Next Article in Journal
Effects of Dietary Components on Mast Cells: Possible Use as Nutraceuticals for Allergies?
Previous Article in Journal
Acne Transcriptomics: Fundamentals of Acne Pathogenesis and Isotretinoin Treatment
Previous Article in Special Issue
Cardiovascular, Pulmonary, and Neuropsychiatric Short- and Long-Term Complications of COVID-19
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

COVID-19 and Alzheimer’s Disease Share Common Neurological and Ophthalmological Manifestations: A Bidirectional Risk in the Post-Pandemic Future

by
Giuseppina Amadoro
1,2,*,†,
Valentina Latina
1,2,†,
Egidio Stigliano
3 and
Alessandra Micera
4,*
1
Institute of Translational Pharmacology (IFT), National Research Council (CNR), Via Fosso del Cavaliere 100, 00133 Rome, Italy
2
European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy
3
Area of Pathology, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Istituto di Anatomia Patologica, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy
4
Research and Development Laboratory for Biochemical, Molecular and Cellular Applications in Ophthalmological Sciences, IRCCS-Fondazione Bietti, Via Santo Stefano Rotondo, 6, 00184 Rome, Italy
*
Authors to whom correspondence should be addressed.
These authors equally contributed to the work.
Cells 2023, 12(22), 2601; https://doi.org/10.3390/cells12222601
Submission received: 4 October 2023 / Revised: 7 November 2023 / Accepted: 8 November 2023 / Published: 10 November 2023
(This article belongs to the Special Issue Neurological Symptoms and COVID-19 Pandemic)

Abstract

:
A growing body of evidence indicates that a neuropathological cross-talk takes place between the coronavirus disease 2019 (COVID-19) -the pandemic severe pneumonia that has had a tremendous impact on the global economy and health since three years after its outbreak in December 2019- and Alzheimer’s Disease (AD), the leading cause of dementia among human beings, reaching 139 million by the year 2050. Even though COVID-19 is a primary respiratory disease, its causative agent, the so-called Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2), is also endowed with high neuro-invasive potential (Neurocovid). The neurological complications of COVID-19, resulting from the direct viral entry into the Central Nervous System (CNS) and/or indirect systemic inflammation and dysregulated activation of immune response, encompass memory decline and anosmia which are typically associated with AD symptomatology. In addition, patients diagnosed with AD are more vulnerable to SARS-CoV-2 infection and are inclined to more severe clinical outcomes. In the present review, we better elucidate the intimate connection between COVID-19 and AD by summarizing the involved risk factors/targets and the underlying biological mechanisms shared by these two disorders with a particular focus on the Angiotensin-Converting Enzyme 2 (ACE2) receptor, APOlipoprotein E (APOE), aging, neuroinflammation and cellular pathways associated with the Amyloid Precursor Protein (APP)/Amyloid beta (Aβ) and tau neuropathologies. Finally, the involvement of ophthalmological manifestations, including vitreo-retinal abnormalities and visual deficits, in both COVID-19 and AD are also discussed. Understanding the common physiopathological aspects linking COVID-19 and AD will pave the way to novel management and diagnostic/therapeutic approaches to cope with them in the post-pandemic future.

Graphical Abstract

1. Introduction

The relationship between Alzheimer’s Disease (AD)—the most prevalent form of neurodegenerative dementia among the elderly with more than 55 million people worldwide [1,2]—and the global pandemic coronavirus disease 2019 (COVID-19), caused by the novel etiological agent known as Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2), is receiving great attention from the scientific community due to its detrimental impact on healthcare and socioeconomic organizations [3].
Even though the most common clinical presentation of COVID-19 is interstitial pneumonia accompanied by a fever and gastrointestinal problems [4,5], a wide range of multisystemic/organ signs classified as “Post-Acute Sequelae of COVID-19 (PASC)”, “Long-term COVID-19 syndrome”, or colloquially “Long COVID”/”Long Haul” has been described in about 30% of affected patients, from 6 months up to 2 years after the initial phase of the viral infection [6,7,8,9,10]. Prolonged mental manifestations—including memory deficits and depression, confusion and anxiety accompanied by a marked sensory decline with a loss of taste and smell—frequently occur during the acute phase and/or the recovery period, as a part of the multi-faced and complex long COVID-19 syndrome. These epidemiological findings are in agreement with the capability of SARS-CoV-2 to penetrate the Central Nervous System (CNS) along multiple ways (neuroinvasion), where it can, directly and/or indirectly, infect both neurons and glial cells (neurotropism) and, possibly, induce and/or contribute to the development of neurological diseases (neurovirulence) [11,12]. This might not be surprising because the Angiotensin-Converting Enzyme 2 (ACE2) receptor for SARS-CoV-2 is widely distributed in different areas of the human brain, including the prefrontal cortex and hippocampus along with the ocular surface and associated structures [13]. Consistently, the neurotropism and the replication capacity of SARS-CoV-2 have been confirmed in neuronal cultures, brain organoids, mice and human brain autopsies [14,15,16,17,18]. Both viral-dependent (the viral invasion of brain parenchyma and vessels and/or replications) and viral-independent mechanisms (the hijacking of host innate immune response with inflammatory cytokine production, including perivascular inflammation) contribute to SARS-CoV-2-induced neuronal injuries and degeneration leading, eventually, to neurological and neuropsychiatric and neurosensorial symptoms [11,19].
AD is a chronic neurodegenerative disorder which is characterized by the progressive deterioration of cognitive functions due to the selective loss of vulnerable brain areas in association with olphactory and visual dysfunctions [20,21,22,23,24,25]. The extracellular senile plaques (SPs), mainly composed of the Amyloid-beta peptide (Aβ) aggregates, and the neurofibrillary tangles (NFTs), comprised of post-translational modified deposits of the intracellular microtubule-associated protein tau, are the two main distinctive histopathological lesions. Although the precise etiology is still unknown, aberrant protein misfolding, vascular damage involving large and small brain vessels, immunosenescence, neuroinflammation and the increased production of pro-inflammatory cytokines, Blood–Brain Barrier (BBB) breakdown, oxidative stress with the overproduction of Reactive Oxygen Species (ROS), mitochondrial dysfunction, synaptic derangement and inappropriate elimination and neural loss are implicated in the onset/progression of AD [20,21,22,23,24,25].
In this review, we highlight the clinical/epidemiological aspects and the molecular physiopathological mechanisms pointing to an increased susceptibility of developing AD in subjects that have experienced the COVID-19 infection. To support this finding, we give detailed insights into the neurochemical interplay occurring between COVID-19 and AD, both in the brain and eye, by paying particular attention to the many common risk factors and neuro-ophthalmological complications shared by these two disorders. We hope that this review, including for the first time a section devoted to ocular manifestations occurring both in COVID-19 and AD, will provide interesting information for researchers, clinicians and ophthalmologists working in the field.

2. Neuroinvasive Mechanisms of SARS-CoV-2 and Neurological Manifestations of COVID-19

Clinical and experimental evidence has shown that the SARS-CoV-2 infection can affect multiple organs beyond the respiratory system, including the Central (CNS) and Peripheral Nervous System (PNS), thus triggering per se neuronal injuries and/or exacerbating the neurodegenerative conditions of pre-existing diseases [26,27,28]. For mechanistic insights, the cerebral and mental complications of COVID-19 are the neuropathological consequences of one or a combination of all of the following factors: (1) direct viral neuronal damage leading to encephalitis (virus-induced neuropathology); (2) systemic inflammation with “cytokine storm” causing the damage of peripheral organs (the liver, kidney and lungs) which indirectly affects the brain’s health (neuroimmunopathology); (3) global ischemia secondary to respiratory insufficiency and the so-called acute respiratory distress syndrome (ARDS); and (4) cerebrovascular damage (blood vessels and coagulopathies) with ischemic or hemorrhagic strokes. Consistently, a high incidence of both CNS and PNS persistent symptoms and/or delayed or long-term neurological, sensorial and motor manifestations are associated with the pathogenesis of SARS-CoV-2 infection. Hyposmia, headache, dizziness, ataxia, cerebrovascular injury, hypogeusia, nausea, encephalitis, fatigue, myalgia, ataxia, neuropathies, conjunctivitis, retinopathy, encephalopathy, myelitis, vomiting, delirium, psychosis, ischemic stroke, epileptic seizures, neurocognitive and psychiatric complications, acute respiratory distress syndrome and affective disorders are recorded in observational studies on COVID-19 survivors [26,29,30,31] (Figure 1). More importantly, the SARS-CoV-2 neuroinvasion of the CNS has been claimed based on quite a few in vitro and in vivo analyses ranging from immunohistochemistry, in situ hybridization, Real-Time Polymerase Chain Reaction (RT-PCR) and Transmission Electron Microscopy (TEM) carried out on autoptic brain tissues and the CerebroSpinal Fluid (CSF) of patients who died of COVID-19 to experimental evidence on human-induced Pluripotent Stem Cells (iPSCs) and brain organoids and animal models [11,17,32,33,34,35,36,37,38]. However, it is also worth noting that, up to now, the definite evidence of SARS-CoV-2’s presence in the nervous system is a matter of debate since neither viral RNA nor particles have been found in human tissues and CSFs by other researchers [39,40,41,42,43]. Moreover, not all animals inoculated with the SARS-CoV-2 virus have shown neurological complications or full-blown CNS infections [44,45].
In general, the replication process of SARS-CoV-2 into host cells requires the initial binding of viral Spike Protein 1 (SP1) to its membrane-anchored ACE2 receptor, even though other proteins such as integrins, neuropilin-1 and the TransMembrane PRoteaSes Serine 2 and Serine 4 (TMPRSS2 and TMPRSS4, respectively) can also take part in it [27,46,47,48,49] (Figure 2). Relevantly, the evidence that the ACE2 receptor as long as two other co-receptors such as TMPRSS2 and neuropilin-1 [27] are widely distributed throughout the CNS and PNS, including the brainstem, cortex, striatum, hypothalamus, choroid plexuses, spinal cord, olfactory neuroepithelium, retinal ganglion cells, tongue gustatory nerve and neuromuscular junction [26,27,28], provides the strong biological rationale for SARS-CoV-2 neurotropism [31,32,50].
In this connection, several both direct and/or indirect routes for the invasion of SARS-CoV-2 into the nervous system (Figure 3) have been proposed [26,30,51,52,53]:
-
The hematogenous pathway or “Trojan horse mechanism” wherein infected circulating immune cells serve as reservoirs for the virus that traverses from the bloodstream to the CNS (cell transmigration); SARS-CoV-2 infects, near the vessel wall, the resident peripheral immune cells of the blood circulation (phagocytic monocytes/macrophages, neutrophils and lymphocytes) which, in turn, penetrate the neurovascular unit of the Blood–Brain Barrier (BBB), becoming a pool of viral dissemination toward the CNS [54,55,56] (Figure 3a);
-
The Blood–CerebroSpinal Fluid (B-CSF) pathway (paracellular migration): SARS-CoV-2 binds to the ACE2 receptors of the endothelial cells and damages the integral citoarchitecture of the BBB. To get into the brain, the virus locally activates the signaling transduction pathway of Nuclear Factor kappa B (NF-kB) transcription factor, leading to an up-regulation in the basal expression level of Matrix MetalloPeptidase 9 (MMP9) which, in turn, degrades the extracellular matrix with consequent increased B-CSF permeability and alterations in immune cell trafficking (MMP8, Monocyte Chemoattractant Protein-1 (MCP-1), InterCellular Adhesion Molecule 1 (ICAM-1), a neuroinflammatory response with the release of pro-inflammatory cytokines and chemokines such as Interleukin (IL) IL-2, IL-6, IL-7 and IL-8, Tumor Necrosis Factor (TNF) TNFα, C-C Motif Chemokine Ligand (CCL) CCL2, CCL3 and CCL7 and C-X-C motif chemokine ligand (CXCL) CXCL10) [54,57] (Figure 3a);
-
The transneuronal spreading or “neuronal route” (via exocytosis/endocytosis or “fast axonal transport” mechanisms of vesicles along the microtubules track in order to move the virus from synaptic terminals back towards neuronal cell bodies) from systemic organs to the CNS throughout the cranial nerves: In this process, the virus first enters the nerve endings (i.e., the peripheral nerves) and then is retrogradely transported to the soma to invade the CNS; in detail, SARS-CoV-2 enters through (i) the olfactory mucosa (causing anosmia), and it spreads via the olfactory nerve to the olfactory cortex; (ii) the lacrimal and salivary glands, and it spreads via the facial VII and glossopharyngeal IX nerves to their respective brainstem nuclei; (iii) the taste buds of gustatory mucosa (triggering ageusia), and it spreads via the VII and IX nerves to the Nucleus Tractus Solitarius (NTS) located in the brainstem; or (iv) the respiratory system, and, via the vagus nerve X, it spreads both to other systemic organs (the heart, kidneys and gastrointestinal tract) innervated by this nerve and to the brainstem [58] (Figure 3b);
-
The circumventricular organs (CVO) lacking the BBB: SARS-CoV-2 enters the CNS through the ACE2-expressing and vascularized subfornical organ, the paraventricular nucleus, the NTS and the rostral ventrolateral medulla by triggering local neurovascular damage (Figure 3b);
-
The ocular system: the epithelial cells of the cornea and conjunctiva, the trabecular meshwork, choroid and retinal cells, optic nerve and geniculo-calcarin tract expressing the ACE2 receptor and neuropilin-1 are also entry points for the SARS-CoV-2 infection towards the occipital cortical areas [26,30] (Figure 3c).
Once in the CNS, as with other viruses endowed with neurotrophic properties, SARS-CoV-2 binds to neurons, astrocytes, oligodendrocytes and microglia, which all express, on their membrane, both ACE2 and TMPRSS2 receptors, and then spreads to multiple brain areas including the cerebral cortex, caudate/putamen, ventral striatum, thalamus, hypothalamus (paraventricular nuclei), spinal cord, hippocampus, frontal cortex, substantia nigra, middle temporal gyrus and along the synapse-interconnected anatomic networks, causing, eventually, neuronal cell dysfunction and degeneration [29,30].

3. Bidirectional Relationships between Long COVID-19 and AD

Severe and debilitating neurological complications that are classically associated with AD symptomatology, such as memory deficits (73%) and cognitive impairments (brain fog) (85%), have also been recorded in follow-ups more than 2 years after the resolution of the acute infection of SARS-CoV-2, occurring at similar rates in hospitalized and non-hospitalized adults [8,9,59,60,61,62]. In line with this finding, a growing body of longitudinal, prospective and retrospective studies indicates that the virus neurotropism can per se significantly trigger and/or contribute to the occurrence of AD-like neuropathological features in the brain, even though how the post-infection sequelae of COVID-19 actually impacts mental processes concerning the acquisition, storage, integration and retrieval of information needs to be fully clarified [30,63,64,65,66,67,68]. In addition, whether the reciprocal association between SARS-CoV-2 and AD implies a direct causal relationship and/or originates from chronic and excessive systemic inflammatory conditions also remains to be determined. Nevertheless, a strong bidirectional relationship existing between the COVID-19 infection and AD development has been clearly documented [69,70]. On one side, elderly individuals with AD are more prone to the SARS-CoV-2 infection, showing an increased chance of severe COVID-19 complications and mortality [71,72,73,74,75,76,77]. On the other side, people who have experienced COVID-19 are at a greater risk of suffering AD, with a global reduction in attention and executive and visuospatial functions [77,78,79,80,81,82,83,84,85,86,87,88,89,90]. Consistently, a marked mental decline in connection with an overall reduction in brain size and a diminution of grey matter thickness in the orbitofrontal cortex and para-hippocampal gyrus—two cerebral areas that are largely affected in AD subjects—have been demonstrated in subjects recovering from COVID-19 when compared to healthy controls, even in non-hospitalized patients [91]. In addition, a mouse model of mild SARS-CoV-2 infection displayed several morphological, molecular and biochemical markers typical of AD, including an impaired hippocampal neurogenesis, microgliosis, myelin disintegration, elevated CSF levels of cytokines/chemokines, such as CCL11, and neuronal loss with cognitive dysfunctions [92]. Bioinformatic screening of the SARS-CoV-2 proteome has also revealed different peptides with a high propensity to self-aggregate into amorphous and fibrillary amyloid clumps which are toxic to neurons, which also occurs in AD brains [93]. Anosmia, due to sustained and protracted inflammation, is caused by the persistence of the SARS-CoV-2 virus in the olfactory mucosa and/or in the olfactory bulb of the COVID-19-affected brain [94,95,96,97,98,99], and, in parallel, this sensorial complication, particularly the inability of olfactory identification/discrimination, is visible in the early/prodromal stages of AD subjects suffering from Mild Cognitive Impairment (MCI) [100]. Hypometabolism detected in AD brains with BBB leakage/dysfunction and cerebral microvascular changes have been also reported in patients with long COVID-19 in correlation with specific cognitive symptoms [101,102]. Furthermore, the activation of Kynurenine signaling—a cellular pathway whose stimulation is involved in the regulation of immune tolerance, neurotoxicity and vascular injury—is dysregulated both in AD [103] and COVID-19. To this point, in a large cohort of cases recovering from mild-moderate to acute SARS-CoV-2 infection across a 12-month period, a causal relationship among the presence of its typical metabolites, such as Quinolinic Acid (QA) and Kynurenine (Kyn) 3-HydroxyKynurenine (3HK) and 3-hydroxyantranilic acid (3HAA), intellectual disabilities and anosmia has been recently reported [104]. In the reminiscence of structural and metabolic mitochondrial alterations responsible of energy deficiency that drives the loss of dendritic spines and synapses occurring in AD development [105,106], abnormal levels of mitochondrial proteins as well as SARS-CoV-2 spikes and nucleocapsid proteins have been also detected both in neuron- and astrocyte-derived exosomes in the plasma of COVID-19 patients with neurological and psychiatric manifestations [107]. More importantly, the SARS-CoV-2 infection provokes and/or precipitates several neurodegenerative processes, in particular, widespread neuroinflammatory response, synaptic pruning, protein misfolding, the disruption of the oxidation-reduction systems, damage to blood vessels by coagulopathy and endothelial dysfunction and neuronal injuries, that are all traits classically discernable in AD brains [108]. As a matter of fact, the virus damages not only the post-mitotic neurons but also the surrounding astrocytes and microglia and, thus, indirectly further aggravates the brain injury, owing to the exaggerated release of pro-inflammatory cytokines and/or deleterious Reactive Oxygen Species (ROS) [32]. Moreover, in addition to triggering neurodegeneration and neuroinflammation, SARS-CoV-2 also promotes the chronicity of these changes, up to months or even years after the acute infection, since it invades and diffusely infiltrates/propagates throughout the brain via trans-synaptic spreading along the motor-based, microtubule-dependent axonal transport [26,31,51,66]. More importantly, ACE2 is co-expressed in both GLUtamatergic and GABAergic neurons, indicating that, in the CNS, SARS-CoV-2 infection is able to interfere with the signaling transduction pathways activated by these two neurotransmitters regulating the cortical excitability. Therefore, SARS-CoV-2 seems to initiate and/or exacerbate the imbalance between excitatory and inhibitory electrical neuronal circuits, leading to excitotoxicity and cell loss, which also occurs in AD progression [109,110]. Finally, based on the evidence connecting the repeated infection of Herpex Simplex Virus type-1 (HSV-1) and amyloidosis, the viral re-activation of SARS-CoV-2 in the CNS in concomitance with an age-dependent physiological decline of innate immunity is more likely to trigger an inflammatory process which, in turn, increases the Aβ synthesis and accumulation, as well as the hyperphosphorylation of tau (pTau) and aggregation, a cascade that is suggestive of the so-called “infection hypotesis of AD” [68,111].

