Next Article in Journal
The Oxidative Stress and Nervous Distress Connection in Gastrointestinal Disorders
Previous Article in Journal
Namodenoson Inhibits the Growth of Pancreatic Carcinoma via Deregulation of the Wnt/β-catenin, NF-κB, and RAS Signaling Pathways
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Neuroprotective Agents with Therapeutic Potential for COVID-19

by
César A. Zaa
1,
Clara Espitia
2,
Karen L. Reyes-Barrera
2,
Zhiqiang An
3 and
Marco A. Velasco-Velázquez
3,4,*
1
School of Biological Sciences, Universidad Nacional Mayor de San Marcos (UNMSM), Lima 15081, Peru
2
Department of Immunology, Institute of Biomedical Research, Universidad Nacional Autónoma de México (UNAM), Mexico City 04510, Mexico
3
Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, TX 77030, USA
4
School of Medicine, Universidad Nacional Autónoma de México (UNAM), Mexico City 04510, Mexico
*
Author to whom correspondence should be addressed.
Biomolecules 2023, 13(11), 1585; https://doi.org/10.3390/biom13111585
Submission received: 12 September 2023 / Revised: 19 October 2023 / Accepted: 20 October 2023 / Published: 27 October 2023
(This article belongs to the Section Natural and Bio-derived Molecules)

Abstract

:
COVID-19 patients can exhibit a wide range of clinical manifestations affecting various organs and systems. Neurological symptoms have been reported in COVID-19 patients, both during the acute phase of the illness and in cases of long-term COVID. Moderate symptoms include ageusia, anosmia, altered mental status, and cognitive impairment, and in more severe cases can manifest as ischemic cerebrovascular disease and encephalitis. In this narrative review, we delve into the reported neurological symptoms associated with COVID-19, as well as the underlying mechanisms contributing to them. These mechanisms include direct damage to neurons, inflammation, oxidative stress, and protein misfolding. We further investigate the potential of small molecules from natural products to offer neuroprotection in models of neurodegenerative diseases. Through our analysis, we discovered that flavonoids, alkaloids, terpenoids, and other natural compounds exhibit neuroprotective effects by modulating signaling pathways known to be impacted by COVID-19. Some of these compounds also directly target SARS-CoV-2 viral replication. Therefore, molecules of natural origin show promise as potential agents to prevent or mitigate nervous system damage in COVID-19 patients. Further research and the evaluation of different stages of the disease are warranted to explore their potential benefits.

Graphical Abstract

1. Introduction

Patients presenting severe pneumonia by an unknown cause were reported in China in December 2019. By January 2020, Chinese authorities reported a novel coronavirus (CoV) as the cause of the illness [1,2]. This novel virus matched the lineage B of the genus betacoronavirus showing >85% identity with a bat SARS-like CoV [1]. Hence, this virus was included in the family containing Severe Acute Respiratory Syndrome (SARS) CoV (79% of genome identity), Middle East respiratory syndrome (MERS) CoV (50% of genome identity), and four other CoVs associated with the common cold [2,3]. Later, the virus was named SARS-CoV-2, and COVID-19 the disease caused by it [2,4]. By the end of January 2020, the World Health Organization (WHO) declared SARS-CoV-2 as a Public Health Emergency of international concern, and by March 2020, COVID-19 was declared a global pandemic [4]. As of 15 March 2023, more than 760 million people worldwide have been infected with SARS-CoV-2, with approximately 6.87 million deaths [5].
Since the beginning of the pandemic, different lineages of SARS-CoV-2 and multiple variants have emerged [6]. The appearance of variants with D614G mutation in the spike (S) protein quickly became dominant strains. This mutation allowed for better binding of the viral S protein to the angiotensin-converting enzyme 2 (ACE2) receptor, accelerating virus infectivity and spread [7]. To date, other variants with important clinical implications have appeared [8]. Such variants are classified according to the potential or known impact of the mutations on the effectiveness of health measures, the severity of the disease, and the ability to spread from person to person [5].
The emerging variants of SARS-CoV-2 are classified into variants of interest (VOI) and variants of concern (VOC). VOI have specific genetic markers associated with changes promoting increased virulence, reduced neutralization by antibodies generated by natural infection or vaccination, and the ability to evade detection or a decrease in the efficacy of vaccination. Eight VOIs have been described: Epsilon (B.1.427 and B.1.429); Zeta (P.2); Eta (B.1.525); Theta (P.3); Iota (B.1.526); Kappa (B.1.617.1); Lambda (C.37), and Mu (B.1.621) [9]. VOC are variants that meet the definition of VOI and that may be associated with one or more of the following: (1) increased transmissibility or detrimental change in the epidemiology of COVID-19; (2) change in the clinical presentation of the disease (more severe manifestations, including death); or (3) decreased effectiveness of public health and social measures [7]. The following VOCs have been described: Alpha (B.1.1.7); Beta (B.1.351); Gamma (P.1); Delta (B.1.617.2); Omicron (B.1.1.529) [9], with variants of the latter currently circulating [10].
Clinical manifestations of COVID-19 range from asymptomatic to mild respiratory tract infections and influenza-like illness to severe illness with lung injury, multiple organ failure, and death. Within such a wide clinical spectrum, multiple varieties of neurological symptoms have been reported. In the present narrative review, we provide a detailed description of the neurological symptomatology in COVID-19 patients and their incidence, compiled from original articles, meta-analyses, and systematic reviews. Furthermore, we discuss the literature generated in the previous four years regarding the mechanisms of SARS-CoV-2 nervous pathogenesis. Finally, we present in vitro, in vivo, and clinical evidence showing that neuroprotective compounds reduce the effects of COVID-19 in the nervous system by targeting the molecular pathways supporting SARS-CoV-2 infection and/or neuronal damage.

