulate amyloid clearance from brain tissue. Approaches using natural compounds as inhibitors of Aβ aggregation in brain seem to be most promising [52,53].
The focus of this review is to highlight new discoveries in the field of COVID-19-mediated amyloid accumulation in the brain associated with the neurological symptoms and the development of promising strategies to stimulate the clearance of amyloids from the brain through lymphatic and other pathways.

2. SARS-CoV-2 Amyloids and COVID-19-Mediated AD Dementia

Many proteins of viruses and bacteria have amyloid properties, which are thought to have an important role in the progression of infectious diseases [54,55,56,57]. The SARS-CoV-2 virus has four main structural proteins, namely the spike, envelope, membrane, and nucleocapsid proteins, as well as non-structural accessory proteins, including ORF6 and ORF10 [58,59] (Figure 1). The SARS-CoV-2 amyloids have been identified in the structural spike [60], the nucleocapsid [30], and the accessory ORF6/ORF10 proteins [4,61]. When the SARS-CoV-2 virus enters an infected cell, it creates many copies that are accompanied by an exponential increase in intracellular SARS-CoV-2 amyloids [62]. The spike proteins of the SARS-CoV-2 virus also bind to Aβ causing Aβ deposition in the brain [63,64,65] (Figure 1). The ORF10 protein causes mitochondrial dysfunction [66] mimicking AD-mediated metabolic disorders [4]. SARS-CoV-2 amyloids can be also released from infected cells into extracellular spaces, forming amyloid depositions leading to neurotoxicity and the pathophysiology of infection [4,30,60,62].
If the SARS-CoV-2 virus contains amyloid-forming sequences in its genome, the question arises, what is their role? It is known that every component of the virus helps it to impede the host immune system and replicate. Therefore, there are several possible roles of SARS-CoV-2 amyloids. The simplest explanation is SARS-CoV-2 amyloids can be an inflammatory stimulus [67] leading to the high expression of the angiotensin-converting enzyme 2 (ACE-2) receptors on the surface of microglia, through which the virus enters the cell, and that increases intracellular transmissibility of the SARS-CoV-2 virus. The nucleocapsid protein of the SARS-CoV-2 virus contains a lot of amyloidogenic sequences that may play an important role in RNA packing during virus replication [68,69,70]. It is also possible that SARS-CoV-2 amyloids might inhibit the host’s antiviral response, similar to other amyloids in other viruses [71].
Recent studies have reported that COVID-19 is associated with structural changes in the brain [72,73,74,75,76]. Therefore, severe COVID-19 patients demonstrate a decrease in grey matter volume of the frontal lobe [77,78,79]. The grey matter in the CNS allows for the control of movement, memory, and emotions [80,81]. Memory deficits have been found in 19.2% of post-COVID patients [78]. The SARS-CoV-2 virus can enter the hippocampus through invasion of the olfactory neurons [82,83]. Since the hippocampus plays a crucial role in regulation of episodic and spatial memory [84,85], hippocampal infection may lead to memory disorders in COVID-19 patients. Many forms of dementia are associated with deposition of different amyloids in the brain, raising the possibility that SARS-CoV-2 amyloids might be involved in the development of COVID-19-mediated dementia and AD or contribute to their progression through an increase of amyloid toxicity and aggregation [4,29,30,63,86,87,88,89,90,91,92].
There is also indirect mechanism of COVID-19-related AD dementia. COVID-19 as a respiratory diseases causes hypoxic changes to which the brain is highly sensitive [93,94,95,96]. It is noted that dementia and Aβ depositions are often accompanied by hypoxia of brain tissues [97,98,99,100,101,102,103]. This means that COVID-19-induced hypoxia might be an additional and independent factor promoting the development of dementia.
In sum, it has been discovered that SARS-CoV-2 amyloids play a potential role in COVID-19-mediated AD dementia [3,4,29,30,60,62,63,64,65,66] (Figure 1). By causing respiratory infection and systemic inflammation, the SARS-CoV-2 virus causes the development of a cytokine storm leading to blood–brain barrier (BBB) disruption [104,105,106,107,108,109]. We discussed the mechanism of COVID-19-related BBB damages in detail in our recent review [104]. The SARS-CoV-2 virus can enter the different regions of the brain through the damaged BBB as well as penetrating into the hippocampus through invasion of the olfactory neurons [82,83,84,85,110,111]. In the brain, the SARS-CoV-2 virus stimulates production of amyloid deposits in both extracellular and intracellular spaces [3,4,29,30,60,62,63,64,65,66]. The toxicity of SARS-CoV-2 amyloids can directly induce brain changes and lead to the development or the progression of AD dementia [4,72,73,74,75,76,78,82,83,84,85,112,113]. Thus, SARS-CoV-2 amyloids can play a causative role in COVID-19-exacerbated dementia and may be the targets for therapeutic approaches in prevention of COVID-19-mediated memory disorders.

3. Meningeal Lymphatics as a Target for Anti-Amyloid Therapy

Recent studies have demonstrated an important role of MLVs in the protection of the brain against pathogens and the development of neurodegenerative diseases [114,115,116,117]. Dysfunction of MLVs aggravates Aβ accumulation in the brain and significantly reduces the efficacy of immunotherapy against AD and brain tumors [43,44,118,119]. Several viruses, such as SARS-CoV-2, herpes simplex virus 1, rabies virus, Zika virus, vesicular stomatitis virus, and Japanese encephalitis virus, can invade the brain and induce inflammation with meningitis and neurologic symptoms [120,121,122,123,124,125]. It is known that the brain is protected against viruses, bacteria, and toxins by the BBB. However, viruses can enter the brain through the olfactory and trigeminal nerves pathways, bypassing the BBB [82,83,123,126,127]. It is important to note that MLVs can transport viruses from the brain to the peripheral lymphatics [114]. Li et al. revealed that that viral infection in mice reduces brain drainage and lymphatic removal of macromolecules from the brain. Photo or surgical damages to MLVs increase neurological symptoms and mortality in virus-infected mice. However, vascular endothelial growth factor C (VEGF-C) pretreatment promotes recovery of MLV functions against viral infection. These data indicate that MLVs facilitate lymphatic clearance of viruses, suggesting a promising MLV-based therapeutic approach against neurotropic viral infection [114]. Since MLVs play an important role in eliminating toxins and wastes from the brain, it is assumed that augmentation of MLV function could be a promising therapeutic target for preventing or delaying age-associated neurocognitive diseases, including COVID-19-mediated AD dementia [43,44,45,46,47,48,49,128,129].
Thus, the development of effective methods for stimulation of MLV functions is highly relevant for medicine. Recently, non-invasive transcranial photobiomodulation (PBM) has been proposed as a novel alternative method for therapy for dementia and AD as well as a promising approach in the improvement of cognitive functions in healthy subjects [128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155,156,157]. It is believed that the therapeutic effects of PBM are based on reducing neuroinflammation and activation of anti-oxidant systems through the improvement of metabolism and microcirculation of the brain, leading to neuroprotection [133,138,139,141]. However, emerging evidence suggests that PBM can also stimulate MLV functions, providing lymphatic removal of wastes and metabolites, including Aβ, from the brain, leading to reduced AD progression [151,152,153,154,155,156,157]. Notably, the U.S. Food and Drug Administration (FDA) recognized PBM as safe. The safe use of PBM is regulated by relevant standards, e.g., ANSI Z136 [158,159].
The mechanisms of PBM-mediated improvement of cognitive function in healthy subjects and with AD are not clear, but it is most likely that it may be due to PBM stimulation of MLV function. Indeed, disruption of MLVs aggravates AD, leading to cognitive deficit and behavioral alterations [44,47,48,49,50,160,161]. Thus, MLV dysfunction might cause cognitive decline and neurodegenerative disease due to aggregation of toxins in the brain, while PBM-stimulation of their lymphatic removal can improve the brain homeostasis, immunity and neuroprotection [43,44,46,47,48,49,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155,156,157].
There is convincing evidence that PBM can modulate lymphatic contractility and pumping, improving drainage and removal of macromolecules from tissues (Figure 2a) [153,154,162]. The PBM-mediated increase in nitric oxide (NO) production could be one possible mechanism responsible for PBM-stimulation of the MLV functions. Indeed, PBM significantly increases NO generation in culture of isolated lymphatic endothelial cells [162]. The stimulating effects of PBM on MLVs are suppressed by the blockade of NO production [162]. How may NO promote contraction of lymphatic vessels? Traditionally, it is believed that NO suppresses lymphatic constriction. It is generally believed that NO causes relaxation of both blood and lymphatic vessels, suppressing their contractility. However, NO-related regulation of the lymphatic endothelium is much more complex than previously thought. It is known that the influx of fluid into the lymph vessel stimulates its contraction and NO production in the lymphatic endothelial cells increases in accordance with the intensity of vessel contractility [163]. Lymphatic constriction is accompanied by an elevated NO production in the lymphatic walls through an increased flow shear forces creating by coming fluids. In this case, NO causes dilation of the lymphatic endothelium after constriction, i.e., inhibits lymphatic pumping during high lymph flow. However, NO’s effects on different parts of the lymphatic vasculature are different. The lymphatic valves contain 50% more endothelial NO-synthase than tubular regions predominantly causing high NO production during the contraction of lymphatic vessels [163]. From a physiological perspective, during lymphatic constriction NO releases from the lymphatic valves into the lumen of the vessel and reaches tubular sites contributing their relaxation that is important for regulation of peristaltic pumping [163,164,165].
The contraction phase of the lymphatic vessels depends on the opening of the Ca2+-channels, while the relaxation cycle is a component of local production of NO in the lymphatic endothelial cells that triggers by transiently elevated shear forces (Figure 2b). Ca2+ and NO cooperate in their regulation of lymph flow via mechanobiological mechanisms. During the contraction cycle, shear stress is increased and triggers activation of the endothelial NO synthase and NO production in the lymphatic valves. In this case, the elevated production of NO reduces intracellular Ca2+ concentration, leading to preparation of the lymphatic endothelial cells for subsequent relaxation. During the relaxation phase, due to the reduced fluid velocity in the now dilated vessel, NO production drops, which is accompanied by its rapid degradation. At this time, intracellular Ca2+ levels and membrane potentials are restored in preparation for another contraction of the vessels, which can be triggered by any signal able to initiate the opening of Ca2+ channels or Ca2+ flux from calmodulin (calcium-binding messenger protein) or through the gap junctions of neighboring cells (Figure 2b).
We hypothesize that PBM can stimulate brain drainage and lymphatic clearance of macromolecules, including amyloids and viruses, from the brain due to PBM-mediated regulation of combination of mechanical and electrophysiological events associated with the pumping of the lymphatic vessels via PBM-mediated stimulation of NO production in the lymphatic endothelial cells (Figure 2c).

4. Plasmalogens as a Potential Therapy for AD

Lipids play a crucial role in the structure and functioning of the brain. The brain contains the second highest amount of lipids after adipose tissue. However, the type of lipids found in the brain is different from that in adipose tissue, with phospholipids being the main component in the brain. The brain contains a significant amount of mono- and polyunsaturated fatty acids such as oleic acid, arachidonic acid, and docosahexaenoic acid, which are mainly stored in phospholipids. These phospholipids are essential components of cell membranes and play a critical role in their proper functioning [166,167,168,169]. Cholesterol and phospholipids containing saturated fatty acids are concentrated in lipid rafts present in cell membranes. These rafts are involved in cell signaling. Membrane fluidity, supported by phospholipids consisting of unsaturated fatty acids, is crucial for membrane-associated functions. It includes the processing of amyloid precursor protein and plays a vital role in the pathogenesis of AD. The composition of phospholipids in cell membranes is involved in important neural functions, such as vesicular fusion and neurotransmitter release, which can be altered in AD [170,171,172,173].
Among phospholipids, plasmalogens play an enormous role in maintaining brain homeostasis. They appear to be the missing link between the biochemical and functional abnormalities observed in AD and its pathological signs, such as the accumulation of Aβ in the brain [174,175,176,177].
Plasmalogens are glycerophospholipids that are vital for brain function [178,179]. They are the primary structural components of lipoproteins, myelin, synaptic membranes, and cell membranes. Plasmalogens possess unique physicochemical characteristics that help regulate membrane fluidity, lipid packaging into lipoproteins, and interaction with nerve receptors and ion channels. These lipids are also necessary for synaptogenesis, myelination, and ion transport [180].
Plasmalogens are synthesized in the peroxisomes and endoplasmic reticulum. The initial reaction involves the acylation of dihydroxyacetone phosphate by acyltransferase. Cells tightly regulate plasmalogen levels. Excessive levels of plasmalogens act as feedback inhibitors, reducing the levels of fatty acyl-CoA reductase 1. This reduction occurs by promoting the degradation of protein, which provides fatty alcohols in the synthesis [179,181]. Peroxisome biogenesis disorders affect the metabolism of lipids and reactive oxygen species, leading to symptoms such as hypomyelination due to low plasmalogen levels and increased inflammation. Cells with a deficiency in plasmalogen synthesis have low oxidation resistance, but adding alkylglycerol as a plasmalogen precursor can increase plasmalogen levels and enhance oxidative resistance [176].
The emergence of age-related diseases such as metabolic syndrome, type 2 diabetes mellitus, Parkinson’s disease, and AD are associated with oxidative stress and chronic neuroinflammation. The disruption of redox homeostasis leads to peroxisome malfunction and impaired plasmalogen production, which could be a mechanism of the development of age-related diseases [174,182,183].
The level of plasmalogens in the brain increases until 30–40 years and then decreases sharply at about 70 years [184,185,186]. This age range coincides with the period of life when the incidence of AD increases exponentially [187,188,189]. Given the sharp decrease in brain plasmalogen levels with age and their critical role in maintaining brain homeostasis, it is not surprising that deficiency of plasmalogens in the brain is closely associated with the progression of aging-related neurodegenerative disorders such as AD and Parkinson’s disease [174,175,190].
Oxidative cleavage of the vinyl ether bond by cytochrome C in the presence of H2O2 [191,192] is a potential mechanism of degradation of plasmalogens. A decrease in peroxisomal function combined with higher H2O2 levels potentially causes a permanent plasmalogen deficiency, which leads to membrane changes, signaling abnormalities, reduced neurotransmission, and suppressed antioxidant protection [193]. Oxidative stress associated with inflammation potentially exacerbates the degradation of plasmalogens by attacking the vinyl–ether bond, further reducing anti-inflammatory and antioxidant capacity of tissues and initiating an irreversible vicious circle that progresses to pathological abnormalities [175].
During COVID-19 infection, the spike viral protein interacts with ACE2, which leads to the excessive production of angiotensin II and the activation of the nicotinamide adenine dinucleotide phosphate oxidase. This subsequently leads to an increase in oxidative stress and release of inflammatory molecules [194]. Unlike other viral infections, COVID-19 infection severity is usually associated with age and may be linked to an imbalance in the redox system. This can cause an accumulation of oxidative damage and a decline in the antioxidant defense system, resulting in an increase in reactive oxygen species [195].
Thus, plasmalogens, which reflect peroxisomes’ functional activity, can be used as biomarkers for diseases related to oxidative stress and aging, as well as a critical therapeutic target [182,196]. A study conducted by Yamashita et al. revealed that administration of Ascidian Viscera glycerophospholipid (EtnGpl), a plasmalogen-rich ethanolamine, to rats with an injected model of AD improves their learning ability and working memory through reducing oxidative stress in the brain [197]. Chronic injection of lipopolysaccharides in mice can lead to the activation of glial cells and accumulation of Aβ proteins in the brain that are associated with cognitive deficit. Hossain et al. clearly show that plasmalogens prevent neuronal cell death by activating protein kinase B and inhibiting caspase 3, improving neuronal survival [198,199]. Wood et al. found that the serum level of plasmalogens is significantly reduced in patients with AD and correlates with severity of dementia [200]. These data indicate that the circulating level of plasmalogens may be an indicator of the progression of AD.
There is growing evidence suggesting improvement of cognitive deficits in both animals and humans with neurodegenerative diseases. Thus, scallop- and ascidian-derived plasmalogens can reduce the incidence of cognitive impairment in subjects with mild forgetfulness, AD and Parkinson’s disease due to an increase in the plasma ethanolamine plasmalogen (PlsEtn) levels [201,202,203,204]. Chicken-derived plasmalogens have similar effects in healthy people with mild memory problems [205]. Phosphatidylserine plasmalogen species attenuate cognitive disorders after cerebrovascular injury [206].
The advantages of marine plasmalogens from ascidians, scallops, and sea cucumbers and the mechanisms underlying their beneficial effects on memory function are not well understood due to differences approaches in experiments, including dose, duration, and ethnicity of volunteers.
In aged mice, marine plasmalogens ameliorate memory deficit via activation of neurogenesis [53]. Amyloid-mediated neuroinflammation is associated with a decrease in the brain PlsEtn levels leading to suppression of the expression of the brain-derived neurotrophic factor (BDNF) and reduced neurogenesis [207,208]. These changes promote the high expression of p75 neurotrophin receptor (p75NTR) and protein kinase Cδ (PKCδ) leading to neural death and neurite degeneration [207,209]. However, the administration of PlsEtn reduces Aβ depositions in the brain and tau hyperphosphorylation by attenuation of the p75NTR and PKCδ expression, which leads to reducing neuroinflammation [207,209,210].
Chicken plasmalogens also enhance memory by attenuation of neuroinflammation [208,211]. However, marine PlsEtn enhances memory function better than chicken-derived plasmalogens, probably because marine PlsEtn is richer in DHA than the chicken derivative [208,210].
It is noted that brain levels of PlsEtn containing DHA in the cortex correlate with cognitive abilities of rats with AD [197,212,213]. In vitro experiments have shown that PlsEtn bearing DHA exhibits strong suppression of neuronal inflammation, apoptosis, γ-secretase activity, and Aβ aggregation [212,213,214]. Interestingly, intravenously injected small-sized liposomes can increase the bioavailability of PlsEtn with DHA, enhancing their beneficial effects on locomotor activity in normal rats [215,216].
Systemic administration of plasmalogens over 7 days reduces microglia inflammation induced by Aβ accumulation [217]. Longer treatment with scallop-derived plasmalogens over 15 months decreases both the expression of markers of inflammation and the protein kinase C-δ (PKCδ) involved in apoptosis [209]. Rothhaar et al. reported that murine plasmalogens reduce gamma-secretase activity in cell membranes isolated from post-mortem brain AD tissue [218].
However, murine plasmalogens can be beneficial for AD and improvement of cognitive function due to an increase in the brain PlsEtn level and neurogenesis, leading to reducing amyloid accumulation, tau hyperpolarization attenuating neuroinflammation and neuron death (Figure 3).