4. Common Risk Factors and Involved Mechanisms That Mediate the Association between COVID-19 and AD

Compelling studies have shown that COVID-19 and AD share several physiopathological aspects including ACE2 expression, age, inflammation with “cytokine storm”, oxidative stress, the APOE4 genetic variant, the neurotransmitter system, hypoxia and the activation of intracellular pathways associated with the altered metabolism of APP/Aβ and tau (Figure 4).

4.1. ACE2 and Ageing

ACE2, the SARS-CoV-2 receptor required for cell entry, is considered the most important determinant in dictating the greater susceptibility of developing AD among COVID-19 patients [108,112]. Microarray, Western blotting, Reverse Transcription quantitative Polymerase Chain Reaction (RT-qPCR) and immunostaining analyses have undoubtedly shown that the expression levels of ACE2 significantly increase in the brain tissues of human AD subjects when compared with healthy, not-demented controls and in close relationship with the severity of clinical dementia and different neuropathological parameters, including the density of dystrophic neurites, the Aβ plaques and NFT accumulation [113,114,115]. In addition, in a SARS-CoV-2 pseudovirus infection model, the fibrillogenic and highly-neurotoxic Aβ1-42 peptide—but not the shorter Aβ1-40 one—binds to both the S1 protein and ACE2 receptor [116], by facilitating the virus invasion and production of IL-6. Apart from being a SARS-CoV-2 receptor, ACE2 is also a key regulator of the Renin-Angiotensin System (RAS) that is one of the most complex hormonal regulatory axes involved in maintaining the body homeostasis and exerting a broad range of other important functions in multiple organs, in particular the cardiovascular and immune systems. In detail, ACE2 catalyzes the Angiotensin II conversion to Angiotensin(1–7) (Ang 1–7) which, in turn, binds to its G Protein-Coupled Receptor (GPCR) MAS to regulate several downstream signaling cascades, for instance the Phosphatidyl-Inositol 3-Kinase (PI3K)/Akt serine/threonine kinase 1 (Akt/) cAMP response element-binding protein(CREB)/Brain-Derived-Neurotrophic Factor (BDNF)/Tropomyosin receptor kinase B (TrkB) [112]. Interestingly, the SARS-CoV-2 infection seems to instigate and/or accelerate the AD phenotype by inhibiting ACE2 enzymatic activity, triggering a hyperinflammatory response and downregulating the secretion of BDNF, a potent neurotrophin endowed with crucial functions in supporting neurogenesis, cognition and the prevention of neurodegeneration upon binding to its cognate transmembrane TrkB receptor protein [117]. In concomitance with an elevation in the mRNA transcript of ACE2 facilitating the entry points of SARS-CoV-2 in the CNS, a high level of its TBS/Detergent-soluble inactive form has also been detected in the parietal cortex of two large cohorts of AD-fully diagnosed subjects when compared to controls, suggesting that a defective brain RAS signaling with a consequent decrease in its anti-inflammatory and neuroprotective properties is more likely to take place in humans with a low cognitive score [115].
Ageing is the greatest contributing factor to AD onset/progression by causing genomic instability, telomere shortening, epigenetic modifications, a loss of proteostasis, a decline in mitochondrial respiration and energy production, deregulated nutrient sensing, altered intercellular communication, an increased permeability of BBB and deregulated inflammation [118]. Another important hallmark of ageing is cellular senescence [119,120], a terminal state of cell-cycle arrest characterized by the proinflammatory Senescence-Associated Secretory Phenotype (SASP) as result of an increased release of various tissue-remodeling (e.g., Tumor Growth Factor (TGF) TGF-β and MMPs) and immune-related (e.g., IL-6, IL-8 and IFNs) factors involved in regeneration/repair and immunosurveillance. In chronic age-associated neurodegenerative diseases such as AD, these events persist for long time and then turn out to be detrimental, with consequent organ dysfunctions, aberrant paracrine senescence and chronic inflammation [121,122]. In line with this notion, the elimination of senescent cells by means of senolytic compounds significantly mitigates the extent of neuropathology [123]. It is noteworthy that SARS-CoV-2 infection can induce per se a condition of premature senescence both directly, by increasing the secretion of InterFeroNs (IFNs) and other pro-inflammatory mediators such as CXCL-10, CCL-2, IL-6, IL-8, IL-12, IL-1β, IFN-γ and TNF-α from infected cells, and/or indirectly, by promoting the release of Danger-Associated-Molecular Patterns (DAMPs) via necroptosis and pyroptosis [124,125]. Moreover, and more importantly, older COVID-19 patients are more likely to accumulate huge levels of cellular senescence, since aged tissues show a decreased intrinsic capacity of repairing damages and/or eliminating senescent cells via the immune system [126]. Relevantly, the age-dependent decay of immune defense against SARS-CoV-2 infection, the so called “immunosenescence and inflamm-aging”, plays a major role in boosting the vulnerability to severe COVID-19 outcomes in older adults [127,128,129,130]. In agreement, in COVID-19 patients, a strong association has been documented between the severity of infection with more severe-to-lethal outcomes and the presence of the immunosenescence phenotype with a high level of the Neutrophils-to-Lymphocytes Ratio (NLR) [131] and IL-6 production [132]. Interestingly, the reduction in telomere elongation and the reactivation of reverse transcriptase telomerase [133], two important molecular hallmarks of cellular senescence in chronic neurodegenerative diseases, critically influence the severity of COVID-19 symptoms, as proved by the observation that an elevated risk of developing grave and fatal complications is found in SARS-CoV-2-infected patients carrying shorter telomeres from their peripheral blood lymphocytes [134]. Several senolytic compounds, such as the flavonoid Quercetin and the mammalian Target of Rapamycin (mTOR) kinase inhibitor Sirolimus—both known to reduce the SASP and prevent the senescence induction (geroconversion)—are currently exploited in clinical trials to counteract the long-COVID-19 syndrome [126]. Finally, the up-regulation of the steady-state expression level of ACE2 occurring in several human tissues with an increasing age, mainly in the nasal neuroepithelium, which is one of the most accessible routes of SARS-CoV-2 invasion in the CNS, also accounts for the elevated risk of contracting COVID-19 in the elderly population in connection with poor clinical outcomes [135].

4.2. Neuroinflammation, Oxidative Stress and Nicotinic Cholinergic System

Apart from the premature synapses’ elimination and neuronal deterioration tightly associated with a cognitive decline, a pronounced neuroinflammation characterized by reactive microglia, astrogliosis and the infiltration of cytotoxic CD8-positive T cells is among the most prominent neuropathological traits discernable in the brains of patients who died from both AD [136,137,138,139] and COVID-19 [37,140,141]. In this regard, COVID-19 chronic inflammation is caused both directly, by the SARS-CoV-2 infection of the CNS, and indirectly, by peripheral inflammation via immune-to-brain signaling [52]. Consistently, IL-6, IL-1, TNFα, complement proteins and Galectin-3/9 are common prognostic biomarkers for the activation of inflammatory immune responses in the CNS, following both SARS-CoV-2 neuroinvasion and AD [142]. By single-nucleus RNA sequencing (snRNA-seq), followed by immunohistochemical staining validation, an excessive stimulation of microglia and brain-barrier inflammatory signals in concomitance with a downregulation in the expression of neuronal genes encoding several synaptic vesicle components, such as synaptobrevins (VAMP1 and VAMP2), SynTaXin 1B (STX1B) and the SyNAPtosome-associated protein of 25 kDa (SNAP25), which regulate the glutamate release and excitatory neurotransmission, have been also documented in post-mortem brain tissues from individuals with AD [143] and COVID-19 (frontal cortex and choroid plexus) [144]. Moreover, the activation of PYrin (PYD)-Domain-containing protein 3 inflammasome (NLRP3) [145], which affects the microglial-dependent clearance of Aβ [146] and promotes tau pathology [147], is triggered in the brain as a consequence of SARS-CoV-2 neuroinvasion, just as described in the AD etiology.
Oxidative stress with the excessive production of the harmful ROS provoking Aβ accumulation/aggregation and tau hyperphosphorylation in AD brains [148] also takes part in the innate response against SARS-CoV-2 invasion [149,150]. A large amount of activated radical-producing neutrophils are found in COVID-19 patients, consistent with a massive production of ROS [151,152]. Changes in mitochondrial respiration and associated redox imbalances have been detected in Peripheral Blood Mononuclear Cells (PBMCs) from patients with COVID-19, in agreement with the energy supply required for the production of pro-inflammatory cytokines during the virus-triggered immune response [153]. Interestingly, upon exposure to SARS-CoV-2, the activation of inducible Nitric Oxide Synthase (iNOS), an important biological mediator of inflammation and immunoregulation producing Nitric Oxide (NO) from L-arginine, causes an overproduction of the superoxide radical ion (O2) in people who have survived the acute phase of COVID-19 that becomes self-perpetuating, even when the virus has been cleared, turning into a persistent and protracted free radical-induced damage [154].
In addition to being involved in the CNS in high-order cognitive processing, sensory information integration, sleep and wakefulness, Acetylcholine (Ach) and its nicotinic Receptors (nAChRs) play a pivotal role in the homeostatic regulation of inflammatory response, owing to the high expression of the α7 receptor (α7nAChR) on the surface of immune cells (B cells, macrophages and T cells) [155,156,157]. Therefore, it is not surprising that the dysregulation of the nicotinic cholinergic system is involved, in parallel, in both COVID-19 and AD pathophysiology [158,159,160]. In support of this finding, the SARS-CoV-2 S1 glycoprotein protein has proved to interact with the cholinergic nicotinic ACh receptor α7 (α7nAChR) and to negatively impair its function by preventing the acetylcholine’s binding and, in turn, the specific intracellular activation of its downstream signaling(s) [161,162,163]. Interestingly, nicotine—a selective agonist of α7nAChR—was recently approved as a promising therapeutic option to counteract the neurological complications in COVID-19 syndrome, as a direct result of its potent anti-inflammatory and neuroprotective actions [159,164]. Likewise, the neocortical cholinergic innervation is also gradually destroyed in AD brains—mainly due to the sequential and aberrant deposition of the insoluble, ThioflavinT (ThT)-positive, dense SP and NFTs—with consequent clinical changes in cognition, behavior, mood and emotions [160]. In particular, alterations in acetylcholine release in concomitance with a decrease in high-affinity choline uptake and a downregulation in muscarinic and nicotinic acetylcholine receptor expressions represent a solid ground for the cholinergic hypofunction detected in AD. In line with this so-called “cholinergic hypothesis”, in vitro and in vivo studies have clearly shown that the Low Molecular Weight (LMW) oligomeric Aβ conformers—which are the most neurotoxic species to synapses found in autoptic AD brains—actually bind to and impair the function of the α7nAChR, both in cultured hippocampal neurons and in mouse models [158,159,160]. Finally, similar to COVID-19, a pharmacological strategy in the clinical management of AD is based on the preservation/restoration of cholinergic neurotransmission, and, in this framework, several FDA-approved cholinesterase inhibitors such as donepezil, rivastigmine and galantamine are currently used in therapy to slow down the cognitive and functional decline in affected patients [158,159,160].

4.3. APOE Genetic Variant and Signal Pathways

Another strong risk factor which accounts for more than 95% of all sporadic AD cases is the genetic variant APOE4 [164,165,166] that plays a causal role in the alteration of cellular trafficking/the metabolism of cholesterol and in the regulation of the aggregation state and deposition of Aβ peptide(s). Interestingly, the APOE polymorphism is also involved in the COVID-19 syndrome by increasing the incidence and severity of the SARS-CoV-2 infection and neurodegeneration [167,168,169,170,171,172,173,174]. In this connection, several lines of evidence have shown that the APOE4 genotype (i) increases the permeability of the BBB, which makes patients more susceptible to viral infections [175]; (ii) augments the production of pro-inflammatory cytokines by peripheral macrophages and CNS microglia [176]; (iii) elevates the infectivity of SARS-CoV-2 both in neurons and astrocytes [177]; (iv) controls the cholesterol homeostasis which, in turn, facilitates the binding of the S1 protein to the ACE2 receptor during the first step of the SARS-CoV-2 infection [178]; (v) decreases the expression of several antiviral defense genes, including InterFeron-Induced TransMembrane Proteins (IFITM) IFITM2 and IFITM3, InterFeroN Alpha(α) and Beta(β) Receptor Subunit 1 (IFNAR1) and Lymphocyte Antigen 6 Family Member E (LY6E) [179,180,181]; and (vi) downregulates the ACE2 expression, followed by an imbalance in Renin-Angiotensin System (RAS) that catalyzes the degradation of Angiotensin II (Ang II) to Angiotensin(1–7) (Ang 1–7), associated with the chronic hyperinflammatory state of COVID-19 [182].
The activation of intracellular signaling transduction pathways associated with APP/Aβ and tau pathologies represents an additional relevant commonality, putting in mutual relation the occurrence of COVID-19 and AD. It is widely acknowledged that the main histopathological features detected in the vulnerable regions of AD brains, such as the entorhinal region and the hippocampus which are involved in memory/learning and synaptic plasticity, are the SP and the NFTs. These two lesions are, respectively, composed in their aggregated forms of Aβ peptide(s)—generated by the aberrant and sequential beta/gamma (β/γ)-mediated amyloidogenic proteolysis of its membrane precursor Amyloid Precursor Protein (APP) involved in synaptogenesis and neurogenesis—and by abnormally hyperphosphorylated and/or truncated tau protein, whose function is the modulation of the intracellular stability of axonal microtubules [183,184]. Consistently, the mRNA of the holoprotein APP has turned out to be greatly upregulated in single-cell RNA-seq studies carried out on blood samples from COVID-19 survivors in comparison with controls ones [185] and on oligodendrocytes isolated from their post-mortem brain tissues [186], hinting at the deregulation of APP metabolism and its proteolytic cleavage. Furthermore, just as detected in AD, a tendency toward an accelerated APP amyloidogenic processing/Aβ deposition in the brains from patients with the COVID-19 neurological syndrome is evident when compared to healthy controls and in connection with significant lower amounts of the soluble Amyloid Precursor Protein alpha and beta fragments (sAPPα and sAPPβ) as well as the Aβ40, Aβ42 and Aβ42/Aβ40 ratio in their peripheral CSFs [187]. The transcriptomic and interactomic profiles from the frontal cortex of fully diagnosed AD subjects with cognitive decline who have also experienced COVID-19 have revealed that SARS-CoV-2 can indirectly amplify the Aβ toxicity in the brain, giving rise to neuroinflammation and an imbalance in the relative levels of cellular pro-oxidants and antioxidants [188].
Relevantly, Aβ1-42, but not Aβ1-40, binds with high affinity to both the viral S1 protein and ACE2 receptor of SARS-CoV-2 [189]. Immunohistochemical staining studies using different specific antibodies clearly decorate insoluble, proteinaceous Aβ-positive aggregates in the autopsied brains of patients who died of COVID-19 [190]. From a mechanistic point of view, immunofluorescence and RNA-seq analyses performed on the cortical and hippocampal tissues of transgenic mice expressing human Angiotensin-Converting Enzyme 2 (hACE2) suggest novel insights by showing, for the first time, that the SARS-CoV-2 spike protein S2 subunit is able per se to enhance the Aβ production via direct binding to and the modulation of the processing enzymatic activity of the γ-secretase complex [191]. A persistent brain neuropathology with the accumulation of AT8 (ptauSer202, Thr205), an AD-pathognomonic site of tau hyperphosphorylation, has been also reported in Syrian golden hamsters after the intranasal inoculation of SARS-CoV-2 [192]. Furthermore, the activation of NF-κB signaling driving the neuroinflammatory cascade in response of SARS-CoV-2 neurotropism [193,194] also activates the beta(β)-site APP cleaving enzyme 1 (BACE-1) activity, thereby triggering the first step of proteolytic cleavages sub-serving the sequential generation of Aβ peptide(s) [195]. A marked elevation in tau phosphorylation at multiple AD-like epitopes, such as pSer262, pSer214 and pSer356 and pSer199/202, has also been ascertained in COVID-19 brains in concomitance with the activation of several known tau-directed kinase, including the AMP-activated Protein Kinase (AMPK), Glycogen Synthase Kinase 3 beta (GSK3β), Protein Kinase A (PKA) and Calcium/Calmodulin-Dependent Protein Kinase II (CAMKII) [196]. Moreover, the levels of specific peripheral biomarkers that are routinely used in clinical practice for AD diagnosis—such as total tau protein in association with IL-6, Neurofilament Light Chain (NFL) and Glial Fibrillary Acidic Protein (GFAP)—are elevated in the CSF and serum of COVID-19 patients [197,198]. More importantly, SARS-CoV-2 targets the neurons of 3D human brain organoids, inducing an aberrant subcellular redistribution of tau from axonal processes to soma, hyperphosphorylation at Threonine 231 (Thr231), apoptotic caspase-3 activation and, eventually, neuronal death, specific cellular changes that are reminiscent of key features associated with AD neuropathology [199]. In a similar way, hyperphosphorylation at the Ser262 and Ser396 sites and the mislocalization and increased aggregation of tau are also detected in human neuron-like SH-SY5Y cells after in vitro infection with different clinical strains (B.1, B.1.1.7 and B.1.617.2) of SARS-CoV-2 [200]. Furthermore, by using a combination of ThT assay, Transmission Electron Microscopy (TEM) staining, analytical High-Performance Liquid Chromatography (HPLC) and Mass Spectrometry (MS), Eberle and colleagues have recently reported that tau is proteolytically cleaved in vitro by the viral SARS-CoV-2 3CL protease with a consequent release of the 25kDa fragment, triggering the formation of amorphous fibrils, resembling the paired helical (PHF) and/or straight filaments (SFs) typically detected in AD brains [201]. Finally, in brain cortices from Murine Hepatitis Virus-1 (MHV-1) coronavirus-inoculated mice—an in vivo model which is very similar to the SARS-CoV-2 infection observed in humans—a significant increase in AT8-tau hyperphosphorylation along with reactive astrocytes and microglia (GFAP and Iba1-positive, respectively) and reduced synaptophysin-1 synaptic protein are found up to 12 months post-infection, again recapitulating several characteristic features of chronic neurodegenerative human tauopathies, including AD [202].