2. SARS-CoV-2 Structure and Biology

SARS-CoV-2 is a single-stranded positive-sense RNA virus with an approximate 60–140 nm diameter with spike projections that emerge from the virions’ surface, a characteristic of the Coronaviridae family [1,11].
The initial 20 kb downstream the 5′ end of the viral genomic region is occupied by the open reading frame (ORF) 1a and 1b, which encodes the polyproteins (pp) 1a and 1ab that contain nonstructural proteins (NSPs). Polyprotein pp1a generates NSP1 to NSP11 whereas pp1ab comprises NSP1 to NSP16. Although NSPs are not included in the viral particle, they play an important role in RNA synthesis and processing, contributing to viral propagation [12]. The remaining 10 kb, preceding the 3′ end, code for four structural proteins (10): spike (S), membrane (M), envelope (E), and nucleocapsid (N) [13].
The S protein plays a key role in virus pathogenesis, infectivity, induction of immune response, and evolutionary mutation [14]. The receptor binding domain (RBD) of the S protein binds to ACE2, mediating SARS-CoV-2 entry to host cells [15,16]. Notably, protein S shows a high diversity, caused by selective pressures and adaptive changes over time, which determine a stabilizing interaction in the spike-ACE2 complex [17].
After binding, early viral entry via membrane fusion is promoted by S protein cleavage between S1 and S2 domains by the host protease TMPRSS2 (transmembrane protease serine 2) [18]. ACE2 and TMPRSS2 are expressed in multiple tissues, including lung, kidneys, small intestine, colon, brain, heart, liver, and blood vessels [19,20,21,22], making these tissues susceptible to viral infection. In cells expressing insufficient TMPRSS2, the ACE2-bound virus can be internalized via clathrin-mediated endocytosis in the late endolysosome, where the junction of S1–S2 subunits of the S protein is cleaved by endosomal proteases, especially cathepsin L [18,23]. After the S1 subunit is shed, the S2′ site is cleaved, either by TMPRSS2 on the surface cell or by cathepsins in endosomes, activating membrane fusion [18].
TMPRSS2 inhibitors and cathepsin inhibitors reduce virus infection with different efficacy, indicating that both entry routes are active and cooperate. Thus, the combined treatment with TMPRSS2 inhibitors and cathepsin inhibitors further reduces virus infectivity [15,24]. Accordingly, ACE2 [24], TMPRSS2 [25], and cathepsin L [26] are host-based targets for the development of anti-SARS-CoV-2 therapies.
After RNA release into the host cells, multiple NSPs control RNA transcription, translation, and protein synthesis required for viral replication; thus, such proteins have been targeted for therapeutic development. For example: (i) RNA-dependent RNA polymerase (RdRp, also known as Nsp12) is a crucial component of the genome replication/transcription complex of SARS-CoV-2; (ii) helicase (Nsp13) can unravel double-stranded (ds) DNA and RNA along the 5′–3′ direction and is vital to viral replication; and (iii) Mpro (also known as 3CLPro) and PLPro (papain-like protease) participate in the viral replication through proofreading and excision of the polyproteins [27].
Detailed reviews of SARS-CoV-2 structure and biology have been published recently [28,29].