5. Natural Compounds as a Novel Treatment for AD

Current pharmacological therapy for AD includes use of cholinesterase inhibitors, anti-Aβ vaccine, and anti-neuroinflammation drugs [219,220,221]. Rivastigmine, galantamine, memantine, and donepezil are approved for treatment of AD [219,220,221]. However, these medications only partly improve quality of people with AD [222,223]. Monoclonal antibody anti-Aβ therapy, including aducanumab, lecanemab, and GV971, is a new strategy in therapy for AD in recent years. However, the safety and effectiveness of these drugs are still controversial, and these drugs are very expensive [224,225,226,227]. There are 43 new clinical drug trials registered in the National Library of Medicine database (ClinicalTrials.gov: accessed on 28 April 2024). However, the most common outcome was a lack of efficacy or only control of early symptoms of AD.
Thus, no effective cure for AD currently exists, and available pharmacological strategies have shown limited effectiveness. Therefore, the search for new approaches to AD therapy is crucial to address the growing burden of this pathology. Medicinal herbs are a promising avenue for the treatment of AD [228,229,230,231,232,233]. Natural compounds were the first therapy used for AD [233]. Although the mechanisms of therapeutic effects of ayurvedic medicinal herbs remain largely unknown, extensive experimental and clinical studies on phytochemical actions have found a wide range of beneficial effects for AD within these plants [228,229,230,231,232,233]. There are several natural components exhibiting therapeutic effectiveness for AD, including flavonoids, polyphenols, sterols, alkaloids, triterpenes, lignans, and tannins with effects against to oxidative stress, neuroinflammation, amyloid deposition, and activity of cholinesterase.
Here, we highlight the applications of various natural compounds as a novel treatment for AD (Figure 4).

5.1. Curcuma longa and Piper nigrum

Turmeric is the root of Curcuma longa, and its main active ingredients are curcuminoids. The most important and most abundant of these curcuminoids is curcumin [234,235]. Curcumin has been approved by the FDA as a natural dietary supplement due to its safety and non-toxicity. Scientists’ interest in curcumin increased due to its unique molecular structure, which has multiple effects, including anti-inflammatory, antioxidant, and neuroprotective actions [236]. It also has the ability to disaggregate tau proteins [237] and target amyloid plaques, which are one of the main causes of AD [238]. Studies have shown that among people aged 70–79, there is a 4.4-fold lower incidence of AD in India compared to the USA. This may be due to the fact that people in India consume more curry, which contains curcumin, and perform better on cognitive function tests [239].
Piperine is an alkaloid found in black pepper (Piper nigrum) and long pepper (Piper longum) [239]. It has been found that piperine can increase the absorption of various drugs [240], and it has also been observed that when curcumin is taken with piperine, the absorption of curcumin increases by 154% in rats and 2000% in humans [241,242]. It has been shown to improve the solubility, stability, and absorption of curcumin. However, curcumin has a poor oral bioavailability. To improve oral curcumin delivery to the brain, self-nanoemulsifying drug delivery systems (S-SNEDDS) have been developed using a nanotechnological approach [243]. In this study, the authors clearly demonstrate a significant dose-dependent therapeutic effect of combined curcumin with S-SNEDDS than drug alone in an AD model.

5.2. Ginkgo biloba

Ginkgo biloba is commonly used in the treatment of AD cognitive impairment due to its antioxidant and anti-apoptotic properties [244,245]. Its leaves contain various essential components, such as flavonoids, steroids, organic acids, ginkgolides, and terpenoids. These compounds, including bilobalide and ginkgolide, are crucial for its therapeutic effects [246,247]. Flavone glycosides make up approximately 22–27% of the extract, while terpene lactones comprise about 5–7% [246]. Specific terpene lactone compounds include A, B, and C ginkgolides and bilobalide.
Ginkgo biloba contains varies flavone glycosides, such as quercetin, isorhamnetin and kaempferol, that play a crucial role in preventing oxidative stress in AD [246]. The herb is thought to work through several mechanisms, including preventing apoptosis, reducing oxidative stress, inhibiting the formation of amyloid plaques, and scavenging free radicals. These actions help to prevent and treat AD. The plant extract also inhibits the neurotoxic effects of Aβ by regulating the activity of glutathione peroxidase and superoxide dismutase. This helps to limit neuronal apoptosis, reduce the build-up of reactive oxygen species, and improve glucose uptake and mitochondrial function. It also prevents the activation of the extracellular signal-regulated kinase and c-Jun N-terminal kinase signaling pathways. There is convincing evidence that Ginkgo biloba can enhance cognitive function in patients with AD by improving oxygen supply to brain tissues and reducing the brain level of free radicals [246,247].

5.3. Bacopa monnieri

Bacopa monnieri is a well-respected nootropic herb that has been used for centuries to treat neurological conditions. A chemical analysis of extract from Bacopa monnieri has revealed the presence of various bioactive compounds, including triterpenoids, alkaloids (such as nicotine, brahmine, and herpestine), saponins, glycosides, D-mannitol, herpaponin, monnierin, and alcohols. This plant also contains a variety of phytocompounds, such as bacosides A and B, bacosaponins A, B, C, bacosapeptides III to V, bacosapans D, E, F, jujubogenin, betulic acid, alkaloids, polyphenols, steroids, and sulfur compounds, all of which indicate its antioxidant properties. The antioxidant and neuroprotective effects of Bacopa monnieri make it a promising herb for AD. The main substances responsible for its neuroprotective effects are bacosides A and B due to their beneficial effects on the transmission of nerve signals and recovery of injured neurons [248]. The antioxidant activity of Bacopa monnieri also leads to an increase in various antioxidant molecules, including superoxide dismutase, thus reducing the harmful effects of H2O2-induced oxidative stress. In addition, this herb decreases lipoxygenase activity, assisting in the recovery from oxidative stress [249]. The diversity of bioactive compounds in Bacopa monnieri emphasizes its potential as a natural remedy for enhancing brain health and cognitive function.

5.4. Salvia officinalis

Salvia officinalis is a fragrant herb that is widely used in the treatment of AD [250,251]. Salvia contains more than 160 different polyphenolic compounds, including phenolic acids, and flavonoids. These include yunnaninic acid, lithospermum acids, salvianic acid, and others. It also contains flavonoids such as kaempferol, apigenin, and luteolin. In addition to these compounds, salvia is rich in terpenes such as alpha- and beta-thujones, 1,8-cineole, alpha-humulene, and camphor. These compounds can be found in the essential oils of the plant. Plants also contain significant quantities of diterpenes and triterpene acids, including tanshinones, carnosic acid, carnosol, and ursolic acid [249,250,251]. Salvia has great potential for promoting brain homeostasis and improving cognitive function. Indeed, Salvia officinalis has been shown to have anti-Aβ, anti-inflammatory and anxiolytic properties [249,250,251]. Salvia officinalis also demonstrates stimulating effects on the muscarinic and cholinergic pathways of the regulation of memory retention and formation, suggesting its supporting influences on cognitive properties.

5.5. Melissa officinalis

Melissa officinalis, also known as lemon balm, is a perennial herb with heart-shaped, bushy leaves that have a rough surface and grow upright [252,253]. The extracts from Melissa officinalis are rich in beneficial substances, including phenolic acids, flavonoids, and triterpenes. Thanks to these substances, the plant exhibits pronounced sedative, anti-depressant, and anti-inflammatory effects [254].
Studies have shown that lemon balm has the ability to inhibit acetylcholinesterase and exhibit antioxidant activity, making it valuable in preventing and treating AD [252,253,254,255,256,257]. In addition, lemon balm has anticholinesterase activity, binds to cholinergic receptors, and exhibits neuroprotective properties [256]. Some substances in lemon balm interact with muscarinic acetylcholine and nicotine receptors, leading to a decrease in acetylcholinesterase activity [256]. By modulating the cholinergic system, lemon balm is a promising treatment for AD, as it has been observed to reduce arousal and improve cognitive function in people with mild to moderate AD [252,253,254,255,256,257,258].

5.6. Huperzia serrata

Huperzia A, an essential alkaloid extracted from H. serrata, can effectively cross the BBB and acts as a reversible and selective acetylcholine esterase inhibitor demonstrating anti-AD effects [259,260].

5.7. Plants That Increase the Drainage Function of the Lymphatic System

MLVs play an important role in removal of wastes, metabolites, and toxins from the brain [44,45,46,47,48,49]. Therefore, MLVs are considered a promising target in therapy for brain diseases, including AD [43,44,45,46,47,48,49,128,129]. However, there are very limited pharmacological and non-pharmacological methods for augmentation of the MLV functions.
Recently, it has been proposed to introduce VEGF-C into the cisterna magna as a new method of pharmacological stimulation of MLV function [43,44,118,119]. However, this method has several limitations: (i) it is an invasive and traumatic method; (ii) VEFG-C has different side effects; and (iii) there are only few positive results in clinical studies in this area [261].
Herbal medicine has been proposed as alternative method in reducing lymphedema burden and drainage of different tissues [262,263]. Indeed, coumarin, juniper, black pepper, geranium, sage, and fennel are known to reduce swelling, and French oak wood extract containing robuvit leads to a sharp reduction in swelling [262]. Particular effectiveness of plant flavonoids has been shown in filarial lymphedema (edema of infectious origin) and in patients with breast cancer (edema of tumor origin) [264].
It is assumed that plant substances can enhance the drainage function of the lymphatic system via activation of the mechanisms lymphoneogenesis, including the high expression of VEFG proteins. Thus, Panax pseudoginseng increases mRNA levels of VEGF-C and vascular endothelial growth factor receptor-3 (VEGFR-3), leading to the stimulation and formation of new lymphatic vessels [265]. The stimulating effects on lymphangiogenesis have been found also for licoricidin, astragalus, and cinobufacini [263].
Plant substances can also be used to improve the drainage function of MLVs. Borneol (C10H18O, molecular weight 154.25) can quickly penetrate the BBB and enter the brain after oral administration within 5 min and reaches maximum concentration in the brain within 1 h [266,267], with the most efficient accumulation in the cerebral cortex and slightly less in the hippocampus and the hypothalamus [268,269]. Some studies demonstrate the effectiveness of borneol in therapy for AD, stroke, and epilepsy [270,271,272,273,274]. Indeed, Borneol improves removal of amyloids from the brain due to activation of lymphangiogenesis leading to improvement of cholinergic regulation of memory function.

6. Discussion

The recently discovered SARS-CoV-2 amyloids can play a crucial role in COVID-19-exacerbated dementia. Once the virus has penetrated the brain, it stimulates production of amyloid deposits at both extracellular and intracellular spaces. The toxicity of the SARS-CoV-2 amyloids can directly damage the brain, leading to the development or the progression of AD dementia. Therefore, SARS-CoV-2 amyloids may be targets for therapeutic approaches in prevention of COVID-19-mediated memory disorders and cognitive deficits. In this aspect, MLVs play a special role in the elimination of both amyloids and the SARS-CoV-2 virus from the brain [44,45,46,47,48,49,114]. It is actively being discussed whether increasing MLV functions will open a new page in the history of therapy for brain diseases associated with amyloidosis and lymphatic dysfunction, including AD, Parkinson’s disease, intracranial hemorrhages, brain trauma, and oncology [44,45,46,47,48,49,118,119,162,275,276]. However, this direction is in its infancy, which explains the limitation of technologies and pharmacological strategies for augmentation of MLVs to prevent or slow amyloidosis and MLV dysfunction. Among MLV stimulation technologies, the most promising is PBM because it is safe, without side effects, and approved by the FDA. There is emerging evidence suggesting that PBM effectively stimulates lymphatic removal of macromolecules, including Aβ and red blood cells from the brain as well as enhances brain’s drainage [153,154,155,156,157,162], leading to a significant improvement of memory function and cognitive properties in both healthy and AD subjects [128,157]. A pioneering direction is the development of portable technologies for PBM under the control of deep sleep. The animal and human data suggest that deep sleep is accompanied by strong activation of brain’s drainage and elimination of wastes and metabolites from the brain with the flow of brain fluids to the peripheral lymphatics [277,278]. It is obvious to assume that PBM of the MLV function and brain’s drainage will be most effective at the moment of their natural activation, i.e., during deep sleep [128,129]. Indeed, pilot results clearly show that PBM during deep sleep more effectively stimulates lymphatic removal of Aβ in an AD model and activates the brain’s drainage in both healthy mice and MLV-defective animals than PBM during wakefulness [45,128,129,151,157]. The use of PBM during sleep is also a promising direction in the treatment of brain tumors [279].
Among the pharmacological strategies for increasing the MLV functions, the direction of increasing lymphangiogenesis by introducing VEGF-C into the cisterna magna is widely discussed [43,118,119]. However, this method has limitations due to its invasiveness, side effects, and unproven clinical effectiveness. Therefore, the development of new alternative strategies to stimulate the MLV functions and remove macromolecules from the brain is a priority in medicine. Plasmalogens are a unique class of phospholipids playing an important role in membrane structure and in neural functions. Interestingly, the level of plasmalogens in the brain is dramatically reduced in AD subjects and is one of key mechanisms responsible for AD. The growing evidence demonstrates that plasmalogens can help to prevent some of AD progression through suppression of amyloid and tau accumulation, reducing neuroinflammation and activation of neurogenesis leading to significant improvement of cognitive and memory properties [53,197,198,199,200,201,202,203,204,205,206,207,208,209,210,211,212,213,214,215,216,217,218]. Plasmalogens also effectively increase resistance to chronic stress via activation of the immune system [280,281].
Large families of medical herbs have emerged as promising avenues in the treatment of AD [234,235,236,237,238,239,240,241,242,243,244,245,246,247,248,249,250,251,252,253,254,255,256,257,258,259,260,261,262,263,264,265,266,267,268,269,270,271,272,273,274]. Although the mechanisms of their therapeutic action remain largely unknown, extensive studies have identified various beneficial compounds in their extracts, including flavonoids, alkaloids, tannins, triterpenes with anti-inflammatory, anti-amyloidogenic, anticholinesterase, and antioxidant effects. These therapeutic effects suggest their potential as promising alternative treatments for AD and offer hope for addressing the difficulties posed by this incurable disease.
Given the new challenges associated with the rapidly growing problems of brain damage by the SARS-CoV-2 virus, immediate solutions aimed at increasing the resistance of the brain and its immune system to the COVID-19-mediated brain injuries are required. Since it takes 10 to 15 years for new pharmacological drugs to appear on the market, non-pharmacological methods of treating brain dysfunction induced by COVID-19 infection, including the use of PBM, plasmalogens, and medicinal herbs, should be more actively studied to develop effective alternative strategies to help COVID-19 patients.

7. Conclusions

Aβ is a normal product of neuronal metabolism eliminating from brain tissues through MLVs [44,45,128,129,151,157,277]. The conditions that can impair the function of lymphatic drainage processes, such as COVID-19 infection, or chronic sleep deprivation (less than 6 h), promote excessive accumulation of Aβ in brain tissues [3,4,29,30,128,129,282,283]. Recent findings have shown that SARS-CoV-2 virus-related accumulation of Aβ, which is identical to amyloids in AD, leads to the development of COVID-19-induced dementia [3,4,29,30]. Since virus-induced dementia develops quickly (in just a few weeks), it is logical to assume that this process may be reversible, as is the case with mild cognitive impairment [284,285,286,287,288,289]. Sleep deficiency over 20 years can lead to the development of dementia in 30% of cases, probably due to long-term Aβ deposition in brain tissues [283]. However, 70% of people experiencing chronic sleep deficiency do not develop dementia, indicating that they are either resistant to this process or it is reversible in them [283]. Thus, dementia and accumulation of Aβ in the brain associated with COVID-19 infection and sleep deficiency can be reversible and therefore treatable.
There is evidence that the dissolved form of Aβ, located in the perivascular spaces, is most toxic to neurons and synapses [290]. Soluble Aβ is rapidly cleared from the human brain within 1–2.5 h [291]. However, this clearance is impaired in AD and other forms of dementia, leading to Aβ aggregation in the brain. Because MLVs play an important role in draining brain tissues and removing toxins and metabolites from the brain, future treatments for reversible forms of dementia are expected to involve stimulation of lymphatic drainage processes. Given the growing evidence of the existence of lymphatic vessels directly in the human brain [292,293,294,295], and not only in its meninges [296,297], technologies for activating lymphatic drainage processes will contribute to progress in the treatment of a wide number of brain diseases associated with dysfunction of the cerebral lymphatic system, such as AD, Parkinson’s disease, brain trauma and oncology [44,45,46,47,48,49,118,119,162,275,276].
Recent animal studies have proposed PBM as an effective technology for stimulating the lymphatic clearance of Aβ and blood products [45,46,151,162,298,299]. However, PBM has limitations due to light scattering as it passes through the skull and the low doses used for photo-therapy [300]. Therefore, the use of PBM alone may not be sufficient to effective treat dementia, even if it is reversible. The alternative methods that could enhance the stimulatory effects of PBM may improve its therapeutic efficacy. In this review, we analyze the potential effectiveness of natural compounds, such as plasmalogens and medical herbs, to enhance lymphatic drainage processes in the brain. It is assumed that the combination of PBM and natural ingredients can become a reliable alternative in the treatment of various forms of reversible dementia.
Unlike moderate dementia, dementia associated with AD is irreversible, and there are no prospects for the emergence of new methods for completely curing this disease. However, PBM has been shown to be effective in pilot clinical trials in controlling progression of AD [128,134,135,136,137,140,141,146,147,148,149,150]. It is expected that while the search for a reliable and safe pharmacological treatment for AD is underway, the combination of PBM with natural products will help prevent the rapid progression of the disease, as well as increase the brain’s resistance to the toxic effects of Aβ.