4.4. Hypoxia

Among the major clinical manifestations of COVID-19 with acute respiratory distress syndrome (ARSD) stands out hypoxia, a pathological condition that is caused by the lack of oxygen and/or the accumulation of mitochondrial ROS into the lungs’ upper airways due to a maladaptive inflammatory response to viral penetration [203]. Relevantly, the SARS-CoV-2 infection induces the cellular expression of Hypoxia-Inducible Factor 1α (HIF-1α), an important transcriptional factor responsible for cellular adaption to low oxygen tension, which, in turn, plays a key role in driving the virus-mediated inflammatory response (cytokines storm), metabolic reprogramming and oxidative stress [204,205,206]. Just as SARS-CoV-2 damages the pulmonary tissue locally inhibiting the gas exchange, post-mitotic neurons with high energy requirements are particularly vulnerable, in the brain, to any subtle change in oxygen saturation, resulting in the activation of pro-apoptotic signaling pathways and, eventually, neuronal injuries [203]. Therefore, it is not surprising that hypoxia significantly increases the risk of developing neurodegenerative diseases, in particular AD, with the dysregulation of the HIF-1α pathway leading to AβPP amyloidogenic processing with Aβ accumulation, due to increased production [207,208,209] and/or decreased degradation [210], tau hyperphosphorylation and microglial activation [211]. Focal deposits of Aβ have been detected in the brains of young (less than 60 years old) hospitalized patients who died of COVID-19 in correlation with widespread hypoxic damage [212]. Interestingly, the important contribution of oxygen dyshomeostasis to long-term cognitive impairment in the post-COVID-19 syndrome has been highlighted in a recent follow-up study reporting that an improved memory and attentional capacities, likely due to delayed hippocampal damage, are observed in a small cohort of patients that underwent oxygen therapy and were prospectively recruited after 3–9 months of the SARS-CoV-2 infection [213]. In line with this, prolonged hyperbaric oxygen treatment can reduce hypoxia, neuroinflammation, the accumulation of Aβ and phosphorylated tau, leading to a significant improvement of cognitive performances in a 3xTg AD mouse model carrying three mutations associated with familial AD (APP Swedish, MAPT P301L, and PSEN1 M146V) when tested in hippocampal-dependent behavioral tasks [214] and, possibly, in affected patients [215].

4.5. Serotonin or 5-Hydroxytryptamine (5-HT)

Serotonin or 5-HydroxyTryptamine (5-HT), a monoamine neurotransmitter involved in the control of mood/reward and learning, is an additional link to explaining the neurocognitive impairments associated with COVID-19 and AD. In this regard, an elegant study recently reported that the long COVID syndrome is associated with a low peripheral level of circulating serotonin which, in turn, impairs the hippocampal-dependent memory function via the reduced stimulation of the vagus nerve signaling [216]. Interestingly, Selective Serotonin Reuptake Inhibitors (SSRIs) are widely prescribed to treat neurobehavioral symptoms associated with dementia [217], and lower levels of serotonin have been detected in AD brains [218].

5. Neuro-Ophthalmic Complications Shared by COVID-19 and AD

Compelling experimental, molecular, histological and clinical studies suggest that the neuro-ophthalmic system and related visual manifestations are another important pathogenetic connection between the COVID-19 and AD syndromes (Figure 4).
In this context, the eye and associated ocular structures are possible transmission routes of SARS-CoV-2 penetration to the brain [219,220,221,222,223,224]. Moreover, in addition to the visual pathway which provides a direct anatomical connection between the ocular surface and the brain, the hematogenous route has been recently proposed as an alternative mode of penetration/transmission of the virus from eye to body. Indeed, after the infection of the iris and conjunctival cells both expressing the ACE2 receptor, SARS-CoV-2 can reach the blood capillaries and then gain access through the Blood–Retinal Barrier (BRB) in the Retinal Pigment Epithelium (RPE) and blood vessel endothelial cells to reach, eventually, the bloodstream and infect the extraocular areas [220]. Thus, in humans, the localization of the ACE2 receptor, required for an efficient viral entry from the eyes, can be considered both intra- and extra-ocular, with large expressions in conjunctival and corneal cells, retinas and retinal pigment epithelium [224,225,226,227,228,229]. In agreement, SARS-CoV-2 can infect and replicate in retinal organoids, and quantitative Real-Time Polymerase Chain Reaction (RT-PCR) analyses have confirmed the presence of SARS-CoV-2 genomic RNA in different ocular tissues including human retina, cornea, conjunctiva, lacrimal sacs and tears from deceased cases with COVID-19 [230,231,232,233,234]. Finally, ocular complications are frequently described by patients both during and after recovery from COVID-19, especially conjunctivitis, retinopathy (retinitis, retinal hemorrhages, retinal venous and arterial occlusion), uveitis, vitritis, optic neuritis [219,222,235,236,237,238,239,240] in association with signs of excessive inflammation, nerve fiber loss, increased dendritic cell density, impaired retinal microcirculation and poor vision [239,241,242,243,244,245,246].
Regarding AD, there is a growing body of literature endorsing the concept that the two hallmarks classically discernable in the brains of affected patients and preclinical animal models—i.e., the deposits of Aβ and hyperphosphorylated tau protein—are also present in their eyes, sometimes even before the appearance of clinical cognitive symptoms, in close association with other ocular pathophysiological alterations such as nerve fiber layer thinning, the degeneration of retinal ganglion cells, vascular alterations, local inflammatory responses and gliosis [247,248,249,250,251,252]. These findings are in agreement with the fact that the retina and optic nerve are neurodevelopmental outgrowths of the CNS, while the aqueous humor and tear film located in the anterior eye segment are more likely to be comparable to the CSF. Consistently, changes in functional visual processing are detected in subjects suffering from AD, including a loss of the visual field, decreased contrast sensitivity, low visual acuity, impaired color vision or motion perception and visuospatial deficits [253,254,255,256,257,258,259,260]. More importantly, in light of the great accessibility of the eyes, which are considered a direct “window” to brain, advanced high-resolution imaging techniques detecting ocular Aβ and pTau in the retina are currently used as predictive and diagnostic biomarkers in the clinical management of AD by allowing for the large-scale noninvasive screening and monitoring of at-risk populations [246,261,262].

6. Conclusions and Future Perspectives

Despite the great interest in the COVID-19 pandemic outbreak and its neurological consequences, it is important to remember that there are still several controversial results concerning the presence of the SARS-CoV-2 virus in the brain [263]. To this point, several studies have identified the direct neuro-invasive capacity of SARS-CoV-2 to enter the brain [264,265] while others do not confirm the presence of the virus within the brain [38,266,267] or report very low levels of detectable RNA and viral protein brains [37,140], suggesting that the virus neuropathology is more likely to be mediated by cytokines through systemic effects [52].
Nevertheless, among the long-term manifestations of post-COVID-19, AD-like dementia stands out as the most frequent disorder with higher susceptibility of subjects exposed to the SARS-CoV-2 infection toward more severe clinical outcomes [268]. Relevantly, even though COVID-19 and AD have different clinical presentations, there are multiple, neurological, psychiatric and ophthalmological, physiopathological aspects linking with each other and increasing the patients’ complications and mortality.
The cellular mechanisms underlying the COVID-19-induced cognitive impairment and visual deficits mainly include the SARS-CoV-2 neurotropism to the CNS and the eyes as a potential route of the virus’s invasion of the brain [269,270]. In addition, several common risk factors such as excessive neuroinflammation, ACE2 expression, APOE4 genotype, age, oxidative stress, hypoxia, neurotransmitter system, the activation of signaling pathways associated with APP/Aβ and tau pathologies provide solid neurobiochemical correlates for reciprocal associations between COVID-19 and AD [271]. In addition, the “inflamm-aging” not only predisposes one to the SARS-CoV-2 infection but also reduces the antibody response to vaccinations, reinforcing COVID-19 as a risk factor in developing cognitive impairments and dementia in frail and elderly patients [272,273]. Clinical follow-up studies with the intent of evaluating the extent and the duration of cognitive impairment in large cohorts of COVID-19 patients along with further experimental investigations on SARS-CoV-2-infected human brain organoids possibly recapitulating the phenotypic expression of key AD hallmarks are still needed. From a translational point of view, the concerted effort from clinicians, researchers, patients, caregivers and health and social care agencies, in association with a deep understanding of the biological aspects linking COVD-19 and AD physiopathologies, will help in designing specific diagnostic/therapeutic strategies [274,275], in order to mitigate the impact of long-lasting neurological and ophthalmological COVID-19 complications in the aging population.

Author Contributions

Conceptualization, G.A. and A.M.; writing—original draft, G.A., V.L., E.S. and A.M. All authors have read and agreed to the published version of the manuscript.