3. SARS-CoV-2 Infection and Clinical Manifestations

SARS-CoV-2 affects homeostasis, which can lead to life-threatening systemic complications. In the critical SARS-CoV-2 infections, the routes of viral spread in blood vessels are lymphatic, hematogenous, direct invasion of adjacent tissues, and pathogenic implantation. SARS-CoV-2 has been detected in oral and nasal mucosa, stomach, heart, small intestine, colon, lymph nodes, blood samples, thymus, spleen, liver, kidney, and brain [30,31]. SARS-CoV-2, as SARS-CoV, causes pneumonia with severe lung damage in the worst cases, but SARS-CoV-2 is more transmissible due to a higher basic reproductive rate and drives more severe illness [32]. SARS-CoV-2 enters the lung from the mouth and throat [33]. Since the lung has a large surface area, it has increased susceptibility to inhaled viruses. In addition, its high vascularization allows the rapid spread of viral particles.
Direct SARS-CoV-2 infection of lung cells has been described. For example, ACE2 is expressed in the type II alveolar epithelial cells [34], ciliated columnar epithelial [35], and AT2 (alveolar stem cells) [36]. These types of cells express multiple functional genes associated with the viral life cycle including TMPRSS2 [34,37]. SARS-CoV-2 infection of lung cells is accompanied by the infiltration of inflammatory cells, endothelial and inflammatory cell death, alteration of intracellular endothelial junctions, cellular swelling, and cell detachment from the basement membrane [38]. Therefore, pulmonary COVID-19 can be subdivided into four morphological stages, which include: (i) an early stage (day 0–1) with the presence of edema, initial epithelial damage and capillaritis/endothelialitis; (ii) diffuse alveolar damage (DAD) (days 1–7); (iii) the organizing phase (type II pneumocyte hyperplasia) (1 to several weeks); and iv) fibrotic stage of DAD (weeks to months) [39]. Detailed reviews of the pathogeny of severe pneumonia by SARS-CoV-2 infection were published previously [40,41,42,43].
As mentioned before, other organs/tissues can also be infected and damaged by SARS-CoV-2. For example, impaired kidney function has been reported in 13–14.4% of COVID-19 patients [44,45]. SARS-CoV-2 infects the kidney by binding to ACE2 in podocytes. Then, the virus could reach the apical membrane of the proximal tubule either by accessing the tubular fluid or during proximal tubule cell injury in patients with acute kidney injury (AKI) [46]. AKI in COVID-19 patients could be mediated by innate immune system over-activation, cytokine release, complement activation, angiotensin II (Ang II) hyperactivity, development of a hypercoagulable state, hypovolemia secondary to excessive diuresis, and/or increased venous pressure secondary to high positive pressure at the end of expiration [45,47].
A detailed review of the SARS-CoV-2 target organs and the local pathogenesis mechanisms are available in [48].

COVID-19 Neurological Symptoms

A comprehensive exploration of neurological disorders conducted by specialists from the World Federation of Neurology reports that SARS-CoV-2 infection affects the central nervous system (CNS), the peripheral nervous system (PNS), and skeletal muscle [49]. Neurological symptoms in COVID-19 patients (Figure 1), which are more frequent in severely ill patients (45.5%) [50], include:
-
Taste and olfactory dysfunctions (ageusia/anosmia). These are the most common PNS neurological symptoms of COVID-19. They develop in the early stages of the disease and can precede most symptoms; thus, they are considered useful diagnostic markers [51].
-
Headache: Headache is the most common nonspecific neurological symptom, with an estimated combined prevalence of 14.7% [52].
-
Altered mental status/confusion/delirium: Acute confusion/delirium may be a primary manifestation and the only presenting symptom of COVID-19 without evident lung disease [53]. The combined prevalence of altered consciousness/altered mental status is around 9.6% [52].
-
Dizziness: Its combined prevalence in patients with COVID-19 is 8.77%, according to a systematic review [54].
-
Stroke: The prevalence of acute ischemic cerebrovascular disease in hospitalized COVID-19 patients with severe infections reaches approximately 6% [50]. In a retrospective study of 221 patients, 11 (5%) had ischemic stroke, one (0.5%) had cerebral venous thrombosis, and one (0.5%) showed cerebral hemorrhage [55]. Elderly patients with COVID-19 either with vascular risk factors or concomitant diseases such as hypertension or diabetes mellitus are at increased risk of developing cerebrovascular complications [55,56,57]. COVID-19 patients with acute ischemic stroke report visual deficits including hemianopia [58].
-
Epilepsies and seizures: COVID-19 lowers the seizure threshold in patients with existing seizure disorder and may also worsen a controlled condition [59]. SARS-CoV-2-associated seizures can also occur because of meningitis/encephalitis [60]. Several studies have reported that the incidence of acute symptomatic seizures due to COVID-19 is low compared to SARS or MERS. Two large studies each with >4000 COVID-19 patients from Iran or New York reported an incidence of <1% [61]. However, the prevalence might be higher in COVID-19 patients with preexisting or other comorbidities [61,62].
-
Encephalitis: A systematic meta-analysis study found that the incidence of encephalitis as a complication of COVID-19 is <1% for all patients but rises to 6.7% in those with severe disease. In addition, the mortality rate of patients with encephalitis as a complication of COVID-19 is 13.4%, almost four times that of the general population of COVID-19 patients [63].
-
Guillain-Barré syndrome (GBS): COVID-19 patients with GBS can present weakness and paraesthesia of the lower extremities, progressing over several days and that can lead to generalized tetraparesis or tetraplegia [64]. Most of these patients have a demyelinating electrophysiological subtype corresponding to acute inflammatory demyelinating polyneuropathy [65]. In addition, acute motor axonal neuropathy and acute motor and sensory axonal neuropathy have been reported in COVID-19 patients [66,67].
-
Cognitive damage: COVID-19 can cause a cognitive deficit, mainly in attention and executive function, and verbal learning; and the incidence is associated with the severity of COVID-19 [68,69]. The prevalence of cognitive impairment due to COVID-19 infection is not well determined. Reported studies have been limited by sample sizes or suboptimal measures of cognitive functioning [70]. Some of the post-COVID cognitive symptoms may be associated with other systemic symptoms [37]. However, systematic reviews analyzing COVID-19 patients show that cognitive impairment ranges from 2.6% to 81% before or at 12 weeks of infection. After 12 weeks, cognitive decline ranged from 21% to 65% [34,71]. Another meta-analysis study that included 27 studies with 2049 individuals found impairment in executive functions (16%), attention (10%), and memory (24%) in post-COVID-19 patients [70,72]. Consistent results have been reported, even in recovered COVID-19 patients, showing lower overall cognition compared to healthy controls up to 7 months post-infection [72].
-
Impaired cranial nerves: Cranial nerve symptoms are more frequent and severe in COVID-19 than in previous SARS and MERS outbreaks, suggesting that SARS-CoV-2 has a more neurotrophic and aggressive neuroinvasion. Multiple cranial nerve abnormalities in COVID-19 patients have been reported [73]. Although most olfactory sensory neurons (OSNs) do not express ACE2 and TMPRSS2 [74,75], there is evidence that sustentacular cells can serve as a vehicle for the virus, through transcytosis or exosomes, to infect OSNs and reach the brain. Moreover, the virus can impair the OSN renewal process by sustentacular cells in the olfactory epithelium or cause direct damage to CNS neurons. Another possible mechanism is that stem cells in the olfactory epithelium expressing ACE2 are infected with virus from sustentacular cells, and when these cells mature in OSNs they can carry SARS-CoV-2 to the CNS [74,76].
Since the prevalence of hyposmia/anosmia (27.2%) and hypogeusia/ageusia (30.8%) is high in several studies, it is likely that the most frequently involved cranial nerves are I, VII, IX, and X [77]. However, if these common symptoms are produced by virus damage to the CNS or systemic immune response, it can be expected that other cranial nerves are affected [78].
In addition, it is known that the neurons of the terminal nerve (cranial nerve “0”) enter the brain through the cribriform plate and connect the nasal epithelium with the brain centers’ caudal to the olfactory bulb, the medial forebrain (septum), the preoptic area, and the hypothalamus. Although they are rare in humans (in contrast to the greater numbers found in marine mammals), they innervate blood vessels and are in direct contact with the subarachnoid space. These characteristics make it an almost ideal conduit for the transmission of SARS-CoV-2 to the caudal centers of the brain, cerebrospinal fluid, and the vascular system [76].
-
Skeletal muscle symptoms: COVID-19 patients present fatigue, myositis, myalgia, and skeletal muscle injury. Most coronavirus infections can cause functional defects and myalgia or generalized weakness in skeletal muscles with elevated levels of creatine kinase [79]. In SARS-CoV-2-positive individuals, several cases report skeletal muscle symptoms, including back pain, dyskinesia, and lower limb paresthesia [80]. Myalgia prevalence varies widely between studies, from 3.36% to over 64%, with an estimated combined prevalence of around 19.3% [52].
An investigation of 213 COVID-19 cases indicated that 85.2% of patients had significantly elevated serum creatine kinase [81], which can be caused by skeletal muscle injury [79]. Likewise, cases of rhabdomyolysis have been described [82].