Author Contributions

O.S.-G., N.N. and S.D. initiated and supervised this review article; O.S.-G. wrote the Abstract, Introduction, and Conclusions; O.S.-G. prepared the section “SARS-CoV-2 amyloids and COVID-19-mediated AD dementia” and “Strategies for lymphatic removal of amyloids from the brain: photobiomodulation of MLVs”; V.T., A.E. (Arina Evsiukova), A.E. (Anna Eroshova), E.D. and E.V. prepared the section “Plasmalogens as a potential therapy for AD”; N.N., D.Z. and V.A. prepared the section “Natural compounds as inhibitors of amyloid aggregation in the brain”. All authors were also involved in the editing of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

N.N., V.T., D.Z., V.A., A.E., O.S.-G. were supported by the Russian Science Foundation grant (23-75-30001).

Acknowledgments

We thank a designer, Elena Saranseva, for preparing the figures for our manuscript.

Conflicts of Interest

The authors declare that they have no competing interests. Author Sergey Diduk, Anna Eroshova, Elina Dosadina was employed by the company Leeners LLC. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

  1. Choi Matthews, K.A.; Xu, W.; Gaglioti, A.H.; Holt, J.B.; Croft, J.B.; Mack, D.; McGuire, L.C. Racial and ethnic estimates of Alzheimer’s disease and related dementias in the United States (2015–2060) in adults aged ≥65 years. Alzheimer’s Dement. 2018, 15, 17–24. [Google Scholar] [CrossRef]
  2. 2023 Alzheimer’s disease facts and figures. Alzheimer’s Dement. 2023, 19, 1598–1695. [CrossRef]
  3. Milton, N. SARS-CoV-2 amyloid, is COVID-19-exacerbated dementia an amyloid disorder in the making? Front. Dement. 2023, 2, 1233340. [Google Scholar] [CrossRef]
  4. Charnley, M.; Islam, S.; Bindra, G.K.; Engwirda, J.; Ratcliffe, J.; Zhou, J.; Mezzenga, R.; Hulett, M.D.; Han, K.; Berryman, J.T.; et al. Neurotoxic amyloidogenic peptides in the proteome of SARS-Cov-2: Potential implications for neurological symptoms in COVID-19. Nat. Commun. 2022, 13, 3387. [Google Scholar] [CrossRef]
  5. Rudnicka-Drozak, E.; Drozak, P.; Mizerski, G.; Zaborowski, T.; S’lusarska, B.; Nowicki, G.; Drożak, M. Links between COVID-19 and Alzheimer’s Disease—What Do We Already Know? Int. J. Environ. Res. Public Health 2023, 20, 2146. [Google Scholar] [CrossRef] [PubMed]
  6. Wang, L.; Davis, P.B.; Volkow, N.D.; Berger, N.A.; Kaelber, D.C.; Xu, R. Association of COVID-19 with New-Onset Alzheimer’s Disease. J. Alzheimer’s Dis. 2022, 89, 411–414. [Google Scholar] [CrossRef] [PubMed]
  7. Liu, N.; Jiang, X.; Li, H. The viral hypothesis in Alzheimer’s disease: SARS-CoV-2 on the cusp. Front. Aging Neurosci. 2023, 15, 1129640. [Google Scholar] [CrossRef] [PubMed]
  8. Rahman, M.R.; Akter, R.; Neelotpol, S.; Mayesha, I.I.; Afrose, A. The Neuropathological Impacts of COVID-19: Challenges and Alternative Treatment Options for Alzheimer’s Like Brain Changes on Severely SARS-CoV-2 Infected Patients. Am. J. Alzheimer’s Dis. Other Dement. 2023, 38, 15333175231214974. [Google Scholar] [CrossRef]
  9. Shahbaz, M.A.; Kuivanen, S.; Lampinen, R.; Mussalo, L.; Hron, T.; Závodná, T.; Ojha, R.; Krejčík, Z.; Saveleva, L.; Tahir, N.A.; et al. Human-derived air-liquid interface cultures decipher Alzheimer’s disease-SARS-CoV-2 crosstalk in the olfactory mucosa. J. Neuroinflamm. 2023, 20, 299. [Google Scholar] [CrossRef]
  10. Chen, F.; Chen, Y.; Wang, Y.; Ke, Q.; Cui, L. The COVID-19 pandemic and Alzheimer’s disease: Mutual risks and mechanisms. Transl. Neurodegener. 2022, 11, 40. [Google Scholar] [CrossRef]
  11. Amadoro, G.; Latina, V.; Stigliano, E.; Micera, A. COVID-19 and Alzheimer’s Disease Share Common Neurological and Ophthalmological Manifestations: A Bidirectional Risk in the Post-Pandemic Future. Cells 2023, 12, 2601. [Google Scholar] [CrossRef] [PubMed]
  12. Golzari-Sorkheh, M.; Weaver, D.F.; Reed, M.A. COVID-19 as a Risk Factor for Alzheimer’s Disease. J. Alzheimer’s Dis. 2023, 91, 1–23. [Google Scholar] [CrossRef] [PubMed]
  13. Chen, J.; Chen, J.; Lei, Z. Amyloid precursor protein facilitates SARS-CoV-2 virus entry into cells and enhances amyloid-β-associated pathology in APP/PS1 mouse model of Alzheimer’s disease. Transl. Psychiatry 2023, 13, 396. [Google Scholar] [CrossRef] [PubMed]
  14. Baranova, A.; Cao, H.; Zhang, F. Causal effect of COVID-19 on Alzheimer’s disease: A Mendelian randomization study. J. Med. Virol. 2023, 95, e28107. [Google Scholar] [CrossRef] [PubMed]
  15. Monllor, P.; Kumar, P.; Lloret, M.Á.; Ftara, A.; Leon, J.L.; Lopez, B.; Cervera-Ferri, A.; Lloret, A. Multifactorial Causation of Alzheimer’s Disease Due to COVID-19. J. Alzheimer’s Dis. 2023, 96, 1399–1409. [Google Scholar] [CrossRef] [PubMed]
  16. Li, W.; Sun, L.; Yue, L.; Xiao, S. Alzheimer’s disease and COVID-19: Interactions, intrinsic linkages, and the role of immunoinflammatory responses in this process. Front. Immunol. 2023, 14, 1120495. [Google Scholar] [CrossRef] [PubMed]
  17. Itzhaki, R.F. COVID-19 and Alzheimer’s Disease: What Is the Connection? J. Alzheimer’s Dis. 2023, 91, 1273–1276. [Google Scholar] [CrossRef] [PubMed]
  18. Frontera, J.A.; Yang, D.; Lewis, A.; Patel, P.; Medicherla, C. A prospective study of long-term outcomes among hospitalized COVID-19 patients with and without neurological complications. J. Neurol. Sci. 2021, 426, 117486. [Google Scholar] [CrossRef]
  19. Ousseiran, Z.H.; Fares, Y.; Chamoun, W.T. Neurological manifestations of COVID-19: A systematic review and detailed comprehension. Int. J. Neurosci. 2023, 133, 754–769. [Google Scholar] [CrossRef]
  20. Hanganu, A.R.; Niculae, C.M.; Dulămea, A.O.; Moisă, E.; Constantin, R.; Neagu, G.; Hristea, A. The outcome and risk factors associated with central and peripheral nervous system involvement in hospitalized COVID-19 patients: A retrospective cohort study. Front. Neurol. 2024, 14, 1338593. [Google Scholar] [CrossRef]
  21. Priyal; Sehgal, V.; Kapila, S.; Taneja, R.; Mehmi, P.; Gulati, N. Review of Neurological Manifestations of SARS-CoV-2. Cureus 2023, 15, 38194. [Google Scholar]
  22. Jumagaliyeva, M.; Ayaganov, D.; Saparbayev, S.; Tuychibaeva, N.; Abdelazim, I.A.; Kurmambayev, Y.; Khamidullina, Z.; Yessenamanova, S. Possible mechanism of central nervous system targeting and neurological symptoms of the new-coronavirus (COVID-19): Literature review. Eur. Rev. Med. Pharmacol. Sci. 2023, 27, 9420–9428. [Google Scholar] [PubMed]
  23. Tyagi, K.; Rai, P.; Gautam, A.; Kaur, H.; Kapoor, S.; Suttee, A.; Jaiswal, P.K.; Sharma, A.; Singh, G.; Barnwal, R.P. Neurological manifestations of SARS-CoV-2: Complexity, mechanism and associated disorders. Eur. J. Med. Res. 2023, 28, 307. [Google Scholar] [CrossRef] [PubMed]
  24. Itzhaki, R.F.; Golde, T.E.; Heneka, M.T.; Readhead, B. Do infections have a role in the pathogenesis of Alzheimer disease? Nat. Rev. Neurol. 2020, 16, 193–197. [Google Scholar] [CrossRef]
  25. Wang, F.; Xu, J.; Xu, S.J.; Guo, J.J.; Wang, F.; Wang, Q.W. Analysis and identification genetic effect of SARS-CoV-2 infections to Alzheimer’s disease patients by integrated bioinformatics. J. Alzheimer’s Dis. 2022, 85, 729–744. [Google Scholar] [CrossRef]
  26. Tang, T.; Jia, J.; Garbarino, E.; Chen, L.; Ma, J.; Li, P. Human herpesvirus 6A U4 inhibits proteasomal degradation of the amyloid precursor protein. J. Virol. 2022, 96, 168821. [Google Scholar] [CrossRef]
  27. Linard, M.; Letenneur, L.; Garrigue, I.; Doize, A.; Dartigues, J.F.; Helmer, C. Interaction between ApoE4 and herpes simplex virus type 1 in Alzheimer’s disease. Alzheimer’s Dement. 2020, 16, 200–208. [Google Scholar] [CrossRef]
  28. Niklasson, B.; Lindquist, L.; Klitz, W.; Englund, E. Picornavirus identified in Alzheimer’s disease brains: A pathogenic path? J. Alzheimer’s Dis. Rep. 2020, 4, 141–146. [Google Scholar] [CrossRef]
  29. Nystrom, S.; Hammarstrom, P. Amyloidogenesis of SARS-CoV-2 spike protein. J. Am. Chem. Soc. 2022, 144, 8945–8950. [Google Scholar] [CrossRef]
  30. Tayeb-Fligelman, E.; Bowler, J.T.; Tai, C.E.; Sawaya, M.R.; Jiang, Y.X.; Garcia, G., Jr.; Griner, S.L.; Cheng, X.; Salwinski, L.; Lutter, L.; et al. Low complexity domains of the nucleocapsid protein of SARS-CoV-2 form amyloid fibrils. Nat. Commun. 2023, 14, 2379. [Google Scholar] [CrossRef]
  31. Galkin, A.P. Hypothesis: AA amyloidosis is a factor causing systemic complications after coronavirus disease. Prion 2021, 15, 53–55. [Google Scholar] [CrossRef] [PubMed]
  32. Huang, Z. A function of amyloid-beta in mediating activity-dependent Axon/Synapse competition may unify its roles in brain physiology and pathology. J. Alzheimer’s Dis. 2023, 92, 29–57. [Google Scholar] [CrossRef] [PubMed]
  33. Bourgade, K.; Dupuis, G.; Frost, E.H.; Fulop, T. Anti-viral properties of amyloid-beta peptides. J. Alzheimer’s Dis. 2016, 54, 859–878. [Google Scholar] [CrossRef] [PubMed]
  34. Powell-Doherty, R.D.; Abbott, A.R.N.; Nelson, L.A.; Bertke, A.S. Amyloid-β and p-Tau Anti-Threat Response to Herpes Simplex Virus 1 Infection in Primary Adult Murine Hippocampal Neurons. J. Virol. 2020, 94, 01874-19. [Google Scholar] [CrossRef] [PubMed]
  35. Pearson, H.A.; Peers, C. Physiological roles for amyloid beta peptides. J. Physiol. 2006, 575, 5–10. [Google Scholar] [CrossRef] [PubMed]
  36. Passeri, E.; Elkhoury, K.; Morsink, M.; Broersen, K.; Linder, M.; Tamayol, A.; Malaplate, C.; Yen, F.T.; Arab-Tehrany, E. Alzheimer’s Disease: Treatment Strategies and Their Limitations. Int. J. Mol. Sci. 2022, 23, 13954. [Google Scholar] [CrossRef] [PubMed]
  37. Se Thoe, E.; Fauzi, A.; Tang, Y.Q.; Chamyuang, S.; Chia, A.Y.Y. A review on advances of treatment modalities for Alzheimer’s disease. Life Sci. 2021, 276, 119129. [Google Scholar] [CrossRef] [PubMed]
  38. Livingston, G.; Huntley, J.; Sommerlad, A.; Ames, D.; Ballard, C.; Banerjee, S.; Brayne, C.; Burns, A.; Cohen-Mansfield, J.; Cooper, C. Dementia Prevention, Intervention, and Care: 2020 Report of the Lancet Commission. Lancet 2020, 396, 413–446. [Google Scholar] [CrossRef] [PubMed]
  39. Howard, R.; Liu, K.Y. Questions EMERGE as Biogen claims aducanumab turnaround. Nat. Rev. Neurol. 2020, 16, 63–64. [Google Scholar] [CrossRef]
  40. Panza, F.; Lozupone, M.; Seripa, D.; Imbimbo, B. Amyloid-β immunotherapy for Alzheimer disease: Is it now a long shot? Ann. Neurol. 2019, 85, 303–315. [Google Scholar] [CrossRef]
  41. Mullard, A. Landmark Alzheimer’s drug approval confounds research community. Nature 2021, 594, 309–310. [Google Scholar] [CrossRef] [PubMed]
  42. Salloway, S.; Chalkias, S.; Barkhof, F.; Burkett, P.; Barakos, J.; Purcell, D.; Suhy, J.; Forrestal, F.; Tian, Y.; Umans, K. Amyloid-Related Imaging Abnormalities in 2 Phase 3 Studies Evaluating Aducanumab in Patients with Early Alzheimer Disease. JAMA Neurol. 2022, 79, 13–21. [Google Scholar] [CrossRef] [PubMed]
  43. Da Mesquita, S.; Papadopoulos, Z.; Dykstra, T.; Brase, L.; Farias, F.G.; Wall, M.; Jiang, H.; Kodira, C.D.; de Lima, K.A.; Herz, J.; et al. Meningeal lymphatics affect microglia responses and anti-Aβ immunotherapy. Nature 2021, 593, 255–260. [Google Scholar] [CrossRef] [PubMed]
  44. Da Mesquita, S.; Louveau, A.; Vaccari, A.; Smirnov, I.; Cornelison, R.C.; Kingsmore, K.M.; Contarino, C.; Onengut-Gumuscu, S.; Farber, E.; Raper, D.; et al. Functional aspects of meningeal lymphatics in ageing and Alzheimer’s disease. Nature 2018, 560, 185–191. [Google Scholar] [CrossRef]
  45. Semyachkina-Glushkovskaya, O.; Shirokov, A.; Blokhina, I.; Fedosov, I.; Terskov, A.; Dubrovsky, A.; Tsoy, M.; Zlatogosrskaya, D.; Adushkina, V.; Evsukova, A.; et al. Mechanisms of phototherapy of Alzheimer’s disease during sleep and wakefulness: The role of the meningeal lymphatics. Front. Optoelectron. 2023, 16, 22. [Google Scholar]
  46. Li, D.; Lin, H.; Sun, S.; Liu, S.; Liu, Z.; He, Y.; Zhu, J.; Xu, J.; Semyachkina-Glushkovskaya, O.; Yu, T.; et al. Photostimulation of lymphatic clearance of β- amyloid from mouse brain: New strategy for the therapy of Alzheimer’s disease. Front. Optoelectron. 2023, 16, 45. [Google Scholar] [CrossRef]
  47. Dupont, G.; Iwanaga, J.; Yilmaz, E.; Tubbs, R.S. Connections Between Amyloid Beta and the Meningeal Lymphatics As a Possible Route for Clearance and Therapeutics. Lymphat. Res. Biol. 2020, 18, 2–6. [Google Scholar] [CrossRef] [PubMed]
  48. Rockson, S.G. Alzheimer’s Disease: Can the Meningeal Lymphatics Provide the Answer? Lymphat. Res. Biol. 2020, 18, 1. [Google Scholar] [CrossRef]
  49. Chen, J.; Pan, Y.; Liu, Q.; Li, G.; Chen, G.; Li, W.; Zhao, W.; Wang, Q. The Interplay between Meningeal Lymphatic Vessels and Neuroinflammation in Neurodegenerative Diseases. Curr. Neuropharmacol. 2024, 22, 1016–1032. [Google Scholar] [CrossRef]
  50. Lemprière, S. Meningeal lymphatics mediate drainage of viruses from the CNS. Nat. Rev. Neurol. 2022, 18, 382. [Google Scholar] [CrossRef]
  51. Goodman, J.R.; Adham, Z.O.; Woltjer, R.L.; Lund, A.W.; Iliff, J.J. Characterization of dural sinus-associated lymphatic vasculature in human Alzheimer’s dementia subjects. Brain Behav. Immun. 2018, 73, 34–40. [Google Scholar] [CrossRef] [PubMed]
  52. Pagano, K.; Tomaselli, S.; Molinari, H.; Ragona, L. Natural Compounds as Inhibitors of Aβ Peptide Aggregation: Chemical Requirements and Molecular Mechanisms. Front. Neurosci. 2020, 14, 619667. [Google Scholar] [CrossRef] [PubMed]
  53. Gu, J.; Chen, L.; Sun, R.; Wang, J.-L.; Wang, J.; Lin, Y.; Lei, S.; Zhang, Y.; Lv, D.; Jiang, F.; et al. Plasmalogens Eliminate Aging-Associated Synaptic Defects and Microglia-Mediated Neuroinflammation in Mice. Front. Mol. Biosci. 2022, 9, 815320. [Google Scholar] [CrossRef]
  54. Ezzat, K.; Pernemalm, M.; Palsson, S.; Roberts, T.C.; Jarver, P.; Dondalska, A.; Bestas, B.; Sobkowiak, M.J.; Levänen, B.; Sköld, M.; et al. The viral protein corona directs viral pathogenesis and amyloid aggregation. Nat. Commun. 2019, 10, 2331. [Google Scholar] [CrossRef] [PubMed]
  55. Leger, P.; Nachman, E.; Richter, K.; Tamietti, C.; Koch, J.; Burk, R.; Kummer, S.; Xin, Q.; Stanifer, M.; Bouloy, M.; et al. NSs amyloid formation is associated with the virulence of Rift Valley fever virus in mice. Nat. Commun. 2020, 11, 3281. [Google Scholar] [CrossRef] [PubMed]
  56. Sampson, T.R.; Challis, C.; Jain, N.; Moiseyenko, A.; Ladinsky, M.S.; Shastri, G.G.; Thron, T.; Needham, B.D.; Horvath, I.; Debelius, J.W.; et al. A gut bacterial amyloid promotes alpha-synuclein aggregation and motor impairment in mice. eLife 2020, 9, e53111. [Google Scholar] [CrossRef] [PubMed]
  57. Saumya, K.U.; Gadhave, K.; Kumar, A.; Giri, R. Zika virus capsid anchor forms cytotoxic amyloid-like fibrils. Virology 2021, 560, 8–16. [Google Scholar] [CrossRef] [PubMed]
  58. Yadav, R.; Chaudhary, J.K.; Jain, N.; Chaudhary, P.K.; Khanra, S.; Dhamija, P.; Sharma, A.; Kumar, A.; Handu, S. Role of structural and non-structural proteins and therapeutic targets of SARS-CoV-2 for COVID-19. Cells 2021, 10, 821. [Google Scholar] [CrossRef] [PubMed]
  59. Crooke, S.N.; Ovsyannikova, I.G.; Kennedy, R.B.; Poland, G.A. Immunoinformatic identification of B cell and T cell epitopes in the SARS-CoV-2 proteome. Sci. Rep. 2020, 10, 14179. [Google Scholar] [CrossRef]
  60. Cao, S.; Song, Z.; Rong, J.; Andrikopoulos, N.; Liang, X.; Wang, Y.; Peng, G.; Ding, F.; Ke, P.C. Spike Protein Fragments Promote Alzheimer’s Amyloidogenesis. ACS Appl. Mater. Interfaces 2023, 15, 40317–40329. [Google Scholar] [CrossRef]
  61. Hassan, S.S.; Choudhury, P.P.; Dayhoff, G.W., 2nd; Aljabali, A.A.A.; Uhal, B.D.; Lundstrom, K.; Rezaei, N.; Pizzol, D.; Adadi, P.; Lal, A. The importance of accessory protein variants in the pathogenicity of SARS-CoV-2. Arch. Biochem. Biophys. 2022, 717, 109124. [Google Scholar] [CrossRef] [PubMed]