Funding

G.A. was supported by Regione Lazio, POR FESR Lazio 2014–2020. “Progetti di Gruppi di Ricerca 2020” (Determinazione dirigenziale n.G08487 del 19 luglio 2020) Project T0002E0001 G04014_13_04_2021 and Alzheimer’s Association Research Grant—Proposal ID: 971925 and Ministry of Health Project RF-2021-12374301 and PNRR (National Plan for Recovery and Resilience Next Generation EU)—PE Neuroscience project-MNESYS A multiscale integrated approach to the study of the nervous system in health and disease. This work was also supported (in part) by Fondo Ordinario Enti (FOE D.M865/2019) funds in the framework of a collaboration agreement between the Italian National Research Council and EBRI. A.M. was supported by Fondazione Roma and Italian Ministry of Health (Ricerca Corrente) (Italy). The funders had no role in the study design, data collection and analysis in the decision to publish or in the preparation of the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Breijyeh, Z.; Karaman, R. Comprehensive review on alzheimer’s disease: Causes and treatment. Molecules 2020, 25, 5789. [Google Scholar] [CrossRef] [PubMed]
  2. Scheltens, P.; De Strooper, B.; Kivipelto, M.; Holstege, H.; Chételat, G.; Teunissen, C.E.; Cummings, J.; van der Flier, W.M. Alzheimer’s disease. Lancet 2021, 397, 1577–1590. [Google Scholar] [CrossRef] [PubMed]
  3. Bchetnia, M.; Girard, C.; Duchaine, C.; Laprise, C. The outbreak of the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2): A review of the current global status. J. Infect. Public Health 2020, 13, 1601–1610. [Google Scholar] [CrossRef]
  4. Hiscott, J.; Alexandridi, M.; Muscolini, M.; Tassone, E.; Palermo, E.; Soultsioti, M.; Zevini, A. The global impact of the coronavirus pandemic. Cytokine Growth Factor Rev. 2020, 53, 1–9. [Google Scholar] [CrossRef] [PubMed]
  5. Larsen, J.R.L.; Martin, M.R.; Martin, J.D.; Kuhn, P.; Hicks, J.B. Modeling the onset of symptoms of COVID-19. Front. Public Health 2020, 8, 473. [Google Scholar] [CrossRef] [PubMed]
  6. Ellul, M.A.; Benjamin, L.; Singh, B.; Lant, S.; Michael, B.D.; Easton, A.; Kneen, R.; Defres, S.; Sejvar, J.; Solomon, T. Neurological associations of COVID-19. Lancet Neurol. 2020, 19, 767–783. [Google Scholar] [CrossRef]
  7. Hampshire, A.; Trender, W.; Chamberlain, S.R.; Jolly, A.E.; Grant, J.E.; Patrick, F.; Mazibuko, N.; Williams, S.C.; Barnby, J.M.; Hellyer, P.; et al. Cognitive deficits in people who have recovered from COVID-19. eClinicalMedicine 2021, 39, 101044. [Google Scholar] [CrossRef]
  8. Davis, H.E.; McCorkell, L.; Vogel, J.M.; Topol, E.J. Long COVID: Major findings, mechanisms and recommendations. Nat. Rev. Microbiol. 2023, 21, 133–146. [Google Scholar] [CrossRef]
  9. Nalbandian, A.; Sehgal, K.; Gupta, A.; Madhavan, M.V.; McGroder, C.; Stevens, J.S.; Cook, J.R.; Nordvig, A.S.; Shalev, D.; Sehrawat, T.S.; et al. Post-acute COVID-19 syndrome. Nat. Med. 2021, 27, 601–615. [Google Scholar] [CrossRef]
  10. Proal, A.D.; VanElzakker, M.B. Long COVID or Post-acute Sequelae of COVID-19 (PASC): An Overview of Biological Factors That May Contribute to Persistent Symptoms. Front. Microbiol. 2021, 12, 698169. [Google Scholar] [CrossRef]
  11. Song, E.; Zhang, C.; Israelow, B.; Lu-Culligan, A.; Vieites Prado, A.; Skriabine, S.; Lu, P.; Weizman, O.E.; Liu, F.; Dai, Y.; et al. Neuroinvasion of SARS-CoV-2 in human and mouse brain. J. Exp. Med. 2021, 218, e20202135. [Google Scholar] [CrossRef]
  12. Harapan, B.N.; Yoo, H.J. Neurological symptoms, manifestations, and complications associated with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and coronavirus disease 19 (COVID-19). J. Neurol. 2021, 268, 3059–3071. [Google Scholar] [CrossRef] [PubMed]
  13. Sharifkashani, S.; Bafrani, M.A.; Khaboushan, A.S.; Pirzadeh, M.; Kheirandish, A.; Bali, H.Y.; Hessami, A.; Saghazadeh, A.; Rezaei, N. Angiotensin-converting enzyme 2 (ACE2) receptor and SARS-CoV-2: Potential therapeutic targeting. Eur. J. Pharmacol. 2020, 884, 173455. [Google Scholar] [CrossRef] [PubMed]
  14. Ackermann, M.; Verleden, S.E.; Kuehnel, M.; Haverich, A.; Welte, T.; Laenger, F.; Vanstapel, A.; Werlein, C.; Stark, H.; Tzankov, A.; et al. Pulmonary vascular endothelialitis, thrombosis, and angiogenesis in COVID-19. N. Engl. J. Med. 2020, 383, 120–128. [Google Scholar] [CrossRef] [PubMed]
  15. Chu, H.; Chan, J.F.; Yuen, T.T.; Shuai, H.; Yuan, S.; Wang, Y.; Hu, B.; Yip, C.C.-Y.; Tsang, J.O.-L.; Huang, X.; et al. Comparative tropism, replication kinetics, and cell damage profiling of SARS-CoV-2 and SARS-CoV with implications for clinical manifestations, transmissibility, and laboratory studies of COVID-19: An observational study. Lancet Microbe 2020, 1, e14–e23. [Google Scholar] [CrossRef]
  16. Sun, S.-H.; Chen, Q.; Gu, H.-J.; Yang, G.; Wang, Y.-X.; Huang, X.-Y.; Liu, S.-S.; Zhang, N.-N.; Li, X.-F.; Xiong, R.; et al. A Mouse Model of SARS-CoV-2 Infection and Pathogenesis. Cell Host Microbe 2020, 28, 124–133.e4. [Google Scholar] [CrossRef] [PubMed]
  17. Von Weyhern, C.H.; Kaufmann, I.; Neff, F.; Kremer, M. Early evidence of pronounced brain involvement in fatal COVID-19 outcomes. Lancet 2020, 395, e109. [Google Scholar] [CrossRef]
  18. Zhang, B.-Z.; Chu, H.; Han, S.; Shuai, H.; Deng, J.; Hu, Y.-F.; Gong, H.-R.; Lee, A.C.-Y.K.; Zou, Z.; Yau, T.; et al. SARS-CoV-2 infects human neural progenitor cells and brain organoids. Cell Res. 2020, 30, 928–931. [Google Scholar] [CrossRef]
  19. Tan, B.-H.; Liu, J.-M.; Gui, Y.; Wu, S.; Suo, J.-L.; Li, Y.-C. Neurological involvement in the respiratory manifestations of COVID-19 patients. Aging 2021, 13, 4713–4730. [Google Scholar] [CrossRef]
  20. Fišar, Z. Linking the Amyloid, Tau, and Mitochondrial Hypotheses of Alzheimer’s Disease and Identifying Promising Drug Targets. Biomolecules 2022, 12, 1676. [Google Scholar] [CrossRef]
  21. Tönnies, E.; Trushina, E. Oxidative Stress, Synaptic Dysfunction, and Alzheimer’s Disease. J. Alzheimers Dis. 2017, 57, 1105–1121. [Google Scholar] [CrossRef]
  22. Bowirrat, A. Immunosenescence and Aging: Neuroinflammation Is a Prominent Feature of Alzheimer’s Disease and Is a Likely Contributor to Neurodegenerative Disease Pathogenesis. J. Pers. Med. 2022, 12, 1817. [Google Scholar] [CrossRef] [PubMed]
  23. Holubiec, M.I.; Gellert, M.; Hanschmann, E.M. Redox signaling and metabolism in Alzheimer’s disease. Front. Aging Neurosci. 2022, 14, 1003721. [Google Scholar] [CrossRef] [PubMed]
  24. Kara, B.; Gordon, M.N.; Gifani, M.; Dorrance, A.M.; Counts, S.E. Vascular and Nonvascular Mechanisms of Cognitive Impairment and Dementia. Clin. Geriatr. Med. 2023, 39, 109–122. [Google Scholar] [CrossRef]
  25. Zhang, H.; Jiang, X.; Ma, L.; Wei, W.; Li, Z.; Chang, S.; Wen, J.; Sun, J.; Li, H. Role of Aβ in Alzheimer’s-related synaptic dysfunction. Front. Cell Dev. Biol. 2022, 10, 964075. [Google Scholar] [CrossRef]
  26. Pacheco-Herrero, M.; Soto-Rojas, L.O.; Harrington, C.R.; Flores-Martinez, Y.M.; Villegas-Rojas, M.M.; León-Aguilar, A.M.; Martínez-Gómez, P.A.; Campa-Córdoba, B.B.; Apátiga-Pérez, R.; Corniel-Taveras, C.N.; et al. Elucidating the Neuropathologic Mechanisms of SARS-CoV-2 Infection. Front. Neurol. 2021, 12, 660087. [Google Scholar] [CrossRef] [PubMed]
  27. Krasemann, S.; Haferkamp, U.; Pfefferle, S.; Woo, M.S.; Heinrich, F.; Schweizer, M.; Appelt-Menzel, A.; Cubukova, A.; Barenberg, J.; Leu, J.; et al. The blood-brain barrier is dysregulated in COVID-19 and serves as a CNS entry route for SARS-CoV-2. Stem Cell Rep. 2022, 17, 307–320. [Google Scholar] [CrossRef]
  28. Karuppan, M.K.M.; Devadoss, D.; Nair, M.; Chand, H.S.; Lakshmana, M.K. SARS-CoV-2 Infection in the Central and Peripheral Nervous System-Associated Morbidities and Their Potential Mechanism. Mol. Neurobiol. 2021, 58, 2465–2480. [Google Scholar] [CrossRef]
  29. Jha, N.K.; Ojha, S.; Jha, S.K.; Dureja, H.; Singh, S.K.; Shukla, S.D.; Chellappan, D.K.; Gupta, G.; Bhardwaj, S.; Kumar, N.; et al. Evidence of Coronavirus (CoV) Pathogenesis and Emerging Pathogen SARS-CoV-2 in the Nervous System: A Review on Neurological Impairments and Manifestations. J. Mol. Neurosci. 2021, 71, 2192–2209. [Google Scholar] [CrossRef]
  30. Dewanjee, S.; Vallamkondu, J.; Kalra, R.S.; Puvvada, N.; Kandimalla, R.; Reddy, P.H. Emerging COVID-19 Neurological Manifestations: Present Outlook and Potential Neurological Challenges in COVID-19 Pandemic. Mol. Neurobiol. 2021, 58, 4694–4715. [Google Scholar] [CrossRef]
  31. Iadecola, C.; Anrather, J.; Kamel, H. Effects of COVID-19 on the Nervous System. Cell 2020, 183, 16–27.e1. [Google Scholar] [CrossRef]
  32. Liu, J.M.; Tan, B.H.; Wu, S.; Gui, Y.; Suo, J.L.; Li, Y.C. Evidence of central nervous system infection and neuroinvasive routes, as well as neurological involvement, in the lethality of SARS-CoV-2 infection. J. Med. Virol. 2021, 93, 1304–1313. [Google Scholar] [CrossRef]
  33. Dolhnikoff, M.; Duarte-Neto, A.N.; Saldiva, P.H.N.; Caldini, E.G. Using EM data to understand COVID-19 pathophysiology. Lancet 2021, 397, 196–197. [Google Scholar] [CrossRef]
  34. Meinhardt, J.; Radke, J.; Dittmayer, C.; Franz, J.; Thomas, C.; Mothes, R.; Laue, M.; Schneider, J.; Brünink, S.; Greuel, S.; et al. Olfactory transmucosal SARS-CoV-2 invasion as a port of central nervous system entry in individuals with COVID-19. Nat. Neurosci. 2021, 24, 168–175. [Google Scholar] [CrossRef]
  35. Al-Dalahmah, O.; Thakur, K.T.; Nordvig, A.S.; Prust, M.L.; Roth, W.; Lignelli, A.; Uhlemann, A.-C.; Happy Miller, E.; Kunnath-Velayudhan, S.; Del Portillo, A.; et al. Neuronophagia and microglial nodules in a SARS-CoV-2 patient with cerebellar hemorrhage. Acta Neuropathol. Commun. 2020, 8, 147. [Google Scholar] [CrossRef]
  36. Schurink, B.; Roos, E.; Radonic, T.; Barbe, E.; Bouman, C.S.C.; De Boer, H.H.; de Bree, G.J.; Bulle, E.B.; Aronica, E.M.; Florquin, S.; et al. Viral presence and immunopathology in patients with lethal COVID-19: A prospective autopsy cohort study. Lancet Microbe 2020, 1, e290–e299. [Google Scholar] [CrossRef] [PubMed]
  37. Matschke, J.; Lutgehetmann, M.; Hagel, C.; Sperhake, J.P.; Schroder, A.S.; Edler, C.; Mushumba, H.; Fitzek, A.; Allweiss, L.; Dandri, M.; et al. Neuropathology of patients with COVID-19 in Germany: A post-mortem case series. Lancet Neurol. 2020, 19, 919–929. [Google Scholar] [CrossRef] [PubMed]
  38. 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]
  39. Kremer, S.; Lersy, F.; Anheim, M.; Merdji, H.; Schenck, M.; Oesterlé, H.; Bolognini, F.; Messie, J.; Khalil, A.; Gaudemer, A.; et al. Neurologic and neuroimaging findings in patients with COVID-19: A retrospective multicenter study. Neurology 2020, 95, e1868–e1882. [Google Scholar] [CrossRef]
  40. Edén, A.; Kanberg, N.; Gostner, J.; Fuchs, D.; Hagberg, L.; Andersson, L.-A.; Lindh, M.; Price, R.W.; Zetterberg, H.; Gisslén, M. CSF Biomarkers in Patients with COVID-19 and Neurologic Symptoms: A Case Series. Neurology 2021, 96, e294–e300. [Google Scholar] [CrossRef]
  41. Destras, G.; Bal, A.; Escuret, V.; Morfin, F.; Lina, B.; Josset, L. COVID-Diagnosis HCL Study Group. Systematic SARS-CoV-2 screening in cerebrospinal fluid during the COVID-19 pandemic. Lancet Microbe 2020, 1, e149. [Google Scholar] [CrossRef] [PubMed]
  42. Espíndola, O.M.; Siqueira, M.; Soares, C.N.; Lima, M.A.S.D.; Leite, A.C.C.B.; Araujo, A.Q.C.; Brandão, C.O.; Silva, M.T.T. Patients with COVID-19 and neurological manifestations show undetectable SARS-CoV-2 RNA levels in the cerebrospinal fluid. Int. J. Infect. Dis. 2020, 96, 567–569. [Google Scholar] [CrossRef]
  43. Lee, M.H.; Perl, D.P.; Steiner, J.; Pasternack, N.; Li, W.; Maric, D.; Safavi, F.; Horkayne-Szakaly, I.; Jones, R.; Stram, M.N.; et al. Neurovascular injury with complement activation and inflammation in COVID-19. Brain 2022, 145, 2555–2568. [Google Scholar] [CrossRef]
  44. Zheng, J.; Roy Wong, L.Y.; Li, K.; Verma, A.K.; Ortiz, M.; Wohlford-Lenane, C.; Leidinger, M.R.; Knudson, C.M.; Meyerholz, D.K.; McCray, P.B.; et al. K18-hACE2 Mice for Studies of COVID-19 Treatments and Pathogenesis Including Anosmia. Nature 2021, 589, 603–607. [Google Scholar] [CrossRef] [PubMed]
  45. Rathnasinghe, R.; Strohmeier, S.; Amanat, F.; Gillespie, V.L.; Krammer, F.; García-Sastre, A.; Coughlan, L.; Schotsaert, M.; Uccellini, M. Comparison of Transgenic and Adenovirus hACE2 Mouse Models for SARS-CoV-2 Infection. Emerg. Microbes Infect. 2020, 9, 2433–2445. [Google Scholar] [CrossRef]
  46. Hamming, I.; Timens, W.; Bulthuis, M.L.C.; Lely, A.T.; Navis, G.J.; van Goor, H. Tissue distribution of ACE2 protein, the functional receptor for SARS coronavirus. A first step in understanding SARS pathogenesis. J. Pathol. 2004, 203, 631–637. [Google Scholar] [CrossRef] [PubMed]
  47. Hoffmann, H.H.; Schneider, W.M.; Rozen-Gagnon, K.; Miles, L.A.; Schuster, F.; Razooky, B.; Jacobson, E.; Wu, X.; Yi, S.; Rudin, C.M.; et al. TMEM41B Is a Pan-flavivirus Host Factor. Cell 2021, 184, 133–148.e20. [Google Scholar] [CrossRef]
  48. Mostafavi, E.; Dubey, A.K.; Teodori, L.; Ramakrishna, S.; Kaushik, A. SARS-CoV-2 Omicron variant: A next phase of the COVID-19 pandemic and a call to arms for system sciences and precision medicine. MedComm 2022, 3, e119. [Google Scholar] [CrossRef]
  49. Cantuti-Castelvetri, L.; Ojha, R.; Pedro, L.D.; Djannatian, M.; Franz, J.; Kuivanen, S.; van der Meer, F.; Kallio, K.; Kaya, T.; Anastasina, M.; et al. Neuropilin-1 facilitates SARS-CoV-2 cell entry and infectivity. Science 2020, 370, 856–860. [Google Scholar] [CrossRef]
  50. Baig, A.M.; Khaleeq, A.; Ali, U.; Syeda, H. Evidence of the COVID-19 Virus Targeting the CNS: Tissue Distribution, Host-Virus Interaction, and Proposed Neurotropic Mechanisms. ACS Chem. Neurosci. 2020, 11, 995–998. [Google Scholar] [CrossRef]
  51. Bergmann, C.C.; Lane, T.E.; Stohlman, S.A. Coronavirus infection of the central nervous system: Host-virus stand-off. Nat. Rev. Microbiol. 2006, 4, 121–132. [Google Scholar] [CrossRef]