4. Mechanisms of SARS-CoV-2-Induced Neurological Damage

Both direct or indirect effects of SARS-CoV-2 on the central nervous system can contribute to neurological and/or neuropsychiatric symptoms of COVID-19 [83]. In the following sections we review the underlying mechanisms.

4.1. Direct Neuronal Damage

SARS-CoV-2 can reach the CNS via the olfactory tract [84]. The human olfactory epithelium is a pseudostratified epithelium composed of Bowman’s glands, horizontal and globose basal cells, microvillar cells, sustentacular cells, and olfactory sensory neurons that extends a single axon towards the olfactory bulb in the brain. Sustentacular cells in the olfactory epithelium exhibit high levels of ACE2 and TMPRSS2 [75] and a study in hamsters showed SARS-CoV-2 active infection in those cells [85]. In agreement, autopsies of COVID-19 patients revealed that the olfactory sensory epithelium was severely damaged [75]. Despite the absence of ACE2 and TMPRSS2 in the olfactory sensory epithelium, the olfactory nerve tissue was found to be positive to SARS-CoV-2 in post-mortem examinations of COVID-19 patients [86]. Thus, it is possible that the replication of virions in the olfactory epithelium leads to infection in the olfactory bulb as blood vessels and pericytes in this brain region express both proteins [87,88]. These neurotrophic properties of SARS-CoV-2 explain the onset of anosmia as a prior symptom [89]. Interestingly, in mice, ACE2 and TMPRSS2 expression in the olfactory epithelium increases with age [75,90], suggesting a possible mechanism by which older patients are more vulnerable to the disease and neurological complications.
Furthermore, the virus can reach brain tissue by the hematogenous route, in which endothelial cells or leukocytes are infected by the virus that passes from the bloodstream to the CNS [75] across the blood–brain barrier (BBB) [91,92] or by transmigration of peripheral immune cells, following the “Trojan horse” mechanism [93,94]. For example, brain vascular cells and choroidal barrier cells robustly express several genes that are relevant for SARS-CoV-2 entry into the brain [95]. SARS-CoV-2 infects and crosses an in vitro model of the BBB comprising primary brain microvascular endothelial cells and astrocytes [96]. Infection of ACE2-overexpressing primary human endothelial cells by SARS-CoV-2 induces the overexpression of coagulation factors, adhesion molecules, and pro-inflammatory cytokines, as well as the formation of multinucleated syncytia and endothelial cell lysis [97]. Consequently, SARS-CoV-2 alters the function and integrity of the BBB, which contributes to viral encephalopathy [98,99].
Furthermore, ACE2 receptors have been found in glial cells of the brain and spinal neurons, so SARS-CoV-2 can adhere, multiply, and cause direct damage to neuronal tissue [100]. Neuronal infection has been associated with neurodegeneration and neurovascular remodeling [101], causing cerebral vascular/endothelial dysfunctions that can generate cerebral circulatory disturbances [102]. Helms et al., using perfusion imaging, demonstrated in patients with COVID-19 that SARS-CoV-2 neuroinvasion causes bilateral frontotemporal hypoperfusion, demonstrating cerebral circulatory impairment [103]. As consequences of cerebral hypoxia, COVID patients can show cerebral vasodilation, brain cell swelling, interstitial edema, obstruction of cerebral blood flow, and even headache due to ischemia and congestion [104].