  62. Abavisani, M.; Rahimian, K.; Mahdavi, B.; Tokhanbigli, S.; Mollapour Siasakht, M.; Farhadi, A.; Kodori, M.; Mahmanzar, M.; Meshkat, Z. Mutations in SARS-CoV-2 structural proteins: A global analysis. Virol. J. 2022, 19, 220. [Google Scholar] [CrossRef] [PubMed]
  63. Idrees, D.; Kumar, V. SARS-CoV-2 spike protein interactions with amyloidogenic proteins: Potential clues to neurodegeneration. Biochem. Biophys. Res. Commun. 2021, 554, 94–98. [Google Scholar] [CrossRef] [PubMed]
  64. Kyriakopoulos, A.M.; Nigh, G.; Mccullough, P.A.; Seneff, S. Mitogen activated protein kinase (MAPK) activation, p53, and autophagy inhibition characterize the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein induced neurotoxicity. Cureus 2022, 14, e32361. [Google Scholar] [CrossRef] [PubMed]
  65. Lee, J.G.; Huang, W.; Lee, H.; Van De Leemput, J.; Kane, M.A.; Han, Z. Characterization of SARS-CoV-2 proteins reveals Orf6 pathogenicity, subcellular localization, host interactions and attenuation by Selinexor. Cell. Biosci. 2021, 11, 58. [Google Scholar] [CrossRef] [PubMed]
  66. Hassan, S.S.; Lundstrom, K.; Serrano-Aroca, A.; Adadi, P.; Aljabali, A.A.A.; Redwan, E.M.; Lal, A.; Kandimalla, R.; Abd El-Aziz, T.M.; Choudhury, P.P.; et al. Emergence of unique SARS-CoV-2 ORF10 variants and their impact on protein structure and function. Int. J. Biol. Macromol. 2022, 194, 128–143. [Google Scholar] [CrossRef]
  67. Patel, N.S.; Paris, D.; Mathura, V.; Quadros, A.N.; Crawford, F.C.; Mullan, M.J. Inflammatory cytokine levels correlate with amyloid load in transgenic mouse models of Alzheimer’s disease. J. Neuroinflamm. 2005, 2, 9. [Google Scholar] [CrossRef] [PubMed]
  68. Tayeb-Fligelman, E.; Cheng, X.; Tai, C.; Bowler, J.T.; Griner, S.; Sawaya, M.R.; Seidler, P.M.; Jiang, Y.X.; Lu, J.; Rosenberg, G.M.; et al. Inhibition of amyloid formation of the Nucleoprotein of SARS-CoV-2. bioRxiv 2021, 5, 434000. [Google Scholar]
  69. Chen, H.; Cui, Y.; Han, X.; Hu, W.; Sun, M.; Zhang, Y.; Wang, P.H.; Song, G.; Chen, W.; Lou, J. Liquid–liquid phase separation by SARS-CoV-2 nucleocapsid protein and RNA. Cell Res. 2020, 30, 1143–1145. [Google Scholar] [CrossRef]
  70. Savastano, A.; Ibáñez de Opakua, A.; Rankovic, M.; Zweckstetter, M. Nucleocapsid protein of SARS-CoV-2 phase separates into RNA-rich polymerase-containing condensates. Nat. Commun. 2020, 11, 6041. [Google Scholar] [CrossRef]
  71. Pham, C.L.; Shanmugam, N.; Strange, M.; O’Carroll, A.; Brown, J.W.; Sierecki, E.; Gambin, Y.; Steain, M.; Sunde, M. Viral M45 and necroptosis-associated proteins form heteromeric amyloid assemblies. EMBO Rep. 2019, 20, e46518. [Google Scholar] [CrossRef] [PubMed]
  72. Koralnik, I.J.; Tyler, K.L. COVID-19: A global threat to the nervous system. Ann. Neurol. 2020, 88, 1–11. [Google Scholar] [CrossRef] [PubMed]
  73. Divani, A.A.; Andalib, S.; Biller, J.; Di Napoli, M.; Moghimi, N.; Rubinos, C.A.; Nobleza, C.O.; Sylaja, P.N.; Toledano, M.; Lattanzi, S.; et al. Central nervous system manifestations associated with COVID-19. Curr. Neurol. Neurosci. Rep. 2020, 20, 60. [Google Scholar] [CrossRef]
  74. Iadecola, C.; Anrather, J.; Kamel, H. Effects of COVID-19 on the nervous system. Cell 2020, 183, 16–27. [Google Scholar] [CrossRef] [PubMed]
  75. Douaud, G.; Lee, S.; Alfaro-Almagro, F.; Arthofer, C.; Wang, C.; Mccarthy, P.; Lange, F.; Andersson, J.L.; Griffanti, L.; Duff, E.; et al. SARS-CoV-2 is associated with changes in brain structure in UK Biobank. Nature 2022, 604, 697–707. [Google Scholar] [CrossRef]
  76. Serrano-Castro, P.J.; Garzon-Maldonado, F.J.; Casado-Naranjo, I.; Ollero-Ortiz, A.; Minguez-Castellanos, A.; Iglesias-Espinosa, M.; Baena-Palomino, P.; Sánchez-Sanchez, V.; Sánchez-Pérez, R.M.; Rubi-Callejon, J.; et al. The cognitive and psychiatric subacute impairment in severe Covid-19. Sci. Rep. 2022, 12, 3563. [Google Scholar] [CrossRef]
  77. Lu, Y.; Li, X.; Geng, D.; Mei, N.; Wu, P.Y.; Huang, C.C.; Jia, T.; Zhao, Y.; Wang, D.; Xiao, A.; et al. Cerebral micro-structural changes in COVID-19 patients—An MRI-based 3-month follow-up study: A brief title: Cerebral changes in COVID-19. eClinicalMedicine 2020, 25, 100484. [Google Scholar] [CrossRef] [PubMed]
  78. Ahmed, M.; Roy, S.; Iktidar, M.A.; Chowdhury, S.; Akter, S.; Islam, A.; Hawlader, M. Post-COVID-19 memory complaints: Prevalence and associated factors. Neurología, 2022; Online ahead of print. [Google Scholar]
  79. Douaud, G.; Lee, S.; Alfaro-Almagro, F.; Arthofer, C.; Wang, C.; Lange, F.; Andersson, J.L.; Griffanti, L.; Duff, E.; Jbabdi, S. Brain imaging before and after COVID-19 in UK Biobank. MedRxiv: Prepr. Serv. Health Sci. 2021. [Google Scholar] [CrossRef]
  80. Köhncke, Y.; Kühn, S.; Düzel, S.; Sander, M.C.; Brandmaier, A.M.; Lindenberger, U. Grey-matter structure in cortical and limbic regions correlates with general cognitive ability in old age. Aging Brain 2023, 5, 100103. [Google Scholar] [CrossRef]
  81. Poole, V.N.; Oveisgharan, S.; Yu, L.; Dawe, R.J.; Leurgans, S.E.; Zhang, S.; Arfanakis, K.; Buchman, A.S.; Bennett, D.A. Volumetric brain correlates of gait associated with cognitive decline in community-dwelling older adults. Front. Aging Neurosci. 2023, 15, 1194986. [Google Scholar] [CrossRef]
  82. Han, A.Y.; Mukdad, L.; Long, J.L.; Lopez, I.A. Anosmia in COVID-19: Mechanisms and significance. Chem. Sens. 2020, 45, 423–428. [Google Scholar] [CrossRef] [PubMed]
  83. Ritchie, K.; Chan, D.; Watermeyer, T. The cognitive consequences of the COVID-19 epidemic: Collateral damage? Brain Commun. 2020, 2, fcaa069. [Google Scholar] [CrossRef] [PubMed]
  84. Eichenbaum, H. The hippocampus and declarative memory: Cognitive mechanisms and neural codes. Behav. Brain Res. 2001, 127, 199–207. [Google Scholar] [CrossRef] [PubMed]
  85. Fortin, N.J.; Agster, K.L.; Eichenbaum, H.B. Critical role of the hippocampus in memory for sequences of events. Nat. Neurosci. 2002, 5, 458–462. [Google Scholar] [CrossRef] [PubMed]
  86. Furcila, D.; Defelipe, J.; Alonso-Nanclares, L. A study of amyloid-beta and phosphotau in plaques and neurons in the hippocampus of Alzheimer’s disease patients. J. Alzheimer’s Dis. 2018, 64, 417–435. [Google Scholar] [CrossRef] [PubMed]
  87. Lopez, O.L.; Becker, J.T.; Chang, Y.; Klunk, W.E.; Mathis, C.; Price, J.; Aizenstein, H.J.; Snitz, B.; Cohen, A.D.; DeKosky, S.T.; et al. Amyloid deposition and brain structure as long-term predictors of MCI, dementia, and mortality. Neurology 2018, 90, e1920–e1928. [Google Scholar] [CrossRef] [PubMed]
  88. Ma, G.; Zhang, D.F.; Zou, Q.C.; Xie, X.; Xu, L.; Feng, X.L.; Li, X.; Han, J.B.; Yu, D.; Deng, Z.H.; et al. SARS-CoV-2 Spike protein S2 subunit modulates gamma-secretase and enhances amyloid-beta production in COVID-19 neuropathy. Cell Discov. 2022, 8, 99. [Google Scholar] [CrossRef]
  89. Priemer, D.S.; Rhodes, C.H.; Karlovich, E.; Perl, D.P.; Goldman, J.E. Abeta deposits in the neocortex of adult and infant hypoxic brains, including in cases of COVID-19. J. Neuropathol. Exp. Neurol. 2022, 81, 988–995. [Google Scholar] [CrossRef]
  90. Ziff, O.J.; Ashton, N.J.; Mehta, P.R.; Brown, R.; Athauda, D.; Heaney, J.; Heslegrave, A.J.; Benedet, A.L.; Blennow, K.; Checkley, A.M.; et al. Amyloid processing in COVID-19-associated neurological syndromes. J. Neurochem. 2022, 161, 146–157. [Google Scholar] [CrossRef]
  91. Chiricosta, L.; Gugliandolo, A.; Mazzon, E. SARS-CoV-2 exacerbates beta-amyloid neurotoxicity, inflammation and oxidative stress in Alzheimer’s disease patients. Int. J. Mol. Sci. 2021, 22, 13603. [Google Scholar] [CrossRef]
  92. Gordon, M.N.; Heneka, M.T.; Le Page, L.M.; Limberger, C.; Morgan, D.; Tenner, A.J.; Terrando, N.; Willette, A.A.; Willette, S.A. Impact of COVID-19 on the onset and progression of Alzheimer’s disease and related dementias: A roadmap for future research. Alzheimer’s Dement. 2022, 18, 1038–1046. [Google Scholar] [CrossRef]
  93. Solomon, I.H.; Normandin, E.; Bhattacharyya, S.; Mukerji, S.S.; Keller, K.; Ali, A.S.; Adams, G.; Hornick, J.L.; Padera, R.F., Jr.; Sabeti, P. Neuropathological Features of Covid-19. N. Engl. J. Med. 2020, 383, 989–992. [Google Scholar] [CrossRef] [PubMed]
  94. Thakur, K.T.; Miller, E.H.; Glendinning, M.D.; Al-Dalahmah, O.; Banu, M.A.; Boehme, A.K.; Boubour, A.; Bruce, S.; Chong, A.M.; Claassen, J.; et al. COVID-19 neuropathology at Columbia University Irving Medical Center/New York Presbyterian Hospital. Brain 2021, 144, 2696–2708. [Google Scholar] [CrossRef] [PubMed]
  95. Adingupu, D.D.; Soroush, A.; Hansen, A.; Twomey, R.; Dunn, J.F. Brain hypoxia, neurocognitive impairment, and quality of life in people post-COVID-19. J. Neurol. 2023, 270, 3303–3314. [Google Scholar] [CrossRef] [PubMed]
  96. Balsak, S.; Atasoy, B.; Donmez, Z.; Yabul, F.C.; Daşkaya, H.; Akkoyunlu, Y.; Yurtsever, İ.; Sarı, L.; Sijahovic, S.; Akcay, A.; et al. Microstructural alterations in hypoxia-related BRAIN centers after COVID-19 by using DTI: A preliminary study. J. Clin. Ultrasound 2023, 51, 1276–1283. [Google Scholar] [CrossRef] [PubMed]
  97. Lall, R.; Mohammed, R.; Ojha, U. What are the links between hypoxia and Alzheimer’s disease? Neuropsychiatr. Dis. Treat. 2019, 15, 1343–1354. [Google Scholar] [CrossRef] [PubMed]
  98. Raz, L.; Knoefel, J.; Bhaskar, K. The neuropathology and cerebrovascular mechanisms of dementia. J. Cereb. Blood Flow Metab. 2016, 36, 172–186. [Google Scholar] [CrossRef]
  99. Hassan, H.; Chen, R. Hypoxia in Alzheimer’s disease: Effects of hypoxia inducible factors. Neural Regen. Res. 2021, 16, 310–311. [Google Scholar]
  100. Shobatake, R.; Ota, H.; Takahashi, N.; Ueno, S.; Sugie, K.; Takasawa, S. The impact of intermittent hypoxia on metabolism and cognition. Int. J. Mol. Sci. 2022, 23, 12957. [Google Scholar] [CrossRef] [PubMed]
  101. Wang, X.; Xie, Y.; Chen, G.; Lu, Y.; Wang, D.; Zhu, L. Intermittent hypoxia therapy ameliorates beta-amyloid pathology via TFEB-mediated autophagy in murine Alzheimer’s disease. J. Neuroinflamm. 2023, 20, 240. [Google Scholar] [CrossRef]
  102. Xie, J.C.; Ma, X.Y.; Liu, X.H.; Yu, J.; Zhao, Y.C.; Tan, Y.; Liu, X.Y.; Zhao, Y.X. Hypoxia increases amyloid-β level in exosomes by enhancing the interaction between CD147 and Hook1. Am. J. Transl. Res. 2018, 10, 150–163. [Google Scholar] [PubMed]
  103. Salminen, A.; Kauppinen, A.; Kaarniranta, K. Hypoxia/ischemia activate processing of Amyloid Precursor Protein: Impact of vascular dysfunction in the pathogenesis of Alzheimer’s disease. J. Neurochem. 2017, 140, 536–549. [Google Scholar] [CrossRef] [PubMed]
  104. Semyachkina-Glushkovskaya, O.; Mamedova, A.; Vinnik, V.; Klimova, M.; Saranceva, E.; Ageev, V.; Yu, T.; Zhu, D.; Penzel, T.; Kurths, J. Brain Mechanisms of COVID-19-Sleep Disorders. Int. J. Mol. Sci. 2021, 22, 6917. [Google Scholar] [CrossRef] [PubMed]
  105. Montazersaheb, S.; Hosseiniyan Khatibi, S.M.; Hejazi, M.S.; Tarhriz, V.; Farjami, A.; Sorbeni, F.G.; Farahzadi, R.; Ghasemnejad, T. COVID-19 infection: An overview on cytokine storm and related interventions. Virol. J. 2022, 19, 92. [Google Scholar] [CrossRef] [PubMed]
  106. Zanza, C.; Romenskaya, T.; Manetti, A.C.; Franceschi, F.; La Russa, R.; Bertozzi, G.; Maiese, A.; Savioli, G.; Volonnino, G.; Longhitano, Y. Cytokine Storm in COVID-19: Immunopathogenesis and Therapy. Medicina 2022, 58, 144. [Google Scholar] [CrossRef] [PubMed]
  107. Hernández-Parra, H.; Reyes-Hernández, O.D.; Figueroa-González, G.; González-Del Carmen, M.; González-Torres, M.; Peña-Corona, S.I.; Florán, B.; Cortés, H.; Leyva-Gómez, G. Alteration of the blood-brain barrier by COVID-19 and its implication in the permeation of drugs into the brain. Front. Cell. Neurosci. 2023, 17, 1125109. [Google Scholar] [CrossRef] [PubMed]
  108. Suprewicz, Ł.; Fiedoruk, K.; Czarnowska, A.; Sadowski, M.; Strzelecka, A.; Galie, P.A.; Janmey, P.A.; Kułakowska, A.; Bucki, R. Blood-brain barrier function in response to SARS-CoV-2 and its spike protein. Neurol. Neurochir. Pol. 2023, 57, 14–25. [Google Scholar] [CrossRef] [PubMed]
  109. Erickson, M.A.; Rhea, E.M.; Knopp, R.C.; Banks, W.A. Interactions of SARS-CoV-2 with the Blood–Brain Barrier. Int. J. Mol. Sci. 2021, 22, 2681. [Google Scholar] [CrossRef] [PubMed]
  110. Crunfli, F.; Carregari, V.C.; Veras, F.P.; Silva, L.S.; Nogueira, M.H.; Antunes, A.; Vendramini, P.H.; Valença, A.G.; Brandão-Teles, C.; Zuccoli, G.D.; et al. Morphological, cellular, and molecular basis of brain infection in COVID-19 patients. Proc. Natl. Acad. Sci. USA 2022, 119, e2200960119. [Google Scholar] [CrossRef]
  111. Stein, S.R.; Ramelli, S.C.; Grazioli, A.; Chung, J.Y.; Singh, M.; Yinda, C.K.; Winkler, C.W.; Sun, J.; Dickey, J.M.; Ylaya, K.; et al. SARS-CoV-2 infection and persistence in the human body and brain at autopsy. Nature 2022, 612, 758–763. [Google Scholar] [CrossRef]
  112. Hosp, J.A.; Dressing, A.; Blazhenets, G.; Bormann, T.; Rau, A.; Schwabenland, M.; Thurow, J.; Wagner, D.; Waller, C.; Niesen, W.D.; et al. Cognitive impairment and altered cerebral glucose metabolism in the subacute stage of COVID-19. Brain 2021, 144, 1263–1276. [Google Scholar] [CrossRef] [PubMed]
  113. Taquet, M.; Sillett, R.; Zhu, L.; Mendel, J.; Camplisson, I.; Dercon, Q.; Harrison, P.J. Neurological and psychiatric risk trajectories after SARS-CoV-2 infection: An analysis of 2-year retrospective cohort studies including 1,284,437 patients. Lancet Psychiatry 2022, 9, 815–827. [Google Scholar] [CrossRef] [PubMed]
  114. Li, X.; Qi, L.; Yang, D.; Hao, S.; Zhang, F.; Zhu, X.; Sun, Y.; Chen, C.; Ye, J.; Yang, J.; et al. Meningeal lymphatic vessels mediate neurotropic viral drainage from the central nervous system. Nat. Neurosci. 2022, 25, 577–587. [Google Scholar] [CrossRef] [PubMed]
  115. Rebejac, J.; Eme-Scolan, E.; Rua, R. Role of meningeal immunity in brain function and protection against pathogens. J. Inflamm. 2024, 21, 3. [Google Scholar] [CrossRef] [PubMed]
  116. Kovacs, M.A.; Cowan, M.N.; Babcock, I.W.; Sibley, L.A.; Still, K.; Batista, S.J.; Labuzan, S.A.; Sethi, I.; Harris, T.H. Meningeal lymphatic drainage promotes T cell responses against Toxoplasma gondii but is dispensable for parasite control in the brain. eLife 2022, 11, e80775. [Google Scholar] [CrossRef] [PubMed]
  117. Abbaoui, A.; Fatoba, O.; Yamashita, T. Meningeal T cells function in the central nervous system homeostasis and neurodegenerative Diseases. Front. Cell. Neurosci. 2023, 17, 1181071. [Google Scholar] [CrossRef]
  118. Song, E.; Mao, T.; Dong, H.; Boisserand, L.S.B.; Antila, S.; Bosenberg, M.; Alitalo, K.; Thomas, J.L.; Iwasaki, A. VEGF-C-driven lymphatic drainage enables immunosurveillance of brain tumours. Nature 2020, 577, 689–694. [Google Scholar] [CrossRef]
  119. Hu, X.T.; Deng, Q.; Ma, L.; Li, Q.; Chen, Y.; Liao, Y.; Zhou, F.; Zhang, C.; Shao, L.; Feng, J.; et al. Meningeal lymphatic vessels regulate brain tumor drainage and immunity. Cell Res. 2020, 30, 229–243. [Google Scholar] [CrossRef]
  120. Silva, M.T.T. Viral encephalitis. Arq. Neuropsiquiatr. 2013, 71, 703–709. [Google Scholar] [CrossRef]
  121. Solomon, T. Flavivirus encephalitis—Reply. N. Engl. J. Med. 2004, 351, 1804. [Google Scholar] [CrossRef]
  122. Tyler, K.L. Herpes simplex virus infections of the central nervous system: Encephalitis and meningitis, including Mollaret’s. Herpes 2004, 11, 57A–64A. [Google Scholar]
  123. Moseman, E.A.; Blanchard, A.C.; Nayak, D.; McGavern, D.B. T cell engagement of cross-presenting microglia protects the brain from a nasal virus infection. Sci. Immunol. 2020, 5, eabb1817. [Google Scholar] [CrossRef] [PubMed]
  124. Raval, U.; Trageser, K.J.; Naughton, S.X.; Griggs, E.; Iqbal, U.H.; Wu, H.; Rahim, M.A.; Harary, J.M.; Gursahai, S.; Pasinetti, G.M. COVID-19 and Alzheimer’s disease: Meninges-mediated neuropathology. Alzheimer’s Dement. 2021, 17, e056418. [Google Scholar]