  52. Chen, F.; Chen, Y.; Wang, Y.; Qiongwei Ke, Q.; Cui, L. The COVID-19 pandemic and Alzheimer’s disease: Mutual risks and mechanisms. Transl. Neurodegener. 2022, 11, 40. [Google Scholar] [CrossRef] [PubMed]
  53. Kujawska, M.; Mostafavi, E.; Kaushik, A. SARS-CoV-2 getting into the brain; neurological phenotype of COVID-19, and management by nano-biotechnology. Neural Regen. Res. 2023, 18, 519–520. [Google Scholar] [CrossRef] [PubMed]
  54. Lima, M.; Siokas, V.; Aloizou, A.-M.; Liampas, I.; Mentis, A.-F.A.; Tsouris, Z.; Papadimitriou, A.; Mitsias, P.D.; Tsatsakis, A.; Bogdanos, D.P.; et al. Unraveling the Possible Routes of SARS-CoV-2 Invasion into the Central Nervous System. Curr. Treat. Options Neurol. 2020, 22, 37. [Google Scholar] [CrossRef]
  55. Ur, A.; Verma, K. Cytokine Storm in COVID-19: A Neural Hypothesis. ACS Chem. Neurosci. 2020, 11, 1868–1870. [Google Scholar] [CrossRef] [PubMed]
  56. MacLean, M.A.; Kamintsky, L.; Leck, E.D.; Friedman, A. The potential role of microvascular pathology in the neurological manifestations of coronavirus infection Fluids Barriers. Fluids Barriers CNS 2020, 17, 55. [Google Scholar] [CrossRef]
  57. Desforges, M.; Le Coupanec, A.; Stodola, J.K.; Meessen-Pinard, M.; Talbot, P.J. Human coronaviruses: Viral and cellular factors involved in neuroinvasiveness and neuropathogenesis. Virus Res. 2014, 194, 145–158. [Google Scholar] [CrossRef]
  58. Fenrich, M.; Mrdenovic, S.; Balog, M.; Tomic, S.; Zjalic, M.; Roncevic, A.; Mandic, D.; Debeljak, Z.; Heffer, M. SARS-CoV-2 Dissemination through Peripheral Nerves Explains Multiple Organ Injury. Front. Cell. Neurosci. 2020, 14, 229. [Google Scholar] [CrossRef]
  59. Perrottelli, A.; Sansone, N.; Giordano, G.M.; Caporusso, E.; Giuliani, L.; Melillo, A.; Pezzella, P.; Bucci, P.; Mucci, A.; Galderisi, S. Cognitive Impairment after Post-Acute COVID-19 Infection: A Systematic Review of the Literature. J. Pers. Med. 2022, 12, 2070. [Google Scholar] [CrossRef]
  60. Miners, S.; Kehoe, P.G.; Love, S. Cognitive impact of COVID-19: Looking beyond the short term. Alzheimers Res. Ther. 2020, 12, 170. [Google Scholar] [CrossRef]
  61. Ceban, F.; Ling, S.; Lui, L.M.W.; Lee, Y.; Gill, H.; Teopiz, K.M.; Rodrigues, N.B.; Subramaniapillai, M.; Di Vincenzo, J.D.; Cao, B.; et al. Fatigue and cognitive impairment in Post-COVID-19 Syndrome: A systematic review and meta-analysis. Brain Behav. Immun. 2022, 101, 93–135. [Google Scholar] [CrossRef]
  62. Crivelli, L.; Palmer, K.; Calandri, I.; Guekht, A.; Beghi, E.; Carroll, W.; Frontera, J.; García-Azorín, D.; Westenberg, E.; Winkler, A.S.; et al. Changes in cognitive functioning after COVID-19: A systematic review and meta-analysis. Alzheimers Dement. 2022, 18, 1047–1066. [Google Scholar] [CrossRef]
  63. Verkhratsky, A.; Li, Q.; Melino, S.; Melino, G.; Shi, Y. Can COVID-19 pandemic boost the epidemic of neurodegenerative diseases? Biol. Direct. 2020, 15, 28. [Google Scholar] [CrossRef] [PubMed]
  64. Barbieri, M.A.; Bagnato, G.; Ioppolo, C.; Versace, A.G.; Irrera, N. Impact of the COVID-19 Pandemic on Chronic Neurological Disorders: Focus on Patients with Dementia. CNS Neurol. Disord. Drug Targets 2022, 21, 1017–1026. [Google Scholar] [CrossRef] [PubMed]
  65. Wang, L.; He, W.; Yu, X.; Hu, D.; Bao, M.; Li, H.; Zhou, J.; Jiang, H. Coronavirus disease 2019 in elderly patients: Characteristics and prognostic factors based on4-week follow-up. J. Infect. 2020, 80, 639–645. [Google Scholar] [CrossRef]
  66. de Erausquin, G.A.; Snyder, H.; Brugha, T.S.; Seshadri, S.; Carrillo, M.; Sagar, R.; Huang, Y.; Newton, C.; Tartaglia, C.; Teunissen, C.; et al. Chronic neuropsychiatric sequelae of SARS-CoV-2: Protocol and methods from the Alzheimer’s Association Global Consortium. Alzheimers Dement. 2022, 8, e12348. [Google Scholar] [CrossRef] [PubMed]
  67. Golzari-Sorkheh, M.; Weaver, D.F.; Reed, M.A. COVID-19 as a Risk Factor for Alzheimer’s Disease. J. Alzheimers Dis. 2023, 91, 1–23. [Google Scholar] [CrossRef]
  68. Olivera, E.; Sáez, A.; Carniglia, L.; Caruso, C.; Lasaga, M.; Durand, D. Alzheimer’s disease risk after COVID-19: A view from the perspective of the infectious hypothesis of neurodegeneration. Neural Regen. Res. 2023, 18, 1404–1410. [Google Scholar] [CrossRef] [PubMed]
  69. Ferini-Strambi, L.; Salsone, M. COVID-19 and neurological disorders: Are neurodegenerative or neuroimmunological diseases more vulnerable? J. Neurol. 2021, 268, 409–419. [Google Scholar] [CrossRef]
  70. Daroische, R.; Hemminghyth, M.S.; Eilertsen, T.H.; Breitve, M.H.; Chwiszczuk, L.J. Cognitive Impairment after COVID-19—A Review on Objective Test Data. Front. Neurol. 2021, 12, 699582. [Google Scholar] [CrossRef]
  71. Numbers, K.; Brodaty, H. The effects of the COVID-19 pandemic on people with dementia. Nat. Rev. Neurol. 2021, 17, 69–70. [Google Scholar] [CrossRef] [PubMed]
  72. Magusali, N.; Graham, A.C.; Piers, T.M.; Panichnantakul, P.; Yaman, U.; Shoai, M.; Reynolds, R.H.; Botia, J.A.; Brookes, K.J.; Guetta-Baranes, T.; et al. A genetic link between risk for Alzheimer’s disease and severe COVID-19 outcomes via the OAS1 gene. Brain 2021, 144, 3727–3741. [Google Scholar] [CrossRef] [PubMed]
  73. Hariyanto, T.I.; Putri, C.; Arisa, J.; Situmeang, R.F.V.; Kurniawan, A. Dementia and outcomes from coronavirus disease 2019 (COVID-19) pneumonia: A systematic review and meta-analysis. Arch. Gerontol. Geriatr. 2021, 93, 104299. [Google Scholar] [CrossRef] [PubMed]
  74. Tahira, A.C.; Verjovski-Almeida, S.; Ferreira, S.T. Dementia is an age-independent risk factor for severity and death in COVID-19 in patients. Alzheimers Dement. 2021, 17, 1818–1831. [Google Scholar] [CrossRef] [PubMed]
  75. Kim, Y.J.; Jee, Y.; Park, S.; Ha, E.H.; Jo, I.; Lee, H.W.; Song, M.S. Mortality risk within 14 days after coronavirus disease 2019 diagnosis in dementia patients: A nation wide analysis. Dement. Geriatr. Cogn. Disord. 2021, 50, 425–436. [Google Scholar] [CrossRef] [PubMed]
  76. Wang, S.M.; Park, S.H.; Kim, N.Y.; Kang, D.W.; Na, H.R.; Um, Y.H.; Han, S.; Park, S.S.; Lim, H.K. Association between dementia and clinical outcome after COVID-19: A nationwide cohort study with propensity score matched control in South Korea. Psychiatry Investig. 2021, 18, 523–529. [Google Scholar] [CrossRef]
  77. Chung, S.J.; Chang, Y.; Jeon, J.; Shin, J.I.; Song, T.-J.; Kim, J. Association of Alzheimer’s Disease with COVID-19 Susceptibility and Severe Complications: A Nationwide Cohort Study. J. Alzheimers Dis. 2022, 87, 701–710. [Google Scholar] [CrossRef]
  78. Wang, F.; Kream, R.M.; Stefano, G.B. Long-Term Respiratory and Neurological Sequelae of COVID-19. Med. Sci. Monit. 2020, 26, e928996. [Google Scholar] [CrossRef]
  79. Wang, L.; Davis, P.B.; Volkow, N.D.; Berger, N.A.; Kaelber, D.C.; Xu, R. Association of COVID-19 with with new-onset Alzheimer’s disease. J. Alzheimers Dis. 2022, 89, 411–414. [Google Scholar] [CrossRef]
  80. Covino, M.; De Matteis, G.; Santoro, M.; Sabia, L.; Simeoni, B.; Candelli, M.; Ojetti, V.; Franceschi, F. Clinical characteristics and prognostic factors in COVID-19 patients aged ≥80 years. Geriatr. Gerontol. Int. 2020, 20, 704–708. [Google Scholar] [CrossRef]
  81. Bianchetti, A.; Rozzini, R.; Guerini, F.; Bofelli, S.; Ranieri, P.; Minelli, G.; Bianchetti, L.; Trabucchi, M. Clinical presentation of COVID-19 in dementia patients. J. Nutr. Health Aging 2020, 24, 560–562. [Google Scholar] [CrossRef] [PubMed]
  82. Negrini, F.; Ferrario, I.; Mazziotti, D.; Berchicci, M.; Bonazzi, M.; de Sire, A.; Negrini, S.; Zapparoli, L. Neuropsychological features of severe hospitalized coronavirus disease 2019 patients at clinical stability and clues for postacute rehabilitation. Arch. Phys. Med. Rehabil. 2021, 102, 155–158. [Google Scholar] [CrossRef]
  83. De Lorenzo, R.; Conte, C.; Lanzani, C.; Benedetti, F.; Roveri, L.; Mazza, M.G.; Brioni, E.; Giacalone, G.; Canti, V.; Sofia, V.; et al. Residual clinical damage after COVID-19: A retrospective and prospective observational cohort study. PLoS ONE 2020, 15, e0239570. [Google Scholar] [CrossRef] [PubMed]
  84. Beaud, V.; Crottaz-Herbette, S.; Dunet, V.; Vaucher, J.; Bernard-Valnet, R.; Du Pasquier, R.; Bart, P.-A.; Clarke, S. Pattern of cognitive deficits in severe COVID-19. J. Neurol. Neurosurg. Psychiatry 2021, 92, 567–568. [Google Scholar] [CrossRef] [PubMed]
  85. van den Borst, B.; Peters, J.B.; Brink, M.; Schoon, Y.; Bleeker-Rovers, C.P.; Schers, H.; van Hees, H.W.H.; van Helvoort, H.; van den Boogaard, M.; van der Hoeven, H.; et al. Comprehensive health assessment three months after recovery from acute COVID-19. Clin. Infect. Dis. 2021, 73, e1089–e1098. [Google Scholar] [CrossRef]
  86. Almeria, M.; Cejudo, J.C.; Sotoca, J.; Deus, J.; Krupinski, J. Cognitive profile following COVID-19 infection: Clinical predictors leading to neuropsychological impairment. Brain Behav. Immun. Health 2020, 9, 100163. [Google Scholar] [CrossRef]
  87. Woo, M.S.; Malsy, J.; Pottgen, J.; Seddiq Zai, S.; Ufer, F.; Hadjilaou, A.; Schmiedel, S.; Addo, M.M.; Gerloff, C.; Heesen, C.; et al. Frequent neurocognitive deficits after recovery from mild COVID-19. Brain Commun. 2020, 2, fcaa205. [Google Scholar] [CrossRef]
  88. Zhou, H.; Lu, S.; Chen, J.; Wei, N.; Wang, D.; Lyu, H.; Shi, C.; Hu, S. The landscape of cognitive function in recovered COVID-19 patients. J. Psychiatr. Res. 2020, 129, 98–102. [Google Scholar] [CrossRef]
  89. Rass, V.; Beer, R.; Schiefecker, A.J.; Lindner, A.; Kofler, M.; Ianosi, B.A.; Mahlknecht, P.; Heim, B.; Peball, M.; Carbone, F.; et al. Neurological outcomes 1 year after COVID-19diagnosis: A prospective longitudinal cohort study. Eur. J. Neurol. 2022, 29, 1685–1696. [Google Scholar] [CrossRef]
  90. Søraas, A.; Bø, R.; Kalleberg, K.T.; Støer, N.C.; Ellingjord-Dale, M.; Landrø, N.I. Self-reported memory problems 8 months after COVID-19 infection. JAMA Netw. Open 2021, 4, e2118717. [Google Scholar] [CrossRef]
  91. Douaud, G.; Lee, S.; Alfaro-Almagro, F.; Arthofer, C.; Wang, C.; McCarthy, P.; Lange, F.; Andersson, J.L.R.; 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]
  92. Fernández-Castañeda, A.; Lu, P.; Geraghty, A.C.; Song, E.; Lee, M.-H.; Wood, J.; O’Dea, M.R.; Dutton, S.; Shamardani, K.; Nwangwu, K.; et al. Mild respiratory COVID can cause multi-lineage neural cell and myelin dysregulation. Cell 2022, 185, 2452–2468.e16. [Google Scholar] [CrossRef] [PubMed]
  93. Charnley, M.; Islam, S.; Bindra, G.B.; 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-CoV2: Potential implications for neurological symptoms in COVID-19. Nat. Commun. 2022, 13, 3387. [Google Scholar] [CrossRef] [PubMed]
  94. de Melo, G.D.; Lazarini, F.; Levallois, S.; Hautefort, C.; Michel, V.; Larrous, F.; Verillaud, B.; Aparicio, C.; Wagner, S.; Gheusi, G.; et al. COVID-19-related anosmia is associated with viral persistence and inflammation in human olfactory epithelium and brain infection in hamsters. Sci. Transl. Med. 2021, 13, eabf8396. [Google Scholar] [CrossRef] [PubMed]
  95. Sodagar, A.; Javed, R.; Tahir, H.; Razak, S.I.A.; Shakir, M.; Naeem, M.; Yusof, A.H.A.; Sagadevan, S.; Hazafa, A.; Uddin, J.; et al. Pathological Features and Neuroinflammatory Mechanisms of SARS-CoV-2 in the Brain and Potential Therapeutic Approaches. Biomolecules 2022, 12, 971. [Google Scholar] [CrossRef]
  96. Mohammadi, S.; Gouravani, M.; Salehi, M.A.; Harandi, H.; Moosaie, F.; Firouzabadi, F.D.; Yousem, D.M. Olfactory system measurements in COVID-19: A systematic review and meta-analysis. Neuroradiology 2023, 65, 25–39. [Google Scholar] [CrossRef]
  97. Ho, C.-Y.; Salimian, M.; Hegert, J.; O’Brien, J.; Choi, S.G.; Ames, H.; Morris, M.; Papadimitriou, J.C.; Mininni, J.; Niehaus, P.; et al. Postmortem Assessment of Olfactory Tissue Degeneration and Microvasculopathy in Patients with COVID-19. JAMA Neurol. 2022, 79, 544–553. [Google Scholar] [CrossRef]
  98. Kay, L.M. COVID-19 and olfactory dysfunction: A looming wave of dementia? J. Neurophysiol. 2022, 128, 436–444. [Google Scholar] [CrossRef]
  99. Ziuzia-Januszewska, L.; Januszewski, M. Pathogenesis of Olfactory Disorders in COVID-19. Brain Sci. 2022, 12, 449. [Google Scholar] [CrossRef]
  100. Tu, L.; Lv, X.; Fan, Z.; Zhang, M.; Wang, H.; Yu, X. Association of odor identification ability with amyloid-β and tau burden: A systematic review and meta-analysis. Front. Neurosci. 2020, 14, 586330. [Google Scholar] [CrossRef]
  101. Guedj, E.; Campion, J.Y.; Horowitz, T.; Barthelemy, F.; Cammilleri, S.; Ceccaldi, M. The impact of COVID-19 lockdown on brain metabolism. Hum. Brain Mapp. 2022, 43, 593–597. [Google Scholar] [CrossRef]
  102. Hugon, J.; Msika, E.F.; Queneau, M.; Farid, K.; Paquet, C. Long COVID: Cognitive complaints (brain fog) and dysfunction of the cingulate cortex. J. Neurol. 2022, 269, 44–46. [Google Scholar] [CrossRef]
  103. Plangár, I.; Zádori, D.; Klivényi, P.; Toldi, J.; Vécsei, L. Targeting the kynurenine pathway-related alterations in Alzheimer’s disease: A future therapeutic strategy. J. Alzheimers Dis. 2011, 24, 199–209. [Google Scholar] [CrossRef] [PubMed]
  104. Cysique, L.A.; Jakabek, D.; Bracken, S.G.; Allen-Davidian, Y.; Heng, B.; Chow, S.; Dehhaghi, M.; Staats Pires, A.; Darley, D.R.; Byrne, A.; et al. The kynurenine pathway relates to post-acute COVID-19 objective cognitive impairment and PASC. Ann. Clin. Transl. Neurol. 2023, 10, 1338–1352. [Google Scholar] [CrossRef] [PubMed]
  105. Wang, W.; Zhao, F.; Ma, X.; Perry, G.; Zhu, X. Mitochondria dysfunction in the pathogenesis of Alzheimer’s disease: Recent advances. Mol. Neurodegener. 2020, 15, 30. [Google Scholar] [CrossRef] [PubMed]
  106. Ashleigh, T.; Swerdlow, R.H.; Beal, M.F. The role of mitochondrial dysfunction in Alzheimer’s disease pathogenesis. Alzheimers Dement. 2023, 19, 333–342. [Google Scholar] [CrossRef] [PubMed]
  107. Peluso, M.J.; Deeks, S.G.; Mustapic, M.; Kapogiannis, D.; Henrich, T.J.; Lu, S.; Goldberg, S.A.; Hoh, R.; Chen, J.Y.; Martinez, E.O.; et al. SARS-CoV-2 and Mitochondrial Proteins in Neural-Derived Exosomes of COVID-19. Ann. Neurol. 2022, 91, 772–781. [Google Scholar] [CrossRef]