4.2. Indirect Effects

Exacerbated inflammation participates in the damage to nervous tissue, as in other target organs. SARS-CoV-2 elicits an exacerbated and deregulated immune response of soluble immune mediators, termed a “cytokine storm” [105]. Multiple immune mediators, such as IL-1β, IL-6, CXCL10, TNFα, and other diverse cytokines are produced in response to SARS-CoV-2 infection and have been associated with functional alterations or tissue damage in different organs, including the brain [106].
In addition, elevated levels of pro-inflammatory cytokines could participate in aggravating neuropathies during critical COVID-19 illness. The overproduction of systemic inflammatory factors (cytokines, nitric oxide, and oxygen radicals) has been associated with the malfunction of peripheral nerves [107] as well as microvascular disorders and electrical and metabolic (channel) disturbances in muscle cells [108].
In addition, chronic damage to other systems can also damage the CNS through ischemia, metabolic dysfunction, and hormonal dysregulation [109]. Coagulopathy and endotheliopathy triggered by cytokine storms are potential mechanisms causing ischemic stroke in COVID-19 patients [110,111]. Furthermore, COVID-19 patients have elevated levels of von Willebrand factor (VWF) antigen, VWF activity, and factor VIII [112], leukocytosis, thrombocytopenia, increased partial thromboplastin time, and low levels of antithrombin activity [113]. COVID-19 patients are at an increased risk of developing venous thromboembolism and disseminated intravascular coagulation [114].
Cerebral venous sinus thrombosis (CVT) can be caused by the hypercoagulable state in SARS-CoV-2 infection, which may be triggered by endothelial dysfunction that predisposes vessels to thrombus formation, platelet dysfunction, hypoxia, and/or alterations of the complement system [115,116]. CVT may cause generalized neurological deficits [117] and there are multiple reports of its association with SARS-CoV-2 infection [118,119].
Moreover, the renin-angiotensin-aldosterone system (RAAS) can contribute to the appearance of brain damage and systemic hyperinflammatory state in COVID-19 patients [120,121]. It has been reported that during SARS-CoV-2 infection: (1) the local levels of angiotensin II (Ang II) increase, acting on angiotensin II type 1 receptors (AT1), and thus increasing arterial pressure; (2) there is endothelial dysfunction in the cerebral vessels in the CNS, which increases the risk of cerebral hemorrhage; and (3) the generation of Ang (1–7) decreases, preventing the vasodilator, neuroprotective, and antifibrotic effects of Ang (1–7)/Mas receptor signaling [122,123].

4.3. Oxidative Stress

An overproduction of reactive oxygen species (ROS) and the deprivation of antioxidant mechanisms are known to be crucial for viral replication and subsequent virus-associated disease, as shown by increased ROS levels and impaired antioxidant defense during SARS-CoV-2 infection [124]. The viral protease Mpro activates nuclear factor kappa B (NF-kB)-mediated transcription, which correlates with increased levels of intracellular ROS [125]. In addition, Mpro causes a significant increase in ROS production in HL-CZ cells, which, in turn, induces cellular apoptosis. Similarly, SARS-CoV-2 increases oxidative stress in nervous tissue, which contributes to neuronal cell death [126,127]. A post-mortem case study showed that 37 of 43 COVID-19 patients had astrogliosis and 34 had microglial activation in the brainstem and cerebellum [128]. In a preclinical trial, neuronal microgliosis in the brain has been observed to persist beyond SARS-CoV-2 clearance [129].

4.4. Protein Misfolding

Protein misfolding and aggregation have also been reported in COVID-19. Interactions between the S protein of SARS-CoV-2 and its receptor ACE2 favor the spread of cytosolic prions and tau aggregates [130].
The RBD domain of the S1 subunit from SARS-CoV-2 S protein (RBD SARS-CoV-2 S1) binds heparin and heparin-binding proteins, accelerating the pathological aggregation of brain proteins, including Aβ (amyloid beta), α-synuclein, tau, prion, and TDP-43 RRM [131]. In addition, SARS-CoV-2-infected hamsters develop microgliosis in the olfactory bulb and selective accumulation of hyperphosphorylated tau and α-synuclein in the cortex after virus clearance, indicating that proteinopathies can be generated in neurons post-infection [129]. Although further studies are required, this evidence suggests that protein misfolding may play a role in the neurological symptoms caused by SARS-CoV-2 infection.