  125. Wostyn, P. COVID-19 and chronic fatigue syndrome: Is the worst yet to come? Med. Hypotheses 2021, 146, 110469. [Google Scholar] [CrossRef]
  126. Patabendige, A.; Michael, B.D.; Craig, A.G.; Solomon, T. Brain microvascular endothelial–astrocyte cell responses following Japanese encephalitis virus infection in an in vitro human blood–brain barrier model. Mol. Cell Neurosci. 2018, 89, 60–70. [Google Scholar] [CrossRef]
  127. Mustafa, Y.M.; Meuren, L.M.; Coelho, S.V.A.; de Arruda, L.B. Pathways exploited by flaviviruses to counteract the blood–brain barrier and invade the central nervous system. Front. Microbiol. 2019, 10, 525. [Google Scholar] [CrossRef] [PubMed]
  128. Semyachkina-Glushkovskaya, O.; Penzel, T.; Poluektov, M.; Fedosov, I.; Tzoy, M.; Terskov, A.; Blokhina, I.; Sidorov, V.; Kurths, J. Phototherapy of Alzheimer’s Disease: Photostimulation of Brain Lymphatics during Sleep: A Systematic Review. Int. J. Mol. Sci. 2023, 24, 10946. [Google Scholar] [CrossRef] [PubMed]
  129. Semyachkina-Glushkovskaya, O.; Fedosov, I.; Penzel, T.; Li, D.; Yu, T.; Telnova, V.; Kaybeleva, E.; Saranceva, E.; Terskov, A.; Khorovodov, A.; et al. Brain Waste Removal System and Sleep: Photobiomodulation as an Innovative Strategy for Night Therapy of Brain Diseases. Int. J. Mol. Sci. 2023, 24, 3221. [Google Scholar] [CrossRef]
  130. Lim, L. The Growing Evidence for Photobiomodulation as a Promising Treatment for Alzheimer’s Disease. J. Biosci. Med. 2018, 6, 100–110. [Google Scholar] [CrossRef]
  131. Berman, M.H.; Nichols, T.W. Treatment of Neurodegeneration: Integrating Photobiomodulation and Neurofeedback in Alzheimer’s Dementia and Parkinson’s: A Review. Photobiomodul. Photomed. Laser Surg. 2019, 37, 623–634. [Google Scholar] [CrossRef]
  132. Caldieraro, M.A.; Laufer-Silva, T.; Cassano, P. Dosimetry and Clinical Efficacy of Transcranial Photobiomodulation for Major Depression Disorder: Could they Guide Dosimetry for Alzheimer’s Disease? J. Alzheimer’s Dis. 2021, 83, 1453–1469. [Google Scholar] [CrossRef]
  133. Pan, W.T.; Liu, P.M.; Ma, D.; Yang, J.J. Advances in photobiomodulation for cognitive improvement by near-infrared derived multiple strategies. J. Transl. Med. 2023, 21, 135. [Google Scholar] [CrossRef] [PubMed]
  134. Saltmarche, A.E.; Naeser, M.A.; Ho, K.F.; Hamblin, M.R.; Lim, L. Significant Improvement in Cognition in Mild to Moderately Severe Dementia Cases Treated with Transcranial Plus Intranasal Photobiomodulation: Case Series Report. Photomed. Laser Surg. 2017, 35, 432–441. [Google Scholar] [CrossRef] [PubMed]
  135. Chao, L.L. Effects of Home Photobiomodulation Treatments on Cognitive and Behavioral Function, Cerebral Perfusion, and Resting-State Functional Connectivity in Patients with Dementia: A Pilot Trial. Photobiomodul. Photomed. Laser Surg. 2019, 37, 133–141. [Google Scholar] [CrossRef]
  136. Maksimovich, I.V. Dementia and cognitive impairment reduction after laser transcatheter treatment of Alzheimer’s disease. World J. Neurosci. 2015, 5, 189–203. [Google Scholar] [CrossRef]
  137. Nizamutdinov, D.; Qi, X.; Berman, M.; Dougal, G.; Dayawansa, S.; Wu, E.; Yi, S.; Stevens, A.; Huang, J. Transcranial near infrared light stimulations improve cognition in patients with dementia. Aging Dis. 2021, 12, 954–963. [Google Scholar] [CrossRef]
  138. Hamblin, M.R. Photobiomodulation for Alzheimer’s Disease: Has the Light Dawned? Photonics 2019, 6, 77. [Google Scholar] [CrossRef]
  139. Yang, M.; Yang, Z.; Wang, P.; Sun, Z. Current application and future directions of photobiomodulation in central nervous diseases. Neural Regen. Res. 2021, 16, 1177–1185. [Google Scholar] [CrossRef]
  140. Stephan, W.; Banas, L.; Hamblin, M. Treatment Efficacy of Photobiomodulation for Moderate and Advanced Dementia or Alzheimer’s Disease: Case Studies. Adv. Alzheimer’s Dis. 2022, 11, 39–47. [Google Scholar] [CrossRef]
  141. Enengl, J.; Hamblin, M.; Dungel, P. Photobiomodulation for Alzheimer’s Disease: Translating Basic Research to Clinical Application. J. Alzheimer’s Dis. 2020, 75, 1073–1082. [Google Scholar] [CrossRef]
  142. Cho, G.; Lee, S.; Park, J.; Kim, M.; Park, M.; Choi, B.; Shin, Y.; Kim, N.; Shin, H. Photobiomodulation using a low-level light-emitting diode improves cognitive dysfunction in the 5XFAD mouse model of Alzheimer’s disease. J. Gerontol. Ser. A Biol. Sci. Med. Sci. 2020, 75, 631–639. [Google Scholar] [CrossRef] [PubMed]
  143. Chan, A.S.; Lee, T.; Yeung, M.; Hamblin, M. Photobiomodulation improves the frontal cognitive function of older adults. Int. J. Geriatr. Psychiatry 2019, 2, 369–377. [Google Scholar] [CrossRef] [PubMed]
  144. Jahan, A.; Nazari, M.; Mahmoudi, J. Transcranial near-infrared photobiomodulation could modulate brain electrophysiological features and attentional performance in healthy young adults. Lasers Med. Sci. 2019, 34, 1193–1200. [Google Scholar] [CrossRef] [PubMed]
  145. Maksimovich, I.V. Laser Technologies as a New Direction in Transcatheter Interventions. Photobiomodul. Photomed. Laser Surg. 2019, 37, 455–456. [Google Scholar] [CrossRef] [PubMed]
  146. Chao, L. Impact of Photobiomodulation (PBM) on Biomarkers of Alzheimer’s Disease (PBMbiomarker). 2022. Available online: https://clinicaltrials.gov/ct2/show/NCT03405662 (accessed on 28 April 2024).
  147. Lah, J. Stimulating Neural Activity to Improve Blood Flow and Reduce Amyloid: Path to Clinical Trials. 2021. Available online: https://clinicaltrials.gov/ct2/show/NCT03543878 (accessed on 28 April 2024).
  148. Salehpour, F.; Hamblin, M.R.; Di Duro, J. Rapid Reversal of Cognitive Decline, Olfactory Dysfunction, and Quality of Life Using Multi-Modality Photobiomodulation Therapy: Case Report. Photobiomodul. Photomed. Laser Surg. 2019, 37, 159–167. [Google Scholar] [CrossRef] [PubMed]
  149. Qi, X.; Nizamutdinov, D.; Berman, M.; Dougal, G.; Chazot, P.; Wu, E.; Stevens, A.; Yi, S.; Huang, J. Gender Differences of Dementia in Response to Intensive Self-Administered Transcranial and Intraocular Near-Infrared Stimulation. Cureus 2021, 13, e16188. [Google Scholar] [CrossRef] [PubMed]
  150. Horner, S.; Berger, L.; Gibas, K. Nutritional Ketosis and photobiomodulation remediate mitochondria warding off Alzheimer’s disease in a diabetic, ApoE4+ patient with mild cognitive impairment: A case report. Photodiagn. Photodyn. Ther. 2020, 30, 101777. [Google Scholar] [CrossRef] [PubMed]
  151. Semyachkina-Glushkovskaya, O.; Penzel, T.; Blokhina, I.; Khorovodov, A.; Fedosov, I.; Yu, T.; Karandin, G.; Evsukova, A.; Elovenko, D.; Adushkina, V.; et al. Night Photostimulation of Clearance of Beta-Amyloid from Mouse Brain: New Strategies in Preventing Alzheimer’s Disease. Cells 2021, 10, 3289. [Google Scholar] [CrossRef] [PubMed]
  152. Semyachkina-Glushkovskaya, O.; Postnov, D.; Lavrova, A.; Fedosov, I.; Borisova, E.; Nikolenko, V.; Penzel, T.; Kurths, J.; Tu-chin, V. Biophotonic Strategies of Measurement and Stimulation of the Cranial and the Extracranial Lymphatic Drainage Function. IEEE J. Sel. Top. 2021, 27, 7400313. [Google Scholar] [CrossRef]
  153. Semyachkina-Glushkovskaya, O.; Abdurashitov, A.; Dubrovsky, A.; Klimova, M.; Agranovich, I.; Terskov, A.; Shirokov, A.; Vinnik, V.; Kuznecova, A.; Lezhnev, N.; et al. Photobiomodulation of lymphatic drainage and clearance: Perspective strategy for augmentation of meningeal lymphatic functions. Biomed. Opt. Express 2020, 11, 725–734. [Google Scholar] [CrossRef]
  154. Semyachkina-Glushkovskaya, O.; Fedosov, I.; Shirokov, A.; Vodovozov, E.; Alekseev, A.; Khorovodov, A.; Blokhina, I.; Ter-skov, A.; Mamedova, A.; Klimova, M.; et al. Photomodulation of lymphatic delivery of liposomes to the brain bypassing the blood-brain barrier: New perspectives for glioma therapy. Nanophotonics 2021, 12, 3215–3227. [Google Scholar] [CrossRef]
  155. Semyachkina-Glushkovskaya, O.; Diduk, S.; Anna, E.; Elina, D.; Artem, K.; Khorovodov, A.; Shirokov, A.; Fedosov, I.; Du-brovsky, A.; Blokhina, I.; et al. Photomodulation of Lymphatic Delivery of Bevacizumab to the Brain: The Role of Singlet Oxygen. Adv. Exp. Med. Biol. 2022, 1395, 53–57. [Google Scholar] [PubMed]
  156. Semyachkina-Glushkovskaya, O.; Shirokov, A.; Blokhina, I.; Telnova, V.; Vodovozova, E.; Alekseeva, A.; Boldyrev, I.; Fedosov, I.; Dubrovsky, A.; Khorovodov, A.; et al. Intranasal delivery of liposomes to glioblastoma by photostimulation of the lymphatic system. Pharmaceutics 2023, 15, 36. [Google Scholar] [CrossRef] [PubMed]
  157. Semyachkina-Glushkovskaya, O.; Fedosov, I.; Zaikin, A.; Ageev, V.; Ilyukov, E.; Myagkov, D.; Tuktarov, D.; Blokhina, I.; Shirokov, A.; Terskov, A.; et al. Technology of the photobiostimulation of the brain’s drainage system during sleep for improvement of learning and memory in male mice. Biomed. Opt. Express 2023, 15, 44–58. [Google Scholar] [CrossRef] [PubMed]
  158. LIA The Laser Institute. Available online: https://www.lia.org/resources/laser-safety-information/laser-safety-standards/ansi-z136-standards/z136-3 (accessed on 28 April 2024).
  159. Webstore International Electrotechnical Commission. IEC TR 60825. Available online: https://webstore.iec.ch/publication/63122 (accessed on 28 April 2024).
  160. Fehervari, Z. Brain lymphatic (dys)function. Nat. Immunol. 2018, 19, 901. [Google Scholar] [CrossRef] [PubMed]
  161. Jiang, H.; Wei, H.; Zhou, Y. Overview of the meningeal lymphatic vessels in aging and central nervous system disorders. Cell Biosci. 2022, 12, 202. [Google Scholar] [CrossRef] [PubMed]
  162. Li, D.; Liu, S.; Yu, T.-T.; Liu, Z.; Sub, S.; Bragin, D.; Shirokov, A.; Navolokin, N.; Bragina, O.; Zheng-Wu, H.; et al. Photostimulation of brain lymphatics in male newborn and adult rodents for therapy of intraventricular hemorrhage. Nat. Commun. 2023, 14, 6104. [Google Scholar] [CrossRef] [PubMed]
  163. Bohlen, H.; Gasheva, O.; Zawieja, D. Nitric oxide formation by lymphatic bulb and valves is a major regulatory component of lymphatic pumping. Am. J. Physiol. Heart Circ. Physiol. 2011, 301, H1897–H1906. [Google Scholar] [CrossRef]
  164. Bohlen, H.; Wang, W.; Gashev, A.; Gasheva, O.; Zawieja, D. Phasic contractions of rat mesenteric lymphatics increase basal and phasic nitric oxide generation in vivo. Am. J. Physiol. Heart Circ. Physiol. 2009, 297, H1319–H1328. [Google Scholar] [CrossRef]
  165. Kunert, C.; Baish, J.W.; Liao, S.; Padera, T.P.; Munn, L.L. Mechanobiological oscillators control lymph flow. Proc. Natl. Acad. Sci. USA 2015, 112, 10938–10943. [Google Scholar] [CrossRef]
  166. Lordan, R.; Tsoupras, A.; Zabetakis, I. Phospholipids of Animal and Marine Origin: Structure, Function, and Anti-Inflammatory Properties. Molecules 2017, 22, 1964. [Google Scholar] [CrossRef] [PubMed]
  167. Ahmmed, M.K.; Ahmmed, F.; Tian, H.S.; Carne, A.; Bekhit, A.E. Marine omega-3 (n-3) phospholipids: A comprehensive review of their properties, sources, bioavailability, and relation to brain health. Compr. Rev. Food Sci. Food Saf. 2020, 19, 64–123. [Google Scholar] [CrossRef] [PubMed]
  168. Gomes, R.; Mendes, I.; Duarte, M.P.; Bandarra, N.M.; Gomes-Bispo, A. New Forms of Neuroactive Phospholipids for DHA Enrichment in Brain. Mar. Drugs 2024, 22, 116. [Google Scholar] [CrossRef]
  169. Ahmmed, M.K.; Hachem, M.; Ahmmed, F.; Rashidinejad, A.; Oz, F.; Bekhit, A.A.; Carne, A.; Bekhit, A.E.A. Marine Fish-Derived Lysophosphatidylcholine: Properties, Extraction, Quantification, and Brain Health Application. Molecules 2023, 28, 3088. [Google Scholar] [CrossRef]
  170. Bennett, S.A.; Valenzuela, N.; Xu, H.; Franko, B.; Fai, S.; Figeys, D. Using neurolipidomics to identify phospholipid mediators of synaptic (dys)function in Alzheimer’s Disease. Front. Physiol. 2013, 4, 168. [Google Scholar] [CrossRef]
  171. Pelucchi, S.; Gardoni, F.; Di Luca, M.; Marcello, E. Synaptic dysfunction in early phases of Alzheimer’s Disease. Handb. Clin. Neurol. 2022, 184, 417–438. [Google Scholar] [PubMed]
  172. Meftah, S.; Gan, J. Alzheimer’s disease as a synaptopathy: Evidence for dysfunction of synapses during disease progression. Front. Synaptic Neurosci. 2023, 15, 1129036. [Google Scholar] [CrossRef]
  173. Li, J.; Huang, X.; An, Y.; Chen, X.; Chen, Y.; Xu, M.; Shan, H.; Zhang, M. The role of snapin in regulation of brain homeostasis. Neural Regen. Res. 2024, 19, 1696–1701. [Google Scholar] [CrossRef] [PubMed]
  174. Senanayake, V.; Goodenowe, D.B. Plasmalogen deficiency and neuropathology in Alzheimer’s disease: Causation or coincidence? Alzheimer’s Dement. Transl. Res. Clin. Interv. 2019, 5, 524–532. [Google Scholar] [CrossRef]
  175. Su, X.Q.; Wang, J.; Sinclair, A.J. Plasmalogens and Alzheimer’s disease: A review. Lipids Health Dis. 2019, 18, 100. [Google Scholar] [CrossRef]
  176. Yamashita, S.; Miyazawa, T.; Higuchi, O.; Kinoshita, M.; Miyazawa, T. Marine Plasmalogens: A Gift from the Sea with Benefits for Age-Associated Diseases. Molecules 2023, 28, 6328. [Google Scholar] [CrossRef] [PubMed]
  177. Azad, A.K.; Sheikh, A.M.; Haque, M.A.; Osago, H.; Sakai, H.; Shibly, A.Z.; Yano, S.; Michikawa, M.; Hossain, S.; Tabassum, S.; et al. Time-Dependent Analysis of Plasmalogens in the Hippocampus of an Alzheimer’s Disease Mouse Model: A Role of Ethanolamine Plasmalogen. Brain Sci. 2021, 11, 1603. [Google Scholar] [CrossRef]
  178. Hossain, M.S.; Mawatari, S.; Fujino, T. Biological Functions of Plasmalogens. Adv. Exp. Med. Biol. 2020, 1299, 171–193. [Google Scholar] [PubMed]
  179. Honsho, M.; Fujiki, Y. Asymmetric Distribution of Plasmalogens and Their Roles-A Mini Review. Membranes 2023, 13, 764. [Google Scholar] [CrossRef] [PubMed]
  180. West, A.; Zoni, V.; Teague, W.E.; Leonard, A.N.; Vanni, S.; Gawrisch, K.; Tristram-Nagle, S.; Sachs, J.N.; Klauda, J.B. How do ethanolamine plasmalogens contribute to order and structure of neurological membranes? J. Phys. Chem. B 2020, 124, 828–839. [Google Scholar] [CrossRef] [PubMed]
  181. Bozelli, J.C., Jr.; Azher, S.; Epand, R.M. Plasmalogens and Chronic Inflammatory Diseases. Front. Physiol. 2021, 12, 730829. [Google Scholar] [CrossRef] [PubMed]
  182. Kytikova, O.Y.; Novgorodtseva, T.P.; Antonyuk, M.V.; Gvozdenko, T.A. Plasmalogens in the Pathophysiology and Therapy of Age-Specific Diseases. Adv. Gerontol. 2020, 10, 272–281. [Google Scholar]
  183. Kling, M.A.; Goodenowe, D.B.; Senanayake, V.; Mahmoudian Dehkordi, S. Circulating ethanolamine plasmalogen indices in Alzheimer’s disease: Relation to diagnosis, cognition, and CSF tau. Alzheimer’s Dement. 2020, 16, 1234–1247. [Google Scholar] [CrossRef] [PubMed]
  184. Goodenowe, D. Breaking Alzheimer’s—A 15 Year Crusade to Expose the Cause and Deliver the Cure; Amazon Digital Services LLC—Kdp: Seattle, WA, USA, 2021; p. 244. [Google Scholar]
  185. Rouser, G.; Yamamoto, A. Curvilinear regression course of human brain lipid composition changes with age. Lipids 1968, 3, 284–287. [Google Scholar] [CrossRef]
  186. Goodenowe, D.B.; Haroon, J.; Kling, M.A.; Zielinski, M.; Mahdavi, K.; Habelhah, B.; Shtilkind, L.; Jordan, S. Targeted Plasmalogen Supplementation: Effects on Blood Plasmalogens, Oxidative Stress Biomarkers, Cognition, and Mobility in Cognitively Impaired Persons. Front. Cell. Dev. Biol. 2022, 10, 864842. [Google Scholar] [CrossRef]
  187. Hugo, J.; Ganguli, M. Dementia and cognitive impairment: Epidemiology, diagnosis, and treatment. Clin. Geriatr. Med. 2014, 30, 421–442. [Google Scholar] [CrossRef] [PubMed]
  188. Vermunt, L.; Sikkes, S.A.M.; van den Hout, A.; Handels, R.; Bos, I.; van der Flier, W.M.; Kern, S.; Ousset, P.J.; Maruff, P.; Skoog, I.; et al. Duration of preclinical, prodromal, and dementia stages of Alzheimer’s disease in relation to age, sex, and APOE genotype. Alzheimer’s Dement. 2019, 15, 888–898. [Google Scholar] [CrossRef] [PubMed]