  108. Xia, X.; Wang, Y.; Zheng, J. COVID-19 and Alzheimer’s disease: How one crisis worsens the other. Transl. Neurodegener. 2021, 10, 15. [Google Scholar] [CrossRef]
  109. Barrantes, F.J. Central Nervous System Targets and Routes for SARS-CoV-2: Current Views and New Hypotheses. ACS Chem. Neurosci. 2020, 11, 2793–2803. [Google Scholar] [CrossRef]
  110. Lukiw, W.J.; Pogue, A.; Hill, J.M. SARS-CoV-2 Infectivity and Neurological Targets in the Brain. Cell Mol. Neurobiol. 2022, 42, 217–224. [Google Scholar] [CrossRef]
  111. Seaks, C.E.; Wilcock, D.M. Infectious hypothesis of Alzheimer disease. PLoS Pathog. 2020, 16, e1008596. [Google Scholar] [CrossRef] [PubMed]
  112. Rudnicka-Drożak, E.; Drożak, P.; Mizerski, G.; Zaborowski, T.; Ślusarska, 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]
  113. Lim, K.-H.; Yang, S.; Kim, S.H.; Joo, J.Y. Elevation of ACE2 as a SARS-CoV-2 entry receptor gene expression in Alzheimer’s disease. J. Infect. 2020, 81, e33–e34. [Google Scholar] [CrossRef]
  114. Ding, Q.; Shults, N.V.; Gychka, S.G.; Harris, B.T.; Suzuki, Y.J. Protein Expression of Angiotensin-Converting Enzyme 2 (ACE2) is Upregulated in Brains with Alzheimer’s Disease. Int. J. Mol. Sci. 2021, 22, 1687. [Google Scholar] [CrossRef]
  115. Louise, R.; Manon, L.; Vincent, E.; Cyntia, T.; Andréanne, L.; Philippe, B.; David, A.B.; Hébert, S.S.; Frédéric, C. Higher angiotensin-converting enzyme 2 (ACE2) levels in the brain of individuals with Alzheimer’s disease. Acta Neuropathol. Commun. 2023, 11, 159. [Google Scholar] [CrossRef]
  116. 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] [PubMed]
  117. Motaghinejad, M.; Gholami, M. Possible Neurological and Mental Outcomes of COVID-19 Infection: A Hypothetical Role of ACE-2\Mas\BDNF Signaling Pathway. Int. J. Prev. Med. 2020, 11, 84. [Google Scholar] [CrossRef] [PubMed]
  118. Hou, Y.; Dan, X.; Babbar, M.; Wei, Y.; Hasselbalch, S.G.; Croteau, D.L.; Bohr, V.A. Ageing as a risk factor for neurodegenerative disease. Nat. Rev. Neurol. 2019, 15, 565–581. [Google Scholar] [CrossRef]
  119. Saez-Atienzar, S.; Masliah, E. Cellular senescence and Alzheimer disease: The egg and the chicken scenario. Nat. Rev. Neurosci. 2020, 21, 433–444. [Google Scholar] [CrossRef] [PubMed]
  120. Azam, S.; Haque, M.E.; Balakrishnan, R.; Kim, I.S.; Choi, D.K. The Ageing Brain: Molecular and Cellular Basis of Neurodegeneration. Front. Cell Dev. Biol. 2021, 9, 683459. [Google Scholar] [CrossRef]
  121. Behfar, Q.; Ramirez Zuniga, A.; Martino-Adami, P.V. Aging, Senescence, and Dementia. J. Prev. Alzheimers Dis. 2022, 9, 523–531. [Google Scholar] [CrossRef] [PubMed]
  122. Liu, R.M. Aging, Cellular Senescence, and Alzheimer’s Disease. Int. J. Mol. Sci. 2022, 23, 1989. [Google Scholar] [CrossRef] [PubMed]
  123. Sahu, M.R.; Rani, L.; Subba, R.; Mondal, A.C. Cellular senescence in the aging brain: A promising target for neurodegenerative diseases. Mech. Ageing Dev. 2022, 204, 111675. [Google Scholar] [CrossRef]
  124. Lee, S.; Yu, Y.; Trimpert, J.; Benthani, F.; Mairhofer, M.; Richter-Pechanska, P.; Wyler, E.; Belenki, D.; Kaltenbrunner, S.; Pammer, M.; et al. Virus-induced senescence is a driver and therapeutic target in COVID-19. Nature 2021, 599, 283–289. [Google Scholar] [CrossRef] [PubMed]
  125. Schmitt, C.A.; Tchkonia, T.; Niedernhofer, L.J.; Robbins, P.D.; Kirkland, J.L.; Lee, S. COVID-19 and cellular senescence. Nat. Rev. Immunol. 2023, 23, 251–263. [Google Scholar] [CrossRef] [PubMed]
  126. Nehme, J.; Borghesan, M.; Mackedenski, S.; Bird, T.G.; Demaria, M. Cellular senescence as a potential mediator of COVID-19 severity in the elderly. Aging Cell. 2020, 19, e13237. [Google Scholar] [CrossRef]
  127. Bajaj, V.; Gadi, N.; Spihlman, A.P.; Wu, S.C.; Choi, C.H.; Moulton, V.R. Aging, Immunity, and COVID-19: How Age Influences the Host Immune Response to Coronavirus Infections? Front. Physiol. 2021, 11, 571416. [Google Scholar] [CrossRef]
  128. Meftahi, G.H.; Jangravi, Z.; Sahraei, H.; Bahari, Z. The possible pathophysiology mechanism of cytokine storm in elderly adults with COVID-19 infection: The contribution of “inflame-aging”. Inflamm. Res. 2020, 69, 825–839. [Google Scholar] [CrossRef]
  129. Chen, Y.; Klein, S.L.; Garibaldi, B.T.; Li, H.; Wu, C.; Osevala, N.M.; Li, T.; Margolick, J.B.; Pawelec, G.; Leng, S.X. Aging in COVID-19: Vulnerability, immunity and intervention. Ageing Res. Rev. 2021, 65, 101205. [Google Scholar] [CrossRef]
  130. Lynch, S.M.; Guo, G.; Gibson, D.S.; Bjourson, A.J.; Rai, T.S. Role of Senescence and Aging in SARS-CoV-2 Infection and COVID-19 Disease. Cells 2021, 10, 3367. [Google Scholar] [CrossRef]
  131. Qin, C.; Zhou, L.; Hu, Z.; Zhang, S.; Yang, S.; Tao, Y.; Tian, D.S. Dysregulation of immune response in patients with COVID-19 in Wuhan, China. Clin. Infect. Dis. 2020, 71, 762–768. [Google Scholar] [CrossRef]
  132. Ulhaq, Z.S.; Soraya, G.V. Interleukin-6 as a potential biomarker of COVID-19 progression. Med. Mal. Infect. 2020, 50, 382–383. [Google Scholar] [CrossRef] [PubMed]
  133. Yu, H.-J.; Koh, S.-H. Is Telomere Length Shortening a Risk Factor for Neurodegenerative Disorders? Dement. Neurocogn. Disord. 2022, 21, 83–92. [Google Scholar] [CrossRef] [PubMed]
  134. Sanchez-Vazquez, R.; Guío-Carrión, A.; Zapatero-Gaviria, A.; Martínez, P.; Blasco, M.A. Shorter telomere lengths in patients with severe COVID-19 disease. Aging 2021, 13, 1–15. [Google Scholar] [CrossRef]
  135. Bunyavanich, S.; Do, A.; Vicencio, A. Nasal Gene Expression of Angiotensin-Converting Enzyme 2 in Children and Adults. JAMA 2020, 323, 2427–2429. [Google Scholar] [CrossRef]
  136. Kinney, J.W.; Bemiller, S.M.; Murtishaw, A.S.; Leisgang, A.M.; Salazar, A.M.; Lamb, B.T. Inflammation as a central mechanism in Alzheimer’s disease. Alzheimers Dement. 2018, 4, 575–590. [Google Scholar] [CrossRef] [PubMed]
  137. Andronie-Cioara, F.L.; Ardelean, A.I.; Nistor-Cseppento, C.D.; Jurcau, A.; Jurcau, M.C.; Pascalau, N.; Marcu, F. Molecular Mechanisms of Neuroinflammation in Aging and Alzheimer’s Disease Progression. Int. J. Mol. Sci. 2023, 24, 1869. [Google Scholar] [CrossRef]
  138. Wang, H.; Shen, Y.; Chuang, H.; Chiu, C.; Ye, Y.; Zhao, L. Neuroinflammation in Alzheimer’s Disease: Microglia, Molecular Participants and Therapeutic Choices. Curr. Alzheimer Res. 2019, 16, 659–674. [Google Scholar] [CrossRef]
  139. Maccioni, R.B.; Navarrete, L.P.; González, A.; González-Canacer, A.; Guzmán-Martínez, L.; Cortés, N. Inflammation: A Major Target for Compounds to Control Alzheimer’s Disease. J. Alzheimers Dis. 2020, 76, 1199–1213. [Google Scholar] [CrossRef]
  140. Thakur, K.T.; Miller, E.H.; Glendinning, M.D.; Al-Dalahmah, O.; Banu, M.A.; Boehme, A.K.; Boubour, A.; Bruce, S.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]
  141. Schwabenland, M.; Salie, H.; Tanevski, J.; Killmer, S.; Lago, M.S.; Schlaak, A.E.; Mayer, L.; Matschke, J.; Püschel, K.; Fitzek, A.; et al. Deep spatial profiling of human COVID-19 brains reveals neuroinflammation with distinct microanatomical microglia-T-cell interactions. Immunity 2021, 54, 1594–1610.e11. [Google Scholar] [CrossRef]
  142. Rahman, M.A.; Islam, K.; Rahman, S.; Alamin, M. Neurobiochemical Cross-Talk between COVID-19 and Alzheimer’s Disease. Mol. Neurobiol. 2021, 58, 1017–1023. [Google Scholar] [CrossRef] [PubMed]
  143. Cai, Z.; Hussain, M.D.; Yan, L.J. Microglia, neuroinflammation, and beta-amyloid protein in Alzheimer’s disease. Int. J. Neurosci. 2014, 124, 307–321. [Google Scholar] [CrossRef] [PubMed]
  144. Savelieff, M.G.; Feldman, E.L.; Stino, E.L. Neurological sequela and disruption of neuron-glia homeostasis in SARS-CoV-2 infection. Neurobiol. Dis. 2022, 68, 105715. [Google Scholar] [CrossRef] [PubMed]
  145. Albornoz, E.A.; Amarilla, A.A.; Modhiran, N.; Parker, S.; Li, X.X.; Wijesundara, D.K.; Aguado, J.; Pliego Zamora, A.; McMillan, C.L.D.; Liang, B.; et al. SARS-CoV-2 drives NLRP3 inflammasome activation in human microglia through spike protein. Mol. Psychiatry 2023, 28, 2878–2893. [Google Scholar] [CrossRef]
  146. Tejera, D.; Mercan, D.; Sanchez-Caro, J.M.; Hanan, M.; Greenberg, D.; Soreq, H.; Latz, E.; Golenbock, D.; Heneka, M.T. Systemic inflammation impairs microglial Aβ clearance through NLRP3 inflammasome. EMBO J. 2019, 38, e101064. [Google Scholar] [CrossRef]
  147. Ising, C.; Venegas, C.; Zhang, S.; Scheiblich, H.; Schmidt, S.V.; Vieira-Saecker, A.; Schwartz, S.; Albasset, S.; McManus, R.M.; Tejera, D.; et al. NLRP3 inflammasome activation drives tau pathology. Nature 2019, 575, 669–673. [Google Scholar] [CrossRef]
  148. Sharma, C.; Kim, S.R. Linking Oxidative Stress and Proteinopathy in Alzheimer’s Disease. Antioxidants 2021, 10, 1231. [Google Scholar] [CrossRef]
  149. Kozlov, E.M.; Ivanova, E.; Grechko, A.V.; Wu, W.K.; Starodubova, A.V.; Orekhov, A.N. Involvement of Oxidative Stress and the Innate Immune System in SARS-CoV-2 Infection. Diseases 2021, 9, 17. [Google Scholar] [CrossRef]
  150. Choe, K.; Park, H.Y.; Ikram, M.; Lee, H.J.; Park, T.J.; Ullah, R.; Kim, M.O. Systematic Review of the Common Pathophysiological Mechanisms in COVID-19 and Neurodegeneration: The Role of Bioactive Compounds and Natural Antioxidants. Cells 2022, 11, 1298. [Google Scholar] [CrossRef]
  151. Golonka, R.M.; Saha, P.; Yeoh, B.S.; Chattopadhyay, S.; Gewirtz, A.T.; Joe, B.; Vijay-Kumar, M. Harnessing innate immunity to eliminate SARS-CoV-2 and ameliorate COVID-19 disease. Physiol. Genom. 2020, 52, 217–221. [Google Scholar] [CrossRef]
  152. Laforge, M.; Elbim, C.; Frère, C.; Hémadi, M.; Massaud, C.; Nuss, P.; Benoliel, J.; Becker, C. Tissue damage from neutrophil-induced oxidative stress in COVID-19. Nat. Rev. Immunol. 2020, 20, 515–516. [Google Scholar] [CrossRef]
  153. Ajaz, S.; McPhail, M.J.; Singh, K.K.; Mujib, S.; Trovato, F.M.; Napoli, S.; Agarwal, K. Mitochondrial metabolic manipulation by SARS-CoV-2 in peripheral blood mononuclear cells of patients with COVID-19. Am. J. Physiol.-Cell Physiol. 2021, 320, C57–C65. [Google Scholar] [CrossRef] [PubMed]
  154. Villaume, W.A. Marginal BH4 deficiencies, iNOS, and self-perpetuating oxidative stress in post-acute sequelae of COVID-19. Med. Hypotheses 2022, 163, 110842. [Google Scholar] [CrossRef] [PubMed]
  155. Ferreira-Vieira, T.H.; Guimaraes, I.M.; Silva, F.R.; Ribeiro, F.M. Alzheimer’s disease: Targeting the Cholinergic System. Curr. Neuropharmacol. 2016, 14, 101–115. [Google Scholar] [CrossRef] [PubMed]
  156. Tracey, K.J. Physiology and immunology of the cholinergic antiinflammatory pathway. J. Clin. Investig. 2007, 117, 289–296. [Google Scholar] [CrossRef]
  157. Fujii, T.; Mashimo, M.; Moriwaki, Y.; Misawa, H.; Ono, S.; Horiguchi, K.; Kawashima, K. Expression and function of the cholinergic system in immune cells. Front. Immunol. 2017, 8, 1085. [Google Scholar] [CrossRef] [PubMed]
  158. Kopańska, M.; Batoryna, M.; Bartman, P.; Szczygielski, J.; Banaś-Ząbczyk, A. Disorders of the Cholinergic System in COVID-19 Era—A Review of the Latest Research. Int. J. Mol. Sci. 2022, 23, 672. [Google Scholar] [CrossRef]
  159. Tizabi, Y.; Getachew, B.; Copeland, R.L.; Aschner, M. Nicotine and the nicotinic cholinergic system in COVID-19. FEBS J. 2020, 287, 3656–3663. [Google Scholar] [CrossRef]
  160. Hampel, H.; Mesulam, M.-M.; Cuello, A.C.; Farlow, M.R.; Giacobini, E.; Grossberg, G.T.; Khachaturian, A.S.; Vergallo, A.; Cavedo, E.; Snyder, P.J.; et al. The cholinergic system in the pathophysiology and treatment of Alzheimer’s disease. Brain 2018, 141, 1917–1933. [Google Scholar] [CrossRef]
  161. Farsalinos, K.; Eliopoulos, E.; Leonidas, D.; Papadopoulos, G.; Tzartos, S.; Poulas, K. Nicotinic cholinergic system and COVID-19: In silico identification of an interaction between SARS-CoV-2 and nicotinic receptors with potential therapeutic targeting implications. Int. J. Mol. Sci. 2020, 21, 5807. [Google Scholar] [CrossRef] [PubMed]
  162. Lagoumintzis, G.; Chasapis, C.T.; Alexandris, N.; Kouretas, D.; Tzartos, S.; Eliopoulos, E.; Farsalinos, K.; Poulas, K. Nicotinic cholinergic system and COVID-19: In silico identification of interactions between α7 nicotinic acetylcholine receptor and the cryptic epitopes of SARS-Co-V and SARS-CoV-2 Spike glycoproteins. Food Chem. Toxicol. 2021, 149, 112009. [Google Scholar] [CrossRef]
  163. Alexandris, N.; Lagoumintzis, G.; Chasapis, C.T.; Leonidas, D.D.; Papadopoulos, G.E.; Tzartos, S.J.; Tsatsakis, A.; Eliopoulos, E.; Poulas, K.; Farsalinos, K. Nicotinic cholinergic system and COVID-19: In silico evaluation of nicotinic acetylcholine receptor agonists as potential therapeutic interventions. Toxicol. Rep. 2020, 8, 73–83. [Google Scholar] [CrossRef] [PubMed]
  164. Leitzke, M. Is the post-COVID-19 syndrome a severe impairment of acetylcholine-orchestrated neuromodulation that responds to nicotine administration? Bioelectron. Med. 2023, 9, 2. [Google Scholar] [CrossRef] [PubMed]
  165. Reitz, C.; Brayne, C.; Mayeux, R. Epidemiology of Alzheimer disease. Nat. Rev. Neurol. 2011, 7, 137–152. [Google Scholar] [CrossRef]
  166. Hauser, P.S.; Ryan, R.O. Impact of Apolipoprotein E on Alzheimer’s Disease. Curr. Alzheimer Res. 2013, 10, 809–817. [Google Scholar] [CrossRef]
  167. Del Ser, T.; Fernandez-Blazquez, M.A.; Valenti, M.; Zea-Sevilla, M.A.; Frades, B.; Alfayate, E.; Saiz, L.; Calero, O.; García-López, F.J.; Rábano, A.; et al. Residence, clinical features, and genetic risk factors associated with symptoms of COVID-19 in a cohort of older people in Madrid. Gerontology 2021, 67, 281–289. [Google Scholar] [CrossRef]
  168. Al-Jaf, S.M.A.; Niranji, S.S.; Ali, H.N.; Mohammed, O.A. Association of apolipoprotein e polymorphism with SARS-CoV-2 infection. Infect. Genet. Evol. 2021, 95, 105043. [Google Scholar] [CrossRef]
  169. Kurki, S.N.; Kantonen, J.; Kaivola, K.; Hokkanen, L.; Mayranpaa, M.I.; Puttonen, H.; Gen, F.; Martola, J.; Pöyhönen, M.; Kero, M.; et al. APOE epsilon4 associates with increased risk of severe COVID-19, cerebral microhaemorrhages and post-COVID mental fatigue: A finnish biobank, autopsy and clinical study. Acta Neuropathol. Commun. 2021, 9, 199. [Google Scholar] [CrossRef]