4.5. Changes in Neurotrophins Expression

Neurotrophins are growth factors acting as regulators of neuronal survival, development, function, and plasticity [132]. Neurotrophins include nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), neurotrophin-3 (NT-3), and neurotrophin-4 (NT-4) [133]. In addition to their classical functions, they regulate axonal and dendritic growth and guidance, synaptic structure and connections, neurotransmitter release, and long-term potentiation, a cellular mechanism underlying memory and learning [134]. The circulating levels of BDNF [135] and NGF [136] are reduced in adult COVID-19 patients compared to healthy individuals. BDNF reduction is higher in patients > 60 years of age [137], indicating age-dependent effects. Reductions in serum BDNF correlate with the severity of the disease [137,138] and cognitive impairment after recovery [139]. Interestingly, adult COVID-19 patients that required supplemental oxygen had even lower BDNF serum concentrations [135], showing an interplay between deregulated BDNF levels and viral hypoxia. These findings support the role of neurotrophins in regulating neurological outcomes in COVID-19 patients. However, further studies are required to define the extent of their participation and the mechanisms involved, especially in the long-lasting effects of this disease.

5. Neurodegenerative Diseases and COVID-19 Share Mechanisms of Neural Dysfunction

Neurodegenerative diseases are triggered by a combination of genetic, epigenetic, and environmental factors [140]. These diseases share mechanisms of neural damage with COVID-19 described above (Figure 2). Since such mechanisms have been studied for several decades, they may provide a starting point for the identification of specific neuroprotective agents with COVID-19 application.
For example, oxidative stress, the formation of free radicals, and/or a dysfunction of the antioxidant system causes brain damage due to its high oxygen demand and its abundance of lipid cells susceptible to peroxidation [141]. This, in turn, induces altered intracellular signaling that can lead to a dysregulation of the inflammatory response [142] and/or cellular damage by altering the mitochondrial oxidative phosphorylation [143]. Thus, reducing ROS generation, lipid peroxidation, DNA damage, and protein oxidation, and modifying the release of mitochondrial factors are desirable goals in the therapeutic intervention of neurodegenerative diseases [144,145].
Similarly, neuroinflammation is a feature of neurodegenerative diseases. Innate immune cells within the CNS (microglia and astrocytes) as well as infiltrating immune cells are chronically activated in multiple sclerosis (MS) [146]. The accumulation of inflammatory cells and soluble mediators sensitizes neurons to further insults, triggering neurodegeneration by inducing apoptosis, necroptosis, neuronal autophagy, retrograde degeneration, and demyelination [147]. Accordingly, altering the concentration of chemokines and inflammatory cytokines, as well as the activation of astrocytes and microglia have been pointed to as goals of therapies in neurodegenerative diseases [144,145].
The death of neuronal cells is an important mechanism of neurodegenerative pathogenesis and is associated with alterations in the signaling cascades of cell death such as apoptosis, necroptosis, pyroptosis, ferroptosis, and autophagy-associated cell death. Aberrant activation of cell death pathways results in an unwanted loss of neuronal cells and function [148]. These processes can be triggered by intracellular or extracellular stimuli and inflammatory processes [149].
Excitotoxicity of neuronal cells (cell death due to excessive exposure to glutamate or overstimulation of NMDA glutamatergic receptors), is also common in neurodegenerative diseases [150]. Because of that, therapeutic goals include the modification of caspase activation and expression of proapoptotic proteins [144,145], and reduction in excitotoxic [151].
Finally, aberrant protein misfolding, aggregation, and accumulation are hallmarks and pathological features of neurodegenerative diseases such as prion diseases, Alzheimer’s disease (AD), and Parkinson’s disease (PD) (see [152] for a review). Reducing the formation of dysfunctional proteins caused by misfolding and agglomeration is a desirable effect in those neurodegenerative diseases [144,145].
However, only a few drugs have shown efficacy in neurodegenerative diseases and have clinical application. For example, AD is treated with acetylcholinesterase inhibitors (donepezil, galantamine, and rivastigmine), which improves cholinergic neurotransmission [153], and/or the N-methyl-D-aspartate (NMDA) receptor antagonist memantine [154], which prevents excessive continuous activation of extrasynaptic NMDA receptors, reducing excitotoxicity [155]. Thus, new potential pharmacological agents, with these or other relevant activities, are currently being sought [154].