  189. Wilson, R.S.; Yu, L.; Schneider, J.A.; Bennett, D.A.; Boyle, P.A. Risk Aversion and Alzheimer Disease in Old Age. Am. J. Geriatr. Psychiatry 2019, 27, 851–861. [Google Scholar] [CrossRef] [PubMed]
  190. Meletis, C.D. Alkyl-Acylglycerols and the Important Clinical Ramifications of Raising Plasmalogens in Dementia and Alzheimer’s Disease. Integr. Med. 2020, 19, 12e–16e. [Google Scholar]
  191. Jenkins, C.M.; Yang, K.; Liu, G.; Moon, S.H.; Dilthey, B.G.; Gross, R.W. Cytochrome c is an oxidative stress-activated plasmalogenase that cleaves plasmenylcholine and plasmenylethanolamine at the sn-1 vinyl ether linkage. J. Biol. Chem. 2018, 293, 8693–8709. [Google Scholar] [CrossRef] [PubMed]
  192. Dorninger, F.; Forss-Petter, S.; Wimmer, I.; Berger, J. Plasmalogens, platelet-activating factor and beyond—Ether lipids in signaling and neurodegeneration. Neurobiol. Dis. 2020, 145, 105061. [Google Scholar] [CrossRef] [PubMed]
  193. Braverman, N.E.; Moser, A.B. Functions of plasmalogen lipids in health and disease. Biochim. Biophys. Acta 2012, 1822, 1442–1452. [Google Scholar] [CrossRef] [PubMed]
  194. Sindona, C.; Schepici, G.; Contestabile, V.; Bramanti, P.; Mazzon, E. NOX2 Activation in COVID-19: Possible Implications for Neurodegenerative Diseases. Medicina 2021, 57, 604. [Google Scholar] [CrossRef] [PubMed]
  195. Wong, L.S.Y.; Loo, E.X.L.; Kang, A.Y.H.; Lau, H.X.; Tambyah, P.A.; Tham, E.H. Age-related differences in immunological responses to SARS-CoV-2. J. Allergy Clin. Immunol. Pract. 2020, 8, 3251–3258. [Google Scholar] [CrossRef]
  196. Feng, T.; Hu, X.; Fukui, Y.; Tadokoro, K.; Bian, Z.; Morihara, R.; Yamashita, T.; Abe, K. Neuroprotective effects of Scallop-derived plasmalogen in a mouse model of ischemic stroke. Brain Res. 2021, 1766, 147516. [Google Scholar] [CrossRef]
  197. Yamashita, S.; Hashimoto, M.; Haque, A.M.; Nakagawa, K.; Kinoshita, M.; Shido, O.; Miyazawa, T. Oral Administration of Ethanolamine Glycerophospholipid Containing a High Level of Plasmalogen Improves Memory Impairment in Amyloid beta-Infused Rats. Lipids 2017, 52, 575–585. [Google Scholar] [CrossRef]
  198. Hossain, M.S.; Mawatari, S.; Fujino, T. Plasmalogens inhibit neuroinflammation and promote cognitive function. Brain Res. Bull. 2023, 192, 56–61. [Google Scholar] [CrossRef]
  199. Hossain, M.S.; Tajima, A.; Kotoura, S.; Katafuchi, T. Oral ingestion of plasmalogens can attenuate the LPS-induced memory loss and microglial activation. Biochem. Biophys. Res. Commun. 2018, 496, 1033–1039. [Google Scholar] [CrossRef] [PubMed]
  200. Wood, P.L.; Mankidy, R.; Ritchie, S.; Heath, D.; Wood, J.A.; Flax, J.; Goodenowe, D.B. Circulating plasmalogen levels and Alzheimer disease Assessment Scale-Cognitive scores in Alzheimer patients. J. Psychiatry Neurosci. 2010, 35, 59–62. [Google Scholar] [CrossRef] [PubMed]
  201. Mawatari, S.; Ohara, S.; Taniwaki, Y.; Tsuboi, Y.; Maruyama, T.; Fujino, T. Improvement of Blood Plasmalogens and Clinical Symptoms in Parkinson’s Disease by Oral Administration of Ether Phospholipids: A Preliminary Report. Park. Dis. 2020, 2020, 2671070. [Google Scholar] [CrossRef]
  202. Fujino, T.; Yamada, T.; Mawatari, S.; Shinfuku, N.; Tsuboi, Y.; Wakana, C.; Kono, S. Effects of Plasmalogen on Patients with Moderate-to-Severe Alzheimer’s Disease and Blood Plasmalogen Changes: A Multi-Center, Open-Label Study. J. Alzheimer’s Dis. Park. 2019, 9, 474. [Google Scholar]
  203. Yamamoto, A.; Aizawa, T.; Kubomura, D.; Akahori, Y.; Yamashita, S.; Nakagawa, K.; Miyazawa, T. Effects of Ascidian-Derived Ethanolamine Plasmalogen on Cognitive Function and Its Safety—A Randomized, Double-Blind, Placebo-Controlled, Parallel-Group Study. Pharmacometrics 2023, 104, 17–24. [Google Scholar]
  204. Watanabe, H.; Okawara, M.; Matahira, Y.; Mano, T.; Wada, T.; Suzuki, N.; Takara, T. The Impact of Ascidian (Halocynthia roretzi)-derived Plasmalogen on Cognitive Function in Healthy Humans: A Randomized, Double-blind, Placebo-controlled Trial. J. Oleo Sci. 2020, 69, 1597–1607. [Google Scholar] [CrossRef]
  205. Kawamura, J.; Kotoura, S.; Ando, T.; Kawasaki, Y.; Ebihara, S. The Evaluation Test of Brain Function by Oral Consumption of the Food Which Contain Plasmalogen—Randomized, Placebo-controlled, Double-blind Parallel-group Study. Jpn. Pharmacol. Ther. 2019, 47, 739–749. [Google Scholar]
  206. Olivera-Pueyo, J.; Pelegrin-Valero, C. Dietary supplements for cognitive impairment. Actas Esp. Psiquiatr. 2017, 45, 37–47. [Google Scholar]
  207. Liu, Y.; Cong, P.; Zhang, T.; Wang, R.; Wang, X.; Liu, J.; Wang, X.; Xu, J.; Wang, Y.; Wang, J.; et al. Plasmalogen attenuates the development of hepatic steatosis and cognitive deficit through mechanism involving p75NTR inhibition. Redox Biol. 2021, 43, 102002. [Google Scholar] [CrossRef] [PubMed]
  208. Hossain, M.S.; Mawatari, S.; Fujino, T. Plasmalogens, the Vinyl Ether-Linked Glycerophospholipids, Enhance Learning and Memory by Regulating Brain-Derived Neurotrophic Factor. Front. Cell Dev. Biol. 2022, 10, 828282. [Google Scholar] [CrossRef] [PubMed]
  209. Sejimo, S.; Hossain, M.S.; Akashi, K. Scallop-derived plasmalogens attenuate the activation of PKCδ associated with the brain inflammation. Biochem. Biophys. Res. Commun. 2018, 503, 837–842. [Google Scholar] [CrossRef]
  210. Che, H.; Li, Q.; Zhang, T.; Ding, L.; Zhang, L.; Shi, H.; Yanagita, T.; Xue, C.; Chang, Y.; Wang, Y. A comparative study of EPA-enriched ethanolamine plasmalogen and EPA-enriched phosphatidylethanolamine on Aβ42 induced cognitive deficiency in a rat model of Alzheimer’s disease. Food Funct. 2018, 9, 3008–3017. [Google Scholar] [CrossRef] [PubMed]
  211. Ifuku, M.; Katafuchi, T.; Mawatari, S.; Noda, M.; Miake, K.; Sugiyama, M.; Fujino, T. Anti-inflammatory/anti-amyloidogenic effects of plasmalogens in lipopolysaccharide-induced neuroinflammation in adult mice. J. Neuroinflamm. 2012, 9, 197. [Google Scholar] [CrossRef]
  212. Yamashita, S.; Kiko, T.; Fujiwara, H.; Hashimoto, M.; Nakagawa, K.; Kinoshita, M.; Furukawa, K.; Arai, H.; Miyazawa, T. Alterations in the Levels of Amyloid-beta, Phospholipid Hydroperoxide, and Plasmalogen in the Blood of Patients with Alzheimer’s Disease: Possible Interactions between Amyloid-beta and These Lipids. J. Alzheimer’s Dis. 2016, 50, 527–537. [Google Scholar] [CrossRef] [PubMed]
  213. Youssef, M.; Ibrahim, A.; Akashi, K.; Hossain, M.S. PUFA-Plasmalogens Attenuate the LPS-Induced Nitric Oxide Production by Inhibiting the NF-kB, p38 MAPK and JNK Pathways in Microglial Cells. Neuroscience 2019, 397, 18–30. [Google Scholar] [CrossRef] [PubMed]
  214. Onodera, T.; Futai, E.; Kan, E.; Abe, N.; Uchida, T.; Kamio, Y.; Kaneko, J. Phosphatidylethanolamine plasmalogen enhances the inhibiting effect of phosphatidylethanolamine on γ-secretase activity. J. Biochem. 2014, 157, 301–309. [Google Scholar] [CrossRef]
  215. Miyazawa, T.; Itaya, M.; Burdeos, G.C.; Nakagawa, K.; Miyazawa, T. A Critical Review of the Use of Surfactant-Coated Nanoparticles in Nanomedicine and Food Nanotechnology. Int. J. Nanomed. 2021, 16, 3937–3999. [Google Scholar] [CrossRef]
  216. Hino, K.; Kaneko, S.; Harasawa, T.; Kimura, T.; Takei, S.; Shinohara, M.; Yamazaki, F.; Morita, S.-y.; Sato, S.; Kubo, Y.; et al. Change in Brain Plasmalogen Composition by Exposure to Prenatal Undernutrition Leads to Behavioral Impairment of Rats. J. Neurosci. 2019, 39, 7689–7702. [Google Scholar] [CrossRef]
  217. Katafuchi, T.; Ifuku, M.; Mawatari, S.; Noda, M.; Miake, K.; Sugiyama, M.; Fujino, T. Effects of plasmalogens on systemic lipopolysaccharide-induced glial activation and β-amyloid accumulation in adult mice. Ann. N. Y. Acad. Sci. 2012, 1262, 85–92. [Google Scholar] [CrossRef] [PubMed]
  218. Rothhaar, T.L.; Grösgen, S.; Haupenthal, V.J.; Burg, V.K.; Hundsdörfer, B.; Mett, J.; Riemenschneider, M.; Grimm, H.S.; Hartmann, T.; Grimm, M.O. Plasmalogens inhibit APP processing by directly affecting γ-secretase activity in Alzheimer’s disease. Sci. World J. 2012, 2012, 141240. [Google Scholar] [CrossRef] [PubMed]
  219. Conti Filho, C.E.; Loss, L.B.; Marcolongo-Pereira, C.; Rossoni Junior, J.V.; Barcelos, R.M.; Chiarelli-Neto, O.; da Silva, B.S.; Passamani Ambrosio, R.; Castro, F.C.A.Q.; Teixeira, S.F.; et al. Advances in Alzheimer’s disease’s pharmacological treatment. Front. Pharmacol. 2023, 14, 1101452. [Google Scholar] [CrossRef] [PubMed]
  220. Marucci, G.; Buccioni, M.; Ben, D.D.; Lambertucci, C.; Volpini, R.; Amenta, F. Efficacy of acetylcholinesterase inhibitors in Alzheimer’s disease. Neuropharmacology 2021, 190, 108352. [Google Scholar] [CrossRef] [PubMed]
  221. Peng, Y.; Jin, H.; Xue, Y.; Chen, Q.; Yao, S.; Du, M.; Liu, S. Current and future therapeutic strategies for Alzheimer’s disease: An overview of drug development bottlenecks. Front. Aging Neurosci. 2023, 15, 1206572. [Google Scholar] [CrossRef] [PubMed]
  222. 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, 10284, 1577–1590. [Google Scholar] [CrossRef] [PubMed]
  223. Botchway, B.O.A.; Moore, M.K.; Akinleye, F.O.; Iyer, I.C.; Fang, M. Nutrition: Review on the possible treatment for Alzheimer’s disease. J. Alzheimer’s Dis. 2018, 3, 867–883. [Google Scholar] [CrossRef] [PubMed]
  224. Mafi, J.N.; Leng, M.; Arbanas, J.C.; Tseng, C.H.; Damberg, C.L.; Sarkisian, C.; Landon, B.E. Estimated annual spending on aducanumab in the US medicare program. JAMA Health Forum 2022, 1, e214495. [Google Scholar] [CrossRef] [PubMed]
  225. Biogen. Biogen Plans Regulatory Filing for Aducanumab in Alzheimer’s Disease Based on New Analysis of Larger Dataset from Phase 3 Studies. Available online: http://investors.biogen.com/news-releases/news-release-details/biogen-plans-regulatory-filing-aducanumab-alzheimers-disease (accessed on 25 April 2024).
  226. Wang, X.; Sun, G.; Feng, T.; Zhang, J.; Huang, X.; Wang, T.; Xie, Z.; Chu, X.; Yang, J.; Wang, H.; et al. Sodium oligomannate therapeutically remodels gut microbiota and suppresses gut bacterial amino acids-shaped neuroinflammation to inhibit Alzheimer’s disease progression. Cell Res. 2019, 29, 787–803. [Google Scholar] [CrossRef]
  227. The Lancet. Lecanemab for Alzheimer’s disease: Tempering hype and hope. Lancet 2022, 400, 1899. [Google Scholar] [CrossRef]
  228. Silva, T.; Reis, J.; Teixeira, J.; Borges, F. Alzheimer’s disease, enzyme targets and drug discovery struggles: From natural products to drug prototypes. Ageing Res. Rev. 2014, 15, 116–145. [Google Scholar] [CrossRef] [PubMed]
  229. Huang, J.; Huang, N.; Mao, Q.; Shi, J.; Qiu, Y. Natural bioactive compounds in Alzheimer’s disease: From the perspective of type 3 diabetes mellitus. Front. Aging Neurosci. 2023, 15, 1130253. [Google Scholar] [CrossRef] [PubMed]
  230. Andrade, S.; Ramalho, M.J.; Loureiro, J.A.; Pereira, M.d.C. Natural Compounds for Alzheimer’s Disease Therapy: A Systematic Review of Preclinical and Clinical Studies. Int. J. Mol. Sci. 2019, 20, 2313. [Google Scholar] [CrossRef] [PubMed]
  231. Firdaus, Z.; Singh, T.D. An Insight in Pathophysiological Mechanism of Alzheimer’s Disease and its Management Using Plant Natural Products. Mini Rev. Med. Chem. 2021, 21, 35–57. [Google Scholar] [CrossRef]
  232. Nagori, K.; Nakhate, K.T.; Yadav, K.; Ajazuddin; Pradhan, M. Unlocking the Therapeutic Potential of Medicinal Plants for Alzheimer’s Disease: Preclinical to Clinical Trial Insights. Future Pharmacol. 2023, 3, 877–907. [Google Scholar] [CrossRef]
  233. Singh, A.K.; Gupta, A.; Mishra, A.K.; Gupta, V.; Bansal, P.; Kumar, S. Medicinal Plant for Curing Alzheimer’s Disease. Int. J. Pharm. Biol. Sci. Arch. 2010, 1, 108–114. [Google Scholar]
  234. Hatab, H.M.; Abdel Hamid, F.F.; Soliman, A.F.; Al-Shafie, T.A.; Ismail, Y.M.; El-Houseini, M.E. A combined treatment of curcumin, piperine, and taurine alters the circulating levels of IL-10 and miR-21 in hepatocellular carcinoma patients: A pilot study. J. Gastrointest. Oncol. 2019, 10, 766–776. [Google Scholar] [CrossRef] [PubMed]
  235. Kotha, R.R.; Luthria, D.L. Curcumin: Biological, pharmaceutical, nutraceutical, and analytical aspects. Molecules 2019, 24, 2930. [Google Scholar] [CrossRef] [PubMed]
  236. Kumar, A.; Singh, A.; Aggarwal, A. Therapeutic potentials of herbal drugs for Alzheimer’s disease—An overview. Indian J. Exp. Biol. 2017, 55, 63–73. [Google Scholar]
  237. Rane, J.S.; Bhaumik, P.; Panda, D. Curcumin inhibits tau aggregation and disintegrates preformed tau filaments in vitro. J. Alzheimer’s Dis. 2017, 60, 999–1014. [Google Scholar] [CrossRef]
  238. Voulgaropoulou, S.D.; van Amelsvoort, T.A.M.J.; Prickaerts, J.; Vingerhoets, C. The effect of curcumin on cognition in Alzheimer’s disease and healthy aging: A systematic review of pre-clinical and clinical studies. Brain Res. 2019, 1725, 146476. [Google Scholar] [CrossRef] [PubMed]
  239. Mishra, S.; Palanivelu, K. The effect of curcumin (turmeric) on Alzheimer’s disease: An overview. Ann. Indian Acad. Neurol. 2008, 11, 13–19. [Google Scholar] [CrossRef]
  240. Kesarwani, K.; Gupta, R. Bioavailability enhancers of herbal origin: An overview. Asian Pac. J. Trop. Biomed. 2013, 3, 253–266. [Google Scholar] [CrossRef]
  241. Prasad, S.; Tyagi, A.K.; Aggarwal, B.B. Recent developments in delivery, bioavailability, absorption and metabolism of curcumin: The golden pigment from golden spice. Cancer Res. Treat. 2014, 46, 2–18. [Google Scholar] [CrossRef] [PubMed]
  242. Shoba, G.; Joy, D.; Joseph, T.; Majeed, M.; Rajendran, R.; Srinivas, P. Influence of piperine on the pharmacokinetics of curcumin in animals and human volunteers. Planta Med. 1998, 64, 353–356. [Google Scholar] [CrossRef]
  243. Ahmad, S.; Hafeez, A. Formulation and Development of Curcumin–Piperine-Loaded S-SNEDDS for the Treatment of Alzheimer’s Disease. Mol. Neurobiol. 2023, 60, 1067–1082. [Google Scholar] [CrossRef] [PubMed]
  244. Canevelli, M.; Adali, N.; Kelaiditi, E.; Cantet, C.; Ousset, P.J.; Cesari, M. Efects of Gingko biloba supplementation in Alzheimer disease patients receiving cholinesterase inhibitors: Data from the ICTUS study. Phytomedicine 2014, 21, 888–892. [Google Scholar] [CrossRef] [PubMed]
  245. Nowak, A.; Kojder, K.; Zielonka-Brzezicka, J.; Wróbel, J.; Bosiacki, M.; Fabiańska, M.; Wróbel, M.; Sołek-Pastuszka, J.; Klimowicz, A. The use of Ginkgo biloba as a neuroprotective agent in the Alzheimer disease. Front. Pharmacol. 2021, 12, 775034. [Google Scholar] [CrossRef]
  246. Xie, L.; Zhu, Q.; Lu, J. Can we use Ginkgo biloba extract to treat Alzheimer disease? Lessons from preclinical and clinical studies. Cells 2022, 11, 479. [Google Scholar] [CrossRef]
  247. Oken, B.S.; Storzbach, D.M.; Kaye, J.A. The efficacy of Ginkgo biloba on cognitive function in Alzheimer disease. Arch. Neurol. 1998, 55, 1409–1415. [Google Scholar] [CrossRef]
  248. Dubey, T.; Chinnathambi, S. Brahmi (Bacopa monnieri): An ayurvedic herb against the Alzheimer disease. Arch. Biochem. Biophys. 2019, 676, 108153. [Google Scholar] [CrossRef] [PubMed]
  249. Lopresti, A.L. Salvia (Sage): A review of its potential cognitive-enhancing and protective effects. Drugs R&D 2017, 17, 53–64. [Google Scholar]
  250. Miraj, S.; Kiani, S. A review study of therapeutic effects of Salvia officanalis L. Pharm Lett. 2016, 8, 299–303. [Google Scholar]
  251. Datta, S.; Patil, S. Evaluation of traditional herb extract Salvia officanalis in treatment of Alzheimer disease. Pharmacogn. J. 2020, 12, 131–143. [Google Scholar] [CrossRef]
  252. Akhondzadeh, S.; Noroozian, M.; Mohammadi, M.; Ohadinia, S.; Jamshidi, A.H.; Khani, M. Melissa offcinalis extract in the treatment of patients with mild to moderate Alzheimer disease: A double blind, randomised, placebo controlled trial. J. Neurol. Neurosurg. Psychiatry 2003, 74, 863–866. [Google Scholar] [CrossRef] [PubMed]