  170. Hubacek, J.A.; Dlouha, L.; Dusek, L.; Majek, O.; Adamkova, V. Apolipoprotein E4 allele in subjects with COVID-19. Gerontology 2021, 67, 320–322. [Google Scholar] [CrossRef]
  171. Abondio, P.; Sazzini, M.; Garagnani, P.; Boattini, A.; Monti, D.; Franceschi, C.; Luiselli, D.; Giuliani, C. The genetic variability of APOE in different human populations and its implications for longevity. Genes 2019, 10, 222. [Google Scholar] [CrossRef]
  172. Dhangadamajhi, G.; Mishra, S.; Mukherjee, P. Association of ApoE isoforms with COVID-19 outcomes: A world-wide epidemiological study. Hum. Cell 2021, 34, 1932–1933. [Google Scholar] [CrossRef] [PubMed]
  173. Zhang, H.; Shao, L.; Lin, Z.; Long, Q.-X.; Yuan, H.; Cai, L.; Jiang, G.; Guo, X.; Yang, R.; Zhang, Z.; et al. APOE interacts with ACE2 inhibiting SARS-CoV-2 cellular entry and inflammation in COVID-19 patients. Signal Transduct. Target Ther. 2022, 7, 261. [Google Scholar] [CrossRef] [PubMed]
  174. Kuo, C.L.; Pilling, L.C.; Atkins, J.L.; Masoli, J.A.H.; Delgado, J.; Kuchel, G.A.; Melzer, D. APOE e4 Genotype Predicts Severe COVID-19 in the UK Biobank Community Cohort. J. Gerontol. A Biol. Sci. Med. Sci. 2020, 75, 2231–2232. [Google Scholar] [CrossRef] [PubMed]
  175. Montagne, A.; Nation, D.A.; Sagare, A.P.; Barisano, G.; Sweeney, M.D.; Chakhoyan, A.; Pachicano, M.; Joe, E.; Nelson, A.R.; D’Orazio, L.M.; et al. APOE4 leads to blood-brain barrier dysfunction predicting cognitive decline. Nature 2020, 581, 71–76. [Google Scholar] [CrossRef]
  176. Moore, J.B.; June, C.H. Cytokine release syndrome in severe COVID-19. Science 2020, 368, 473–474. [Google Scholar] [CrossRef]
  177. Wang, C.; Zhang, M.; Garcia, G., Jr.; Tian, E.; Cui, Q.; Chen, X.; Sun, G.; Wang, J.; Arumugaswami, V.; Shi, Y. ApoE-Isoform-Dependent SARS-CoV-2 Neurotropism and Cellular Response. Cell Stem Cell 2021, 28, 331–342.e5. [Google Scholar] [CrossRef]
  178. Wang, H.; Yuan, Z.; Pavel, M.A.; Jablonski, S.M.; Jablonski, J.; Hobson, R.; Valente, S.; Reddy, C.B.; Hansen, S.B. The role of high cholesterol in age-related COVID-19 lethality. bioRxiv 2021. [Google Scholar] [CrossRef]
  179. Xiong, N.; Schiller, M.R.; Li, J.; Chen, X.; Lin, Z. Severe COVID-19 in Alzheimer’s disease: APOE4′s fault again? Alzheimers Res. Ther. 2021, 13, 111. [Google Scholar] [CrossRef] [PubMed]
  180. Goyal, A.; Kushwah, P.S.; Agrawal, N.; Pathak, S. APOE4: A Culprit for the Vulnerability of COVID-19 in Alzheimer’s Patients. Curr. Neurovasc. Res. 2023, 20, 162–169. [Google Scholar] [CrossRef]
  181. Ramachandran, A.K.; Das, S.; Shenoy, G.G.; Mudgal, J.; Joseph, A. Relation between apolipoprotein e in alzheimer’s disease and Sars-CoV-2 and their treatment strategy: A review. CNS Neurol. Disord. Drug Targets 2022. [Google Scholar] [CrossRef]
  182. Chen, F.; Chen, Y.; Ke, Q.; Wang, Y.; Gong, Z.; Chen, X.; Cai, Y.; Li, S.; Sun, Y.; Peng, X.; et al. ApoE4 associated with severe COVID-19 outcomes via downregulation of ACE2 and imbalanced RAS pathway. J. Transl. Med. 2023, 21, 103. [Google Scholar] [CrossRef] [PubMed]
  183. Goedert, M.; Spillantini, M.G. A century of Alzheimer’s disease. Science 2006, 314, 777–781. [Google Scholar] [CrossRef] [PubMed]
  184. Abubakar, M.B.; Sanusi, K.O.; Ugusman, A.; Mohamed, W.; Kamal, H.; Ibrahim, N.H.; Khoo, C.S.; Kumar, J. Alzheimer’s Disease: An Update and Insights Into Pathophysiology. Front. Aging Neurosci. 2022, 14, 742408. [Google Scholar] [CrossRef]
  185. Overmyer, K.A.; Shishkova, E.; Miller, I.J.; Balnis, J.; Bernstein, M.N.; Peters-Clarke, T.M.; Meyer, J.G.; Quan, Q.; Muehlbauer, L.K.; Trujillo, E.A.; et al. Large-Scale Multi-omic Analysis of COVID-19 Severity. Cell Syst. 2021, 12, 23–40.e7. [Google Scholar] [CrossRef]
  186. Yang, A.C.; Kern, F.; Losada, P.M.; Agam, M.R.; Maat, C.A.; Schmartz, G.P.; Fehlmann, T.; Stein, J.A.; Schaum, N.; Lee, D.P. Dysregulation of brain and choroid plexus cell types in severe COVID-19. Nature 2021, 595, 565–571. [Google Scholar] [CrossRef] [PubMed]
  187. Ziff, O.J.; Ashton, N.J.; Mehta, P.R.; Brown, R.; Athauda, D.; Heaney, J.; Heslegrave, A.J.; Lessa Benedet, A.; Blennow, K.; Checkley, A.M.; et al. Amyloid processing in COVID-19-associated neurological syndromes. J. Neurochem. 2022, 161, 146–157. [Google Scholar] [CrossRef]
  188. 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]
  189. Hsu, J.T.-A.; Tien, C.F.; Yu, G.-Y.; Shen, S.; Lee, Y.-H.; Hsu, P.-C.; Wang, Y.; Chao, P.-K.; Tsay, H.-J.; Shie, F.-S. The Effects of Aβ1-42 Binding to the SARS-CoV-2 Spike Protein S1 Subunit and Angiotensin-Converting Enzyme 2. Int. J. Mol. Sci. 2021, 22, 8226. [Google Scholar] [CrossRef]
  190. Priemer, D.S.; Rhodes, C.H.; Karlovich, E.; Perl, D.P.; Goldman, J.E. 5β 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]
  191. 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 γ-secretase and enhances amyloid-β production in COVID-19 neuropathy. Cell Discov. 2022, 8, 99. [Google Scholar] [CrossRef] [PubMed]
  192. Kaufer, C.; Schreiber, C.S.; Hartke, A.S.; Denden, I.; Stanelle-Bertram, S.; Beck, S.; Kouassi, N.M.; Beythien, G.; Becker, K.; Schreiner, T.; et al. Microgliosis and neuronal proteinopathy in brain persist beyond viral clearance in SARS-CoV-2 hamster model. EBioMedicine 2022, 79, 103999. [Google Scholar] [CrossRef] [PubMed]
  193. Liao, Q.J.; Ye, L.B.; Timani, K.A.; Zeng, Y.C.; She, Y.L.; Ye, L.; Wu, Z.H. Activation of NF-kappa B by the full-length nucleocapsid protein of the SARS coronavirus. Acta Biochim. Biophys. Sin. 2005, 37, 607–612. [Google Scholar] [CrossRef] [PubMed]
  194. De Diego, M.L.; Nieto-Torres, J.L.; Regla-Nava, J.A.; Jimenez-Guardeno, J.M.; Fernandez-Delgado, R.; Fett, C.; Castano-Rodriguez, C.; Perlman, S.; Enjuanes, L. Inhibition of NF-kappaB-mediated inflammation in severe acute respiratory syndrome coronavirus-infected mice increases survival. J. Virol. 2014, 88, 913–924. [Google Scholar] [CrossRef]
  195. Chen, C.H.; Zhou, W.; Liu, S.; Deng, Y.; Cai, F.; Tone, M.; Tone, Y.; Tong, Y.; Song, W. Increased NF-kappaB signalling up-regulates BACE1 expression and its therapeutic potential in Alzheimer’s disease. Int. J. Neuropsychopharmacol. 2012, 15, 77–90. [Google Scholar] [CrossRef]
  196. Reiken, S.; Sittenfeld, L.; Dridi, H.; Liu, Y.; Liu, X.; Marks, A.R. Alzheimer’s-like signaling in brains of COVID-19 patients. Alzheimers Dement. 2022, 18, 955–965. [Google Scholar] [CrossRef]
  197. Virhammar, J.; Nääs, A.; Fällmar, D.; Cunningham, J.L.; Klang, A.; Ashton, N.J.; Jackmann, S.; Westman, G.; Frithiof, R.; Blennow, K.; et al. Biomarkers for central nervous system injury in cerebrospinal fluid are elevated in COVID-19 and associated with neurological symptoms and diseaseseverity. Eur. J. Neurol. 2021, 28, 3324–3331. [Google Scholar] [CrossRef]
  198. Needham, E.; Ren, A.L.; Digby, R.J.; Norton, E.J.; Ebrahimi, S.; Outtrim, J.G.; Chatfield, D.A.; Manktelow, A.E.; Leibowitz, M.M.; Newcombe, V.F.J.; et al. Brain injury in COVID-19 is associated with dysregulated innate and adaptive immune responses. Brain 2022, 145, 4097–4107. [Google Scholar] [CrossRef]
  199. Ramani, A.; Müller, L.; Ostermann, P.N.; Gabriel, E.; Abida-Islam, P.; Müller-Schiffmann, A.; Mariappan, A.; Goureau, O.; Gruell, H.; Walker, A.; et al. SARS-CoV-2 targets neurons of 3D human brain organoids. EMBO J. 2020, 39, e106230. [Google Scholar] [CrossRef]
  200. Di Primio, C.; Quaranta, P.; Mignanelli, M.; Siano, G.; Bimbati, M.; Scarlatti, A.; Piazza, C.R.; Spezia, P.G.; Perrera, P.; Basolo, F.; et al. Severe acute respiratory syndrome coronavirus 2 infection leads to Tau pathological signature in neurons. PNAS Nexus 2023, 2, pgad282. [Google Scholar] [CrossRef]
  201. Eberle, R.J.; Coronado, M.A.; Gering, I.; Sommerhage, S.; Korostov, K.; Stefanski, A.; Stühler, K.; Kraemer-Schulien, V.; Blömeke, L.; Bannach, O.; et al. Tau protein aggregation associated with SARS-CoV-2 main protease. PLoS ONE 2023, 18, e0288138. [Google Scholar] [CrossRef]
  202. Paidas, M.J.; Cosio, D.S.; Ali, S.; Kenyon, N.S.; Jayakumar, A.R. Long-Term Sequelae of COVID-19 in Experimental Mice. Mol. Neurobiol. 2022, 59, 5970–5986. [Google Scholar] [CrossRef] [PubMed]
  203. Sivagurunathan, N.; Calivarathan, L. SARS-CoV-2 Infection to Premature Neuronal Aging and Neurodegenerative Diseases: Is there any Connection with Hypoxia? CNS Neurol. Disord. Drug Targets 2023, 23, 431–438. [Google Scholar] [CrossRef] [PubMed]
  204. Appelberg, S.; Gupta, S.; Akusjärvi, S.S.; Ambikan, A.T.; Mikaeloff, F.; Saccon, E.; Végvári, A.; Benfeitas, R.; Sperk, M.; Ståhlberg, M.; et al. Dysregulation in Akt/mTOR/HIF-1 signaling identified by proteo-transcriptomics of SARS-CoV-2 infected cells. Emerg. Microbes Infect. 2020, 9, 1748–1760. [Google Scholar] [CrossRef] [PubMed]
  205. Tian, M.; Liu, W.; Li, X.; Zhao, P.; Shereen, M.A.; Zhu, C.; Huang, S.; Liu, S.; Yu, X.; Yue, M.; et al. HIF-1α promotes SARS-CoV-2 infection and aggravates inflammatory responses to COVID-19. Signal Transduct. Target Ther. 2021, 6, 308. [Google Scholar] [CrossRef]
  206. Jana, S.; Heaven, M.R.; Stauft, C.B.; Wang, T.T.; Williams, M.C.; D’Agnillo, F.; Alayash, A.I. 1 HIF-1α-Dependent Metabolic Reprogramming, Oxidative Stress, and Bioenergetic Dysfunction in SARS-CoV-2-Infected Hamsters. Int. J. Mol. Sci. 2022, 24, 558. [Google Scholar] [CrossRef] [PubMed]
  207. Salminen, A.; Kauppinen, A.; Kaarniranta, K. Hypoxia/ischemia activate processing of AmyloidPrecursor Protein: Impact of vascular dysfunction in the pathogenesis of Alzheimer’s disease. J. Neurochem. 2017, 140, 536–549. [Google Scholar] [CrossRef]
  208. Sun, X.; He, G.; Quing, H.; Zhou, W.; Dobie, F.; Cai, F.; Staufenbiel, M.; Huang, L.E.; Song, W. Hypoxia facilitates Alzheimer’s disease pathogenesis by up-regulating BACE1 gene expression. Proc. Natl. Acad. Sci. USA 2006, 103, 18727–18732. [Google Scholar] [CrossRef]
  209. Zhang, X.; Zhou, K.; Wang, R.; Cui, J.; Lipton, S.A.; Liao, F.F.; Xu, H.; Zhang, Y.W. Hypoxia-inducible factor 1alpha (HIF-1alpha)-mediated hypoxia increases BACE1 expression and beta-amyloid generation. J. Biol. Chem. 2007, 282, 10873–10880. [Google Scholar] [CrossRef]
  210. Kerridge, C.; Kozlova, D.I.; Nalivaeva, N.N.; Turner, A.J. Hypoxia Affects Neprilysin Expression through Caspase Activation and an APP Intracellular Domain-dependent Mechanism. Front. Neurosci. 2015, 9, 426. [Google Scholar] [CrossRef]
  211. Wang, Y.-Y.; Huang, Z.-T.; Yuan, M.-H.; Jing, F.; Cai, R.-L.; Zou, Q.; Pu, Y.-S.; Wang, S.Y.; Chen, F.; Yi, W.-M.; et al. Role of Hypoxia Inducible Factor-1α in Alzheimer’s Disease2. J. Alzheimers Dis. 2021, 80, 949–961. [Google Scholar] [CrossRef] [PubMed]
  212. Rhodes, C.H.; Priemer, D.S.; Karlovich, E.; Perl, D.P.; Goldman, J. Β-Amyloid Deposits in Young COVID Patients. Lancet 2023. [Google Scholar] [CrossRef]
  213. Dondaine, T.; Ruthmann, F.; Vuotto, F.; Carton, L.; Gelé, P.; Faure, K.; Deplanque, D.; Bordet, R. Long-term cognitive impairments following COVID-19: A possible impact of hypoxia. J. Neurol. 2022, 269, 3982–3989. [Google Scholar] [CrossRef] [PubMed]
  214. Shapira, R.; Solomon, B.; Efrati, S.; Frenkel, D.; Ashery, U. Hyperbaric oxygen therapy ameliorates pathophysiology of 3xTg-AD mouse model by attenuating neuroinflammation. Neurobiol. Aging 2018, 62, 105–119. [Google Scholar] [CrossRef]
  215. Harch, P.G.; Fogarty, E.F. Hyperbaric oxygen therapy for Alzheimer’s dementia with positron emission tomography imaging: A case report. Med. Gas Res. 2019, 8, 181–184. [Google Scholar] [CrossRef] [PubMed]
  216. Wong, A.C.; Devason, A.S.; Umana, I.C.; Cox, T.O.; Dohnalová, L.; Litichevskiy, L.; Perla, J.; Lundgren, P.; Etwebi, Z.; Izzo, L.T.; et al. Serotonin reduction in post-acute sequelae of viral infection. Cell 2023, 186, 4851–4867.e20. [Google Scholar] [CrossRef] [PubMed]
  217. Sood, A.; Wilson, R.S.; Yu, L.; Wang, T.; Schneider, J.A.; Honer, W.G.; Bennett, D.A. Selective serotonin reuptake inhibitor use, age-related neuropathology and cognition in late-life. Psychiatry Res. 2023, 328, 115471. [Google Scholar] [CrossRef]
  218. Gottfries, C.G.; Bartfai, T.; Carlsson, A.; Eckernas, S.; Svennerholm, L. Multiple biochemical deficits in both gray and white matter of Alzheimer brains Prog. Neuropsychopharmacol. Biol. Psychiatry 1986, 10, 405–413. [Google Scholar] [CrossRef]
  219. Dong, J.; Chen, R.; Zhao, H.; Zhu, Y. COVID-19 and ocular complications: A review of ocular manifestations, diagnostic tools, and prevention strategies. Adv. Ophthalmol. Pract. Res. 2023, 3, 33–38. [Google Scholar] [CrossRef]
  220. Szczęśniak, M.; Brydak-Godowska, J. SARS-CoV-2 and the eyes: A review of the literature on transmission, detection, and ocular manifestations. Med. Sci. Monit. 2021, 27, e931863-1–e931863-10. [Google Scholar] [CrossRef]
  221. de Figueiredo, C.S.; Raony, Í.; Giestal-de-Araujo, E. SARS-CoV-2targeting the retina: Host-virus interaction and possible mechanisms of viral tropism. Ocul. Immunol. Inflamm. 2020, 28, 1301–1304. [Google Scholar] [CrossRef] [PubMed]
  222. D’Alessandro, E.; Kawasaki, A.; Eandi, C.M. Pathogenesis of Vascular Retinal Manifestations in COVID-19 Patients: A Review. Biomedicines 2022, 10, 2710. [Google Scholar] [CrossRef] [PubMed]
  223. Barnett, B.P.; Wahlin, K.; Krawczyk, M.; Spencer, D.; Welsbie, D.; Afshari, N.; Chao, D. Potential of Ocular Transmission of SARS-CoV-2: A Review. Vision 2020, 4, 40. [Google Scholar] [CrossRef]
  224. Coroneo, M.T. The eye as the discrete but defensible portal of coronavirus infection. Ocul. Surf. 2021, 19, 176–182. [Google Scholar] [CrossRef]
  225. deS Senanayake, P.; Drazba, J.; Shadrach, K.; Milsted, A.; Rungger-Brandle, E.; Nishiyama, K.; Miura, S.-I.; Karnik, S.; Sears, J.E.; Hollyfield, J.G. Angiotensin II and its receptor subtypes in the human retina. Investig. Ophthalmol. Vis. Sci. 2007, 48, 3301–3311. [Google Scholar] [CrossRef]