6. Natural Products with Reported Neuroprotective Effects Could Reduce COVID-19 Neurological Symptoms

Evidence suggests that a healthy lifestyle that includes a balanced diet rich in bioactive compounds reduces the risk of developing CNS pathologies [156]. Multiple compounds from natural sources, mainly from medicinal plants, have been identified as modifiers of the pathogenic causes for various neurodegenerative disorders [157,158,159].
In this section we review the use of bioactive natural compounds as neuroprotective agents and discuss the evidence that suggests that they may be useful in COVID-19 treatment. Our literature search identified natural compounds with evidence of neuroprotection from common chemical classes, mainly flavonoids, alkaloids, and terpenoids. Furthermore, some of those compounds with neuroprotective activity may elicit direct effects on the SARS-CoV-2 viral cycle, as reported for multiple compounds from natural sources [160,161,162], making them more attractive candidates for adjuvant therapies.

6.1. Flavonoids

Flavonoids are an important group of polyphenols with a wide range of biological activities. Flavonoids have been shown to be particularly effective in blocking pathological pathways associated with aging and neurodegeneration [163]. Bioactive flavonoids have good bioavailability and stability in circulating plasma and many of them reach the CNS [164,165,166], exerting their protective/restorative capacities in different neuronal populations.
Flavonoids can elicit two different effects that may be beneficial in COVID-19 patients: neuroprotection, mainly because of their antioxidant activity, and modulation of inflammation. For example, the flavone apigenin reduces AD-associated memory impairment, prevents oxidative stress, decreases Aβ plaque load [167], inhibits inflammatory stress, limits apoptotic cell death, and reduces neuronal hyperexcitability [96,168,169]. Furthermore, apigenin attenuates microglial activation and neuroinflammation, counteracting dopaminergic neuronal loss, improving locomotor ability in a PD model [170,171].
Quercetin, a flavonoid ubiquitous in fruits and medicinal herbs, has been widely studied for its neuroprotective effects. It antagonizes neuronal toxicity due to oxidative stress, suppresses neuroinflammation by down-regulating the activation of proinflammatory pathways mediated by NF-kB and iNOS, stimulates neuronal regeneration, and inhibits Aβ aggregation and tau phosphorylation [172]. Epicatechin is a flavonol found in blueberries, tea, cocoa, and grapes. It crosses the BBB [173] and exhibits neuroprotection through anti-apoptosis and anti-mitophagy effects in a model of Parkinson’s disease (PD) [174]. The combination of quercetin and epicatechin synergistically reduces ischemic neuronal cell death, preserves mitochondrial respiratory capacity, and confers protection against hypoxic-ischemic brain damage [175].
Hesperidin and neohesperidin, flavonoids present in citrus, enhance the content of glutathione (GSH) and the antioxidant enzymes catalase (CAT) and superoxide dismutase (SOD) in animal models of ischemic stroke [176], and activates the Akt/Nrf2/HO-1 signaling pathway to inhibit oxidative stress and protect brain damage induced by cerebral artery occlusion [177].
Furthermore, we identified multiple flavonoids that display a direct anti-SARS-CoV-2 effect (Figure 3A and Table 1), which may be further beneficial for COVID-19 patients. The direct antiviral activity of flavonoids is not new and has been extensively reported [178,179,180]. The anti-SARS-CoV-2 activity of flavonoids was proposed at the beginning of the pandemic by computational experiments using SARS-CoV-2 Mpro as a target and confirmed later by crystallography (Figure 3D). Quercetin, baicalein, and luteolin show anti-SARS-CoV-2 activity and share structural features with reported Mpro inhibitors from natural sources (Figure 3E), suggesting they target viral proteins in addition to the neuroprotective actions. Subsequent preclinical evaluations confirmed a direct inhibition of viral entry and/or replication, as well as attenuation of systemic inflammation.
Luteolin is a flavone widely distributed in the plant kingdom. Luteolin protects hippocampal damage and prevents learning defects in a rat model of AD [181]. Interestingly, luteolin attenuates microglial activation and excessive production of TNF-α, nitric oxide, and superoxide [182] and can reduce the activation of the TRIF-dependent signaling pathway of Toll-like receptors [183]. In addition, in SHSY human neuronal cells, it inhibits the transcription of β-secretase 1 (BACE-1), responsible for generating Aβ peptides [184]. It also reversibly inhibits human butyrylcholinesterase [185], which is one of the neuroprotective strategies of approved drugs. Luteolin has been shown to be an allosteric modulator of the S protein of SARS-CoV-2 [186] and inhibitor of Mpro [187].
Kaempferol is a flavonol present in various vegetables such as green tea with reported antiviral, immunomodulatory, and antioxidant effects [188,189]. Kaempferol can overcome the blood–brain barrier (BBB) with a single dose, reaching the hippocampus, frontal cortex, striatum, and cerebellum [190]. It has shown in vivo neuroprotective activity by attenuating the activation of the TLR4/NF-κB pathway in LPS-activated microglial cells [191]. Kaempferol modulates the antiepileptic target synaptic vesicle transporter 2A (SV2A) [192], inhibits 5-HT 3A receptors [193] involved in memory and cognitive functions, and blocks acetylcholinesterase (AChE) [194] implicated in cognitive dysfunction and memory loss associated with AD. Kaempferol decreases epileptic seizures in a rat model of chronic epilepsy, comparable to the control antiepileptic drug [192]. Kaempferol interacts with active sites of RdRp and Mpro from SARS-CoV-2 [195,196].