  253. John, O.O.; Ihim, S.A.; Agbo, C.P.; Echezona, A. Phytotherapy: A promising approach for the treatment of Alzheimer disease. Pharmacol. Res. Mod. Chin. Med. 2022, 2, 100030. [Google Scholar] [CrossRef]
  254. López, V.; Martín, S.; Gómez-Serranillos, M.P.; Carretero, M.E.; Jäger, A.K.; Calvo, M.I. Neuroprotective and neurological properties of Melissa officinalis. Neurochem. Res. 2009, 34, 1955–1961. [Google Scholar] [CrossRef] [PubMed]
  255. Obulesu, M.; Rao, D.M. Effect of plant extracts on Alzheimer disease: An insight into therapeutic avenues. J. Neurosci. Rural Pract. 2011, 2, 56–61. [Google Scholar]
  256. Miraj, S.; Azizi, N.; Kiani, S. A review of chemical components and pharmacological effects of Melissa officinalis L. Pharm. Lett. 2016, 8, 229–237. [Google Scholar]
  257. Beheshti, S.; Shahmoradi, B. Therapeutic effect of Melissa offcinalis in an amyloid-β rat model of Alzheimer disease. J. Herbmed Pharmacol. 2018, 7, 193–199. [Google Scholar] [CrossRef]
  258. Mahboubi, M. Melissa offcinalis and rosmarinic acid in management of memory functions and Alzheimer disease. Asian Pac. J. Trop. Biomed. 2019, 9, 47–52. [Google Scholar] [CrossRef]
  259. Friedli, M.J.; Inestrosa, N.C. Huperzine A and Its Neuroprotective Molecular Signaling in Alzheimer’s Disease. Molecules 2021, 26, 6531. [Google Scholar] [CrossRef] [PubMed]
  260. Callizot, N.; Campanari, M.L.; Rouvière, L.; Jacquemot, G.; Henriques, A.; Garayev, E.; Poindron, P. Huperzia serrata Extract ‘NSP01’ with Neuroprotective Effects-Potential Synergies of Huperzine A and Polyphenols. Front. Pharmacol. 2021, 12, 681532. [Google Scholar] [CrossRef] [PubMed]
  261. Wen, Y.R.; Yang, J.H.; Wang, X.; Yao, Z.B. Induced dural lymphangiogenesis facilities soluble amyloid-beta clearance from brain in a transgenic mouse model of Alzheimer’s disease. Neural Regen. Res. 2018, 13, 709–716. [Google Scholar] [PubMed]
  262. Sheikhi-Mobarakeh, Z.; Yarmohammadi, H.; Mokhatri-Hesari, P.; Fahimi, S.; Montazeri, A.; Heydarirad, G. Herbs as old potential treatments for lymphedema management: A systematic review. Complement. Ther. Med. 2020, 55, 102615. [Google Scholar] [CrossRef] [PubMed]
  263. Peng, L.; Dong, Y.; Fan, H.; Cao, M.; Wu, Q.; Wang, Y.; Zhou, C.; Li, S.; Zhao, C.; Wang, Y. Traditional Chinese Medicine Regulating Lymphangiogenesis: A Literature Review. Front. Pharmacol. 2020, 11, 1259. [Google Scholar] [CrossRef] [PubMed]
  264. Cacchio, A.; Prencipe, R.; Bertone, M.; De Benedictis, L.; Taglieri, L.; D’Elia, E.; Centoletti, C.; Di Carlo, G. Effectiveness and safety of a product containing diosmin, coumarin, and arbutin (Linfadren®) in addition to complex decongestive therapy on management of breast cancer-related lymphedema. Support. Care Cancer 2019, 27, 1471–1480. [Google Scholar] [CrossRef] [PubMed]
  265. Yu, J.; Mao, L.; Guan, L.; Zhang, Y.; Zhao, J. Ginsenoside Rg1 enhances lymphatic transport of intrapulmonary silica via VEGF-C/VEGFR-3 signaling in silicotic rats. Biochem. Biophys. Res. Commun. 2016, 472, 182–188. [Google Scholar] [CrossRef] [PubMed]
  266. Zhang, X.G.; Shan, C.; Zhu, J.Z.; Bao, X.Y.; Tong, Q.; Wu, X.F.; Tang, X.C.; Xue, T.; Liu, J.; Zheng, G.Q.; et al. Additive Neuroprotective Effect of Borneol with Mesenchymal Stem Cells on Ischemic Stroke in Mice. Front. Physiol. 2018, 8, 1133. [Google Scholar] [CrossRef]
  267. Li, W.R.; Chen, R.Y.; Yang, L.; Huang, T.L.; Xu, Q.W.; Mi, S.Q.; Wang, N.S. Pharmacokinetics of natural borneol after oral administration in mice brain and its effect on excitation ratio. Eur. J. Drug Metab. Pharmacokinet. 2012, 37, 39–44. [Google Scholar] [CrossRef]
  268. Yu, B.; Ruan, M.; Cui, X.B.; Guo, J.M.; Xu, L.; Dong, X.P. Effects of borneol on the pharmacokinetics of geniposide in cortex, hippocampus, hypothalamus and striatum of conscious rat by simultaneous brain microdialysis coupled with UPLC-MS. J. Pharm. Biomed. Anal. 2013, 77, 128–132. [Google Scholar] [CrossRef] [PubMed]
  269. Zhang, Q.L.; Fu, B.M.; Zhang, Z.L. Borneol, a novel agent that improves central nervous system drug delivery by enhancing blood–brain barrier permeability. Drug Deliv. 2017, 24, 1037–1044. [Google Scholar] [CrossRef] [PubMed]
  270. Tambe, R.F.; Jain, P.K.; Patil, S.; Ghumatkar, P.; Sathaye, S. Antiepileptogenic effects of borneol in pentylenetetrazole-induced kindling in mice. Naunyn. Schmiedebergs Arch. Pharmacol. 2016, 389, 467–475. [Google Scholar] [CrossRef] [PubMed]
  271. Hu, W.H.; Mak, H.; Zheng, Z.Y.; Xia, Y.J.; Xu, M.L.; Duan, R.; Dong, T.T.-X.; Li, S.-P.; Zhan, C.-S.; Shang, X.-H.; et al. Shexiang Baoxin Pill, a Traditional Chinese Herbal Formula, Rescues the Cognitive Impairments in APP/PS1 Transgenic Mice. Front. Pharmacol. 2020, 11, 1045. [Google Scholar] [CrossRef] [PubMed]
  272. Dong, T.W.; Chen, N.; Ma, X.; Wang, J.; Wen, J.; Xie, Q.; Ma, R. The protective roles of L-borneolum, D-borneolum and synthetic borneol in cerebral ischaemia via modulation of the neurovascular unit. Biomed. Pharmacother. 2018, 102, 874–883. [Google Scholar] [CrossRef] [PubMed]
  273. Ye, T.T.; Wu, Y.; Shang, L.; Deng, X.Q.; Wang, S.J. Improved lymphatic targeting: Effect and mechanism of synthetic borneol on lymph node uptake of 7-ethyl-10-hydroxycamptothecin nanoliposomes following subcutaneous administration. Drug Deliv. 2018, 25, 1461–1471. [Google Scholar] [CrossRef] [PubMed]
  274. Wu, Y.; Zhang, T.; Li, X.; Wei, Y.; Li, X.; Wang, S.; Liu, J.; Li, D.; Wang, S.; Ye, T. Borneol-driven meningeal lymphatic drainage clears amyloid-β peptide to attenuate Alzheimer-like phenotype in mice. Theranostics 2023, 13, 106–124. [Google Scholar] [CrossRef]
  275. Ding, X.; Wang, X.; Xia, D.; Liu, H.; Tian, H.; Fu, Y.; Chen, Y.; Qin, C.; Wang, J.; Xiang, Z.; et al. Impaired meningeal lymphatic drainage in patients with idiopathic Parkinson’s disease. Nat. Med. 2021, 27, 411–418. [Google Scholar] [CrossRef]
  276. Bolte, A.C.; Dutta, A.B.; Hurt, M.E. Meningeal lymphatic dysfunction exacerbates traumatic brain injury pathogenesis. Nat. Commun. 2020, 11, 4524. [Google Scholar] [CrossRef]
  277. Xie, L.; Kang, H.; Xu, Q.; Chen, M.J.; Liao, Y.; Thiyagarajan, M.; O’Donnell, J.; Christensen, D.J.; Nicholson, C.; Iliff, J. Sleep drives metabolite clearance from the adult brain. Science 2013, 342, 373–377. [Google Scholar] [CrossRef]
  278. Fultz, N.E.; Bonmassar, G.; Setsompop, K.; Stickgold, R.; Rosen, B.; Polimeni, J.; Lewis, L. Coupled electrophysio-logical, hemodynamic, and cerebrospinal fluid oscillations in human sleep. Science 2019, 366, 628–631. [Google Scholar] [CrossRef] [PubMed]
  279. Shirokov, A.; Blokhina, I.; Fedosov, I.; Ilyukov, E.; Terskov, A.; Myagkov, D.; Tuktarov, D.; Tzoy, M.; Adushkina, V.; Zlatogosrkaya, D.; et al. Different Effects of Phototherapy for Rat Glioma during Sleep andWakefulness. Biomedicines 2024, 12, 262. [Google Scholar] [CrossRef] [PubMed]
  280. Sultanov, R.M.; Poleshchuk, T.S.; Ermolenko, E.V.; Kasyanov, S.P. Protective Properties of Marine Alkyl Glycerol Ethers in Chronic Stress. Mar. Drugs 2023, 21, 202. [Google Scholar] [CrossRef] [PubMed]
  281. Sultanov, R.; Ermolenko, E.; Poleshchuk, T.; Kasyanov, S. Alkyl Glycerol Ethers as Adaptogens. Mar. Drugs 2023, 21, 4. [Google Scholar] [CrossRef]
  282. Carvalho, D.; Knopman, D.; Boeve, D.; Lowe, V.; Roberts, R.; Mielke, M.; Przybelski, S.; Machulda, M.; Petersen, R.; Jack, C.; et al. association of excessive daytime sleepiness with longitudinal beta-amyloid accumulation in elderly persons without dementia. JAMA Neurol. 2018, 75, 672–680. [Google Scholar] [CrossRef]
  283. Sabia, S.; Fayosse, A.; Dumurgier, J.; Van Hees, V.; Paquet, C.; Sommerlad, A.; Kivimäki, M.; Dugravot, A.; Singh-Manoux, A. Association of sleep duration in middle and old age with incidence of dementia. Nat. Commun. 2021, 12, 2289. [Google Scholar] [CrossRef]
  284. Fessel, J. Analysis of Why Alzheimer’s Dementia Never Spontaneously Reverses, Suggests the Basis for Curative Treatment. J. Clin. Med. 2023, 12, 4873. [Google Scholar] [CrossRef]
  285. Li, Z.; Heckman, M.G.; Kanekiyo, T.; Martens, Y.A.; Day, G.S.; Vassilaki, M.; Liu, C.-C.; Bennett, D.A.; Petersen, R.C.; Zhao, N. Clinicopathologic Factors Associated with Reversion to Normal Cognition in Patients with Mild Cognitive Impairment. Neurology 2022, 98, e2036–e2045. [Google Scholar] [CrossRef] [PubMed]
  286. Pandya, S.Y.; Lacritz, L.H.; Weiner, M.F.; Deschner, M.; Woon, F.L. Predictors of reversion from mild cognitive impairment to normal cognition. Dement. Geriatr. Cogn. Disord. 2017, 43, 204–214. [Google Scholar] [CrossRef]
  287. Overton, M.; Pihlsgård, M.; Elmståhl, S. Diagnostic stability of mild cognitive impairment, and predictors of reversion to normal cognitive functioning. Dement. Geriatr. Cogn. Disord. 2020, 48, 317–329. [Google Scholar] [CrossRef]
  288. Malek-Ahmadi, M. Reversion from mild cognitive impairment to normal cognition. Alzheimer Dis. Assoc. Disord. 2016, 30, 324–330. [Google Scholar] [CrossRef] [PubMed]
  289. Iraniparast, M.; Shi, Y.; Wu, Y.; Zeng, L.; Maxwell, C.J.; Kryscio, R.J.; St John, P.D.; SantaCruz, K.S.; Tyas, S.L. Cognitive Reserve and Mild Cognitive Impairment: Predictors and Rates of Reversion to Intact Cognition vs. Progression to Dementia. Neurology 2022, 98, e1114–e1123. [Google Scholar] [CrossRef] [PubMed]
  290. Wells, C.; Brennan, S.; Keon, M.; Ooi, L. The role of amyloid oligomers in neurodegenerative pathologies. Int. J. Biol. Macromol. 2021, 181, 582–604. [Google Scholar] [CrossRef] [PubMed]
  291. Savage, M.; Trusko, S.; Howland, D.; Pinsker, L.; Mistretta, S.; Reaume, A.; Greenberg, B.; Siman, R.; Scott, R. Turnover of amyloid beta-protein in mouse brain and acute reduction of its level by phorbol ester. J. Neurosci. 1998, 18, 1743–1752. [Google Scholar] [CrossRef] [PubMed]
  292. Mezey, E.; Szalayova, I.; Hogden, C.; Brady, A.; Dosa, A.; Sotonui, P.; Palkovits, M. An immunohistochemical study of lymphatic elements in the human brain. Proc. Natl. Acad. Sci. USA 2021, 118, e2002574118. [Google Scholar] [CrossRef] [PubMed]
  293. Chang, J.; Guo, B.; Gao, Y.; Li, W.; Tong, X.; Feng, Y.; Abumaria, N. Characteristic Features of Deep Brain Lymphatic Vessels and Their Regulation by Chronic Stress. Research 2023, 6, 0120. [Google Scholar] [CrossRef] [PubMed]
  294. Prineas, L.W. Multiple sclerosis: Presence of lymphatic capillaries and lymphoid tissue in the brain and spinal cord. Science 1979, 203, 1123–1125. [Google Scholar] [CrossRef] [PubMed]
  295. Semyachkina-Glushkovskaya, O.; Fedosov, I.; Navolokin, N.; Shirokov, A.; Maslyakova, G.; Bucharskaya, A.; Blokhina, I.; Terskov, A.; Khorovodov, A.; Postnov, D.; et al. Pilot identification of the Live-1/Prox-1 expressing lymphatic vessels and lymphatic elements in the unaffected and affected human brain. bioRxiv 2021. [Google Scholar] [CrossRef]
  296. Ahn, J.; Cho, H.; Kim, J.; Kim, S.; Ham, J.; Park, I.; Suh, S.; Hong, S.; Song, J.; Hong, Y.; et al. Meningeal lymphatic vessels at the skull base drain cerebrospinal fluid. Nature 2019, 572, 62–66. [Google Scholar] [CrossRef]
  297. Louveau, A.; Smirnov, I.; Keyes, T.J.; Eccles, J.D.; Rouhani, S.J.; Peske, J.D.; Derecki, N.C.; Castle, D.; Mandell, J.W.; Lee, K.S.; et al. Structural and functional features of central nervous system lymphatic vessels. Nature 2015, 523, 337–341. [Google Scholar] [CrossRef]
  298. Zhinchenko, E.; Klimova, M.; Mamedova, A.; Agranovich, I.; Blokhina, I.; Antonova, T.; Terskov, A.; Shirokov, A.; Navolkin, N.; Morgun, A.; et al. Oxana Semyachkina-Glushkovskaya. Photostimulation of extravasation of beta-amyloid through the model of blood-brain barrier. Electronics 2020, 9, 1056. [Google Scholar] [CrossRef]
  299. Zhinchenko, E.; Navolokin, N.; Shirokov, A.; Khlebcov, B.; Dubrovsky, A.; Saranceva, E.; Abdurashitov, A.; Khorovodov, A.; Terskov, A.; Mamedova, A.; et al. Pilot study of transcranial photobiomodulation of lymphatic clearance of beta-amyloid from the mouse brain: Breakthrough strategies for nonpharmacologic therapy of Alzheimer’s disease. Biomed. Opt. Exp. 2019, 10, 4003–4017. [Google Scholar] [CrossRef] [PubMed]
  300. Semyachkina-Glushkovskaya, O.; Terskov, A.; Khorovodov, A.; Telnova, V.; Blokhina, I.; Saranceva, E.; Kurths, J. Photodynamic Opening of the Blood–Brain Barrier and the Meningeal Lymphatic System: The New Niche in Immunotherapy for Brain Tumors. Pharmaceutics 2022, 14, 2612. [Google Scholar] [CrossRef] [PubMed]
Figure 1. SARS-CoV-2 amyloid formation and the development of COVID-19-mediated AD dementia. SARS-CoV-2 causes respiratory infection associated with systemic inflammation and cytokine storms leading to BBB leakage. The SARS-CoV-2 virus can enter the brain through the damaged BBB and bind the ACE2 receptor. The SARS-CoV-2 virus can enter the hippocampus through invasion of the olfactory neurons. Once it enters the brain, the SARS-CoV-2 virus promotes the accumulation of amyloids both in the extracellular space due to the spike protein binding to Aβs, and intracellularly by forming the SARS-CoV-2 amyloids from the structural (the spikes, envelop, membrane and nucleocapsid) and the accessory (ORF6 and ORF10) proteins. These changes may lead to memory disorders in COVID-19 patients.
Figure 1. SARS-CoV-2 amyloid formation and the development of COVID-19-mediated AD dementia. SARS-CoV-2 causes respiratory infection associated with systemic inflammation and cytokine storms leading to BBB leakage. The SARS-CoV-2 virus can enter the brain through the damaged BBB and bind the ACE2 receptor. The SARS-CoV-2 virus can enter the hippocampus through invasion of the olfactory neurons. Once it enters the brain, the SARS-CoV-2 virus promotes the accumulation of amyloids both in the extracellular space due to the spike protein binding to Aβs, and intracellularly by forming the SARS-CoV-2 amyloids from the structural (the spikes, envelop, membrane and nucleocapsid) and the accessory (ORF6 and ORF10) proteins. These changes may lead to memory disorders in COVID-19 patients.
Pharmaceuticals 17 00788 g001
Figure 2. PBM-stimulation of lymphatic removal of the SARS-CoV-2 amyloids and virus from the brain: (a) Illustration of the effects of PBM on MLVs localized along the main venous sinuses, such as the Sagittal sinus (SS) and the transverse sinus (TS). (b,c) Schematic illustration of cooperation of NO and Ca2+ in regulation of lymphatic contraction and relaxation as well as the PBM-mediated stimulation of lymphatic removal of Aβ and the SARS-CoV-2 virus from the brain (detailed explanation in the text of the manuscript).
Figure 2. PBM-stimulation of lymphatic removal of the SARS-CoV-2 amyloids and virus from the brain: (a) Illustration of the effects of PBM on MLVs localized along the main venous sinuses, such as the Sagittal sinus (SS) and the transverse sinus (TS). (b,c) Schematic illustration of cooperation of NO and Ca2+ in regulation of lymphatic contraction and relaxation as well as the PBM-mediated stimulation of lymphatic removal of Aβ and the SARS-CoV-2 virus from the brain (detailed explanation in the text of the manuscript).
Pharmaceuticals 17 00788 g002
Figure 3. The beneficial effects of murine plasmalogens on cognitive function and AD.
Figure 3. The beneficial effects of murine plasmalogens on cognitive function and AD.
Pharmaceuticals 17 00788 g003
Figure 4. Schematic illustration of beneficial effects of promising plants for AD therapy.
Figure 4. Schematic illustration of beneficial effects of promising plants for AD therapy.
Pharmaceuticals 17 00788 g004
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