  226. Choudhary, R.; Kapoor, M.S.; Singh, A.; Bodakhe, S.H. Therapeutic targets of renin-angiotensin system in ocular disorders. J. Curr. Ophthalmol. 2016, 29, 7–16. [Google Scholar] [CrossRef]
  227. Qing, H.; Li, Z.; Yang, Z.; Shi, M.; Huang, Z.; Song, J.; Song, Z. The possibility of COVID-19 transmission from eye to nose. Acta Ophthalmol. 2020, 98, e388. [Google Scholar] [CrossRef] [PubMed]
  228. Leonardi, A.; Rosani, U.; Brun, P. Ocular Surface Expression of SARS-CoV-2 Receptors. Ocul. Immunol. Inflamm. 2020, 28, 735–738. [Google Scholar] [CrossRef]
  229. Zhou, L.; Xu, Z.; Castiglione, G.M.; Soiberman, U.S.; Eberhart, C.G.; Duh, E.J. ACE2 and TMPRSS2 are expressed on the human ocular surface, suggesting susceptibility to SARS-CoV-2 infection. Ocul. Surf. 2020, 18, 537–544. [Google Scholar] [CrossRef]
  230. Casagrande, M.; Fitzek, A.; Spitzer, M.S.; Püschel, K.; Glatzel, M.; Krasemann, S.; Nörz, D.; Lütgehetmann, M.; Pfefferle, S.; Schultheiss, M. Presence of SARS-CoV-2 RNA in the Cornea of Viremic Patients with COVID-19. JAMA Ophthalmol. 2021, 139, 383–388. [Google Scholar] [CrossRef]
  231. Casagrande, M.; Fitzek, A.; Püschel, K.; Aleshcheva, G.; Schultheiss, H.-P.; Berneking, L.; Spitze, M.S.; Schultheiss, M. Detection of SARS-CoV-2 in Human Retinal Biopsies of Deceased COVID-19 Patients. Ocul. Immunol. Inflamm. 2020, 28, 721–725. [Google Scholar] [CrossRef] [PubMed]
  232. Li, M.; Yang, Y.; He, T.; Wei, R.; Shen, Y.; Qi, T.; Han, T.; Song, Z.; Zhu, Z.; Ma, X.; et al. Detection of SARS-CoV-2 in the ocular surface in different phases of COVID-19 patients in Shanghai, China. Ann. Transl. Med. 2021, 9, 100. [Google Scholar] [CrossRef]
  233. Aiello, F.; Gallo Afflitto, G.; Mancino, R.; Li, J.O.; Cesareo, M.; Giannini, C.; Nucci, C. Coronavirus disease 2019 (SARS-CoV-2) and colonization of ocular tissues and secretions: A systematic review. Eye 2020, 34, 1206–1211. [Google Scholar] [CrossRef] [PubMed]
  234. Sawant, O.B.; Singh, S.; Wright, R.E., 3rd; Jones, K.M.; Titus, M.S.; Dennis, E.; Hicks, E.; Majmudar, P.A.; Kumar, A.; Mian, S.I. Prevalence of SARS-CoV-2 in human post-mortem ocular tissues. Ocul. Surf. 2021, 19, 322–329. [Google Scholar] [CrossRef]
  235. Yeo, S.; Kim, H.; Lee, J.; Yi, J.; Chung, Y.R. Retinal vascular occlusions in COVID-19 infection and vaccination: A literature review. Graefes Arch. Clin. Exp. Ophthalmol. 2023, 261, 1793–1808. [Google Scholar] [CrossRef]
  236. Abdul-Salam State, S.E.; Sfredel, V.; Mocanu, C.L.; Albu, C.V.; Bălășoiu, A.T. Optic neuropathies post-COVID-19-review. Rom. J. Ophthalmol. 2022, 66, 289–298. [Google Scholar] [CrossRef]
  237. Eissa, M.; Abdelrazek, N.A.; Saady, M. COVID-19 and its relation to the human eye: Transmission, infection, and ocular manifestations. Graefes Arch. Clin. Exp. Ophthalmol. 2022, 261, 1771–1780. [Google Scholar] [CrossRef] [PubMed]
  238. Akbari, M.; Dourandeesh, M. Update on overview of ocular manifestations of COVID-19. Front. Med. 2022, 9, 877023. [Google Scholar] [CrossRef] [PubMed]
  239. Sutandi, N.; Lee, F. Vitreoretinal abnormalities in corona virus disease 2019 patients: What we know so far Taiwan. J. Ophthalmol. 2021, 11, 232–243. [Google Scholar] [CrossRef] [PubMed]
  240. Nasiri, N.; Sharifi, H.; Bazrafshan, A.; Noori, A.; Karamouzian, M.; Sharifi, A. Ocular Manifestations of COVID-19: A Systematic Review and Meta-analysis. J. Ophthalmic Vis. Res. 2021, 16, 103–112. [Google Scholar] [CrossRef] [PubMed]
  241. Sharma, A.; Kudchadkar, U.S.; Shirodkar, R.; Usgaonkar, U.P.S.; Naik, A. Unilateral inferior altitudinal visual field defect related to COVID-19. Indian J. Ophthalmol. 2021, 69, 989–991. [Google Scholar] [CrossRef]
  242. Selvaraj, V.; Sacchetti, D.; Finn, A.; Dapaah-Afriyie, K. Acute vision loss in a patient with COVID-19. R. I. Med. J. 2020, 103, 37–38. [Google Scholar]
  243. Kaya, Y.; Kara, S.; Akinci, C.; Kocaman, A.S. Transient cortical blindness in COVID-19 pneumonia; a PRES-like syndrome: Case report. J. Neurol. Sci. 2020, 413, 116858. [Google Scholar] [CrossRef]
  244. Coco-Martín, M.B.; Leal-Vega, L.; Alcoceba-Herrero, I.; Molina-Martín, A.; de-Fez, D.; Luque, M.J.; Dueñas-Gutiérrez, C.; Arenillas-Lara, J.F.; Piñero, D.P. Visual perception alterations in COVID-19: A preliminary study. Int. J. Ophthalmol. 2023, 16, 1–9. [Google Scholar] [CrossRef]
  245. Zhu, R.; Yu, Z.Y.; Lin Han, L. Insights on the possibility of SARS-CoV-2 transmission through the eyes. Int. J. Ophthalmol. 2022, 15, 1857–1863. [Google Scholar] [CrossRef]
  246. Sen, S.; Kannan, N.B.; Kumar, J.; Rajan, R.P.; Kumar, K.; Baliga, G.; Reddy, H.; Upadhyay, A.; Ramasamy, K. Retinal manifestations in patients with SARS-CoV-2 infection and pathogenetic implications: A systematic review. Int. Ophthalmol. 2022, 42, 323–336. [Google Scholar] [CrossRef] [PubMed]
  247. Romaus-Sanjurjo, D.; Regueiro, U.; López-López, M.; Vázquez-Vázquez, L.; Ouro, A.; Lema, I.; Sobrino, T. Alzheimer’s Disease Seen through the Eye: Ocular Alterations and Neurodegeneration. Int. J. Mol. Sci. 2022, 23, 2486. [Google Scholar] [CrossRef] [PubMed]
  248. Hart, N.J.; Koronyo, Y.; Black, K.L.; Koronyo-Hamaoui, M. Ocular indicators of Alzheimer’s: Exploring disease in the retina. Acta Neuropathol. 2016, 132, 767–787. [Google Scholar] [CrossRef]
  249. den Haan, J.; Morrema, T.H.J.; Verbraak, F.D.; de Boer, J.F.; Scheltens, P.; Rozemuller, A.J.; Bergen, A.A.B.; Bouwman, F.H.; Hoozemans, J.J. Amyloid-beta and phosphorylated tau in post-mortem Alzheimer’s disease retinas. Acta Neuropathol. Commun. 2018, 6, 147. [Google Scholar] [CrossRef]
  250. London, A.; Benhar, I.; Schwartz, M. The retina as a window to the brain: From eye research to CNS disorders. Nat. Rev. Neurol. 2013, 9, 44–53. [Google Scholar] [CrossRef]
  251. Chiquita, S.; Rodrigues-Neves, A.C.; Baptista, F.I.; Carecho, R.; Moreira, P.I.; Castelo-Branco, M.; Ambrósio, A.F. The Retina as a Window or Mirror of the Brain Changes Detected in Alzheimer’s Disease: Critical Aspects to Unravel. Mol. Neurobiol. 2019, 56, 5416–5435. [Google Scholar] [CrossRef]
  252. Latina, V.; Giacovazzo, G.; Cordella, F.; Balzamino, B.O.; Micera, A.; Varano, M.; Marchetti, C.; Malerba, F.; Florio, R.; Ercole, B.B.; et al. Systemic delivery of a specific antibody targeting the pathological N-terminal truncated tau peptide reduces retinal degeneration in a mouse model of Alzheimer’s Disease. Acta Neuropathol. Commun. 2021, 9, 38. [Google Scholar] [CrossRef]
  253. Tippett, W.J.; Black, S.E. Regional Cerebral Blood Flow Correlates of Visuospatial Tasks in Alzheimer’s Disease. J. Int. Neuropsychol. Soc. 2008, 14, 1034–1045. [Google Scholar] [CrossRef]
  254. Rizzo, M.; Anderson, S.W.; Dawson, J.; Nawrot, M. Vision and Cognition in Alzheimer’s Disease. Neuropsychologia 2000, 38, 1157–1169. [Google Scholar] [CrossRef] [PubMed]
  255. Lenoir, H.; Siéroff, É. Visual Perceptual Disorders in Alzheimer’s Disease. Geriatr. Psychol. Neuropsychiatr. Vieil. 2019, 17, 307–316. [Google Scholar] [CrossRef]
  256. Kaeser, P.F.; Ghika, J.; Borruat, F.X. Visual Signs and Symptoms in Patients with the Visual Variant of Alzheimer Disease. BMC Ophthalmol. 2015, 15, 65. [Google Scholar] [CrossRef] [PubMed]
  257. Armstrong, R.; Kergoat, H. Oculo-visual changes and clinical considerations affecting older patients with dementia. Ophthalmic Physiol. Opt. 2015, 35, 352–376. [Google Scholar] [CrossRef]
  258. Javaid, F.Z.; Brenton, J.; Guo, L.; Cordeiro, M.F. Visual and Ocular Manifestations of Alzheimer’s Disease and Their Use as Biomarkers for Diagnosis and Progression. Front. Neurol. 2016, 7, 55. [Google Scholar] [CrossRef]
  259. Kusne, Y.; Wolf, A.B.; Townley, K.; Conway, M.; Peyman, G.A. Visual system manifestations of Alzheimer’s disease. Acta Ophthalmol. 2017, 95, e668–e676. [Google Scholar] [CrossRef]
  260. McKee, A.C.; Au, R.; Cabral, H.J.; Kowall, N.W.; Seshadri, S.; Kubilus, C.A.; Drake, J.; Wolf, P.A. Visual association pathology in preclinical Alzheimer disease. Comparative Study. J. Neuropathol. Exp. Neurol. 2006, 65, 621–630. [Google Scholar] [CrossRef]
  261. Shah, T.M.; Gupta, S.M.; Chatterjee, P.; Campbell, M.; Martins, R.N. Beta-amyloid sequelae in the eye: A critical review on its diagnostic significance and clinical relevance in Alzheimer’s disease. Mol. Psychiatry 2017, 22, 353–363. [Google Scholar] [CrossRef]
  262. Mirzaei, N.; Shi, H.; Oviatt, M.; Doustar, J.; Rentsendorj, A.; Fuchs, D.-T.; Sheyn, J.; Black, K.L.; Koronyo, Y.; Koronyo-Hamaoui, M. Alzheimer’s Retinopathy: Seeing Disease in the Eyes. Front. Neurosci. 2020, 14, 921. [Google Scholar] [CrossRef]
  263. Furman, S.; Green, K.; Lane, T.E. COVID-19 and the impact on Alzheimer’s disease pathology. J. Neurochem. 2023; ahead of print. [Google Scholar] [CrossRef]
  264. Bauer, L.; Laksono, B.M.; de Vrij, F.M.S.; Kushner, S.A.; Harschnitz, O.; van Riel, D. The neuroinvasiveness, neurotropism, and neurovirulence of SARS-CoV-2. Trends Neurosci. 2022, 45, 358–368. [Google Scholar] [CrossRef] [PubMed]
  265. Veleri, S. Neurotropism of SARS-CoV-2 and neurological diseases of the central nervous system in COVID-19 patients. Exp. Brain Res. 2022, 240, 9–25. [Google Scholar] [CrossRef]
  266. Jensen, M.P.; Le Quesne, J.; Officer-Jones, L.; Teodosio, A.; Thaventhiran, J.; Ficken, C.; Goddard, M.; Smith, C.; Menon, D.; Allinson, K.S.J. Neuropathological findings in two patients with fatal COVID-19. Neuropathol. Appl. Neurobiol. 2021, 47, 17–25. [Google Scholar] [CrossRef]
  267. Agrawal, U.; Bedston, S.; McCowan, C.; Oke, J.; Patterson, L.; Robertson, C.; Akbari, A.; Azcoaga-Lorenzo, A.; Bradley, D.T.; Fagbamigbe, A.F.; et al. Severe COVID-19 outcomes after full vaccination of primary schedule and initial boosters: Pooled analysis of national prospective cohort studies of 30 million individuals in England, Northern Ireland, Scotland, and Wales. Lancet 2022, 400, 1305–1320. [Google Scholar] [CrossRef]
  268. Gonzalez-Fernandez, E.; Huang, J. Cognitive Aspects of COVID-19. Curr. Neurol. Neurosci. Rep. 2023, 23, 531–538. [Google Scholar] [CrossRef] [PubMed]
  269. Helms, J.; Kremer, S.; Merdji, H.; Clere-Jehl, R.; Schenck, M.; Kummerlen, C.; Collange, O.; Boulay, C.; Fafi-Kremer, S.; Ohana, M.; et al. Neurologic Features in Severe SARS-CoV-2 Infection. N. Engl. J. Med. 2020, 382, 2268–2270. [Google Scholar] [CrossRef] [PubMed]
  270. Pace, J.L.; Richard, D.; Khachik, A.; Mistry, M.; Singh, G.; Mostaghni, N.; Yazdanmehr, S. Ophthalmic Presentations and Manifestations of COVID-19: A Systematic Review of Global Observations. Cureus 2023, 15, e40695. [Google Scholar] [CrossRef]
  271. Zhao, Y.; Lukiw, W.J. SARS-CoV-2 Neuroinvasion, Inflammatory Neurodegeneration and Alzheimer’s Disease. Front. Cell. Neurosci. 2022, 16, 937961. [Google Scholar] [CrossRef]
  272. Asghari, F.; Asghary, A.; Zolbanin, N.M.; Faraji, F.; Jafari, R. Immunosenescence and Inflammaging in COVID-19. Viral Immunol. 2023; ahead of print. [Google Scholar] [CrossRef]
  273. Mantovani, A.; Morrone, M.C.; Patrono, C.; Santoro, M.G.; Schiaffino, S.; Remuzzi, G.; Bussolati, G. COVID-19 Commission of the Accademia Nazionale dei Lincei Long COVID: Where we stand and challenges ahead. Cell. Death Differ. 2022, 29, 1891–1900. [Google Scholar] [CrossRef] [PubMed]
  274. Huang, P.; Zhang, L.Y.; Tan, Y.Y.; Chen, S.D. Links between COVID-19 and Parkinson’s disease/Alzheimer’s disease: Reciprocal impacts, medical care strategies and underlying mechanisms. Transl. Neurodegener. 2023, 12, 5. [Google Scholar] [CrossRef] [PubMed]
  275. Vandersteen, C.; Plonka, A.; Manera, V.; Sawchuk, K.; Lafontaine, C.; Galery, K.; Rouaud, O.; Bengaied, N.; Launay, C.; Guérin, O.; et al. Alzheimer’s early detection in post-acute COVID-19 syndrome: A systematic review and expert consensus on preclinical assessments. Front. Aging Neurosci. 2023, 15, 1206123. [Google Scholar] [CrossRef]
Figure 1. Long-term neurological and cognitive consequences of COVID-19. An illustration of the peripheral and central neurological manifestations of the post-COVID-19 syndrome.
Figure 1. Long-term neurological and cognitive consequences of COVID-19. An illustration of the peripheral and central neurological manifestations of the post-COVID-19 syndrome.
Cells 12 02601 g001
Figure 2. SARS-CoV-2 virus and its receptors. An illustration showing the SARS-CoV-2 structure and its membrane receptors. This figure was created with BioRender.com.
Figure 2. SARS-CoV-2 virus and its receptors. An illustration showing the SARS-CoV-2 structure and its membrane receptors. This figure was created with BioRender.com.
Cells 12 02601 g002
Figure 3. Neuroinvasive mechanisms of SARS-CoV-2. A picture showing the different routes through which SARS-CoV-2 enters the brain. This figure was created with BioRender.com.
Figure 3. Neuroinvasive mechanisms of SARS-CoV-2. A picture showing the different routes through which SARS-CoV-2 enters the brain. This figure was created with BioRender.com.
Cells 12 02601 g003
Figure 4. Common mutual risks and pathophysiological mechanisms in the COVID-19 pandemic and Alzheimer’s Disease (AD). A schematic of key risk factors associating the COVID-19 and AD pathogenesis. This figure was created with BioRender.com.
Figure 4. Common mutual risks and pathophysiological mechanisms in the COVID-19 pandemic and Alzheimer’s Disease (AD). A schematic of key risk factors associating the COVID-19 and AD pathogenesis. This figure was created with BioRender.com.
Cells 12 02601 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

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. https://doi.org/10.3390/cells12222601

AMA Style

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(22):2601. https://doi.org/10.3390/cells12222601

Chicago/Turabian Style

Amadoro, Giuseppina, Valentina Latina, Egidio Stigliano, and Alessandra Micera. 2023. "COVID-19 and Alzheimer’s Disease Share Common Neurological and Ophthalmological Manifestations: A Bidirectional Risk in the Post-Pandemic Future" Cells 12, no. 22: 2601. https://doi.org/10.3390/cells12222601

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