Navolokin, N.; Adushkina, V.; Zlatogorskaya, D.; Telnova, V.; Evsiukova, A.; Vodovozova, E.; Eroshova, A.; Dosadina, E.; Diduk, S.; Semyachkina-Glushkovskaya, O. Promising Strategies to Reduce the SARS-CoV-2 Amyloid Deposition in the Brain and Prevent COVID-19-Exacerbated Dementia and Alzheimer’s Disease. Pharmaceuticals 2024, 17, 788. https://doi.org/10.3390/ph17060788

AMA Style

Navolokin N, Adushkina V, Zlatogorskaya D, Telnova V, Evsiukova A, Vodovozova E, Eroshova A, Dosadina E, Diduk S, Semyachkina-Glushkovskaya O. Promising Strategies to Reduce the SARS-CoV-2 Amyloid Deposition in the Brain and Prevent COVID-19-Exacerbated Dementia and Alzheimer’s Disease. Pharmaceuticals. 2024; 17(6):788. https://doi.org/10.3390/ph17060788

Chicago/Turabian Style

Navolokin, Nikita, Viktoria Adushkina, Daria Zlatogorskaya, Valeria Telnova, Arina Evsiukova, Elena Vodovozova, Anna Eroshova, Elina Dosadina, Sergey Diduk, and Oxana Semyachkina-Glushkovskaya. 2024. "Promising Strategies to Reduce the SARS-CoV-2 Amyloid Deposition in the Brain and Prevent COVID-19-Exacerbated Dementia and Alzheimer’s Disease" Pharmaceuticals 17, no. 6: 788. https://doi.org/10.3390/ph17060788

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