6.2. Alkaloids

Several alkaloids alter the pathophysiology of AD by functioning as muscarinic receptor agonists, antioxidants, acetylcholinesterase and butyrylcholinesterase inhibitors, α-synuclein aggregation inhibitors, anti-amyloid, and monoamine oxidase (MAO) inhibitors [197]. For example, galantamine is an AChE inhibitor that improves cholinergic neurotransmission, which is impaired in AD [198]. Piperine, a major alkaloid found in long pepper (Piper longum), showed efficacy in attenuating oxidative stress and improving cognition in the rat model of AD [199]. In the 6-hydroxydopamine (6-OHDA)-induced parkinsonian rat model, it decreased the inflammatory markers IL-1β and TNF-α [200]. In epilepsy, piperine delays tonic-clonic seizures by raising the cortical and hippocampal level of serotonin and GABA [201]. Others, such as harmaline, were shown to offset the toxic effects of dopamine oxidation in brain mitochondria, and together with harmine, increased antioxidant enzymes such as SOD and GSH peroxidase [202].
As for flavonoids, we identified alkaloids that, in addition to a neuroprotective effect, may display anti-SARS-CoV-2 activity (Figure 3B and Table 1). Berberine is an isoquinoline present in Tinospora cordifolia and roots, rhizomes, and stem bark of several medicinal plants of the Ranunculaceae, Rutaceae, and Berberidaceae families [203]. Berberine effectively crosses the BBB, which allows it to elicit neurotrophic and neuroprotective effects [204]. These effects are associated not only with its antioxidant action, but with the modulation of enzymes, neurotransmitters and molecular targets involved in neuropathology [205,206,207,208,209,210]. For example, berberine directly reduces ROS [211,212] and activates antioxidant mechanisms by regulating key signaling pathways, such as the P13K/AKT/Bcl-2 pathway and the Nrf2/HO-1 pathway [213]. Berberine is reported as an inhibitor of BACE1 and prevents Aβ 1–42 aggregation to delay the pathological process in AD [214,215,216]. It also improves motor stability and reduces dopaminergic neuron loss in PD [217] and reduces the deposition and aggregation of mutant huntingtin in HD, improving the coordination of movement and motor function [218].
Berberine reduces SARS-CoV-2 infectivity and blocks SARS-CoV-2 replication through direct interaction with the virion in Vero E6 cells and in human nasal epithelial cells [219,220]. In silico studies indicate that it may inhibit the function of SARS-CoV-2 Mpro [221,222]. Through molecular docking and network pharmacology, it was found that berberine inhibits pulmonary fibrosis in COVID-19 pneumonia by reducing TNF-α, IL-6, STAT3, and CCL2 [223]. Likewise, berberine/NIT-X nanoparticles inhibited the replication of SARS-CoV-2, the expression of the ACE2 and TMPRSS2 genes in the human lung epithelial cell line infected with SARS-CoV-2, and the expression of inflammatory cytokines and chemokines [81]. In a clinical study, berberine reduced circulating inflammatory mediators in patients with severe COVID-19 [96].

6.3. Terpenoids

The monoterpenoid carvacrol exhibits neuroprotective activities against cerebral infarction and the associated neurological deficits [224]. Those effects can be mediated by the reduction in inflammation caused by NF-κB inhibition, and inhibition of apoptosis through TRPM7 suppression and promotion of the PI3K/Akt pathway [225,226]. Celastrol (Figure 3C and Table 1), showed its neuroprotection in ischemic stroke by inhibiting the JNK/NF-κB pathway to suppress the inflammatory cascade in the ischemic brain [227]. Rotenone reduces apoptosis in a model of Parkinson’s disease by preventing the increase in ROS and the loss of the mitochondrial membrane potential [228].

6.4. Other Compounds

Gallic acid and caffeic acid are phenolic acids with reported neuroprotective effects [229,230,231]. Gallic acid restores mitochondrial dysfunction [232] and improves the outcome of post-stroke depression treatment [233]. Caffeic acid improves neurological dysfunction and decreases infarct volume after focal cerebral ischemia in rats, with the inhibition of NF-κBp65 expression and reduction in malondialdehyde content through the downregulation of 5-lipoxygenase [234].
Table 1. Classification of anti-SARS-CoV-2 and neuroprotective compounds from natural sources.
Table 1. Classification of anti-SARS-CoV-2 and neuroprotective compounds from natural sources.
TypeCompoundSourceAnti-SARS-CoV-2
Effect
Neuroprotective
Activity
AnalysisReferences
FlavonoidsBaicaleinScutellaria
baicalensis
Antiviral activity in vitro
(EC50: 4.5 μM)
Inhibits Mpro, RdRp, and NSP14 in vitro. Reduces viral load and lung damage in infected mice.
Neuroprotective against AD, PD, cerebral ischemia, epilepsy, aging, and cognitive deficits.In silico
In vitro
In vivo
Clinical trial
[162,235,236,237]
LuteolinCapsicum annuumAntiviral activity in vitro (IC50: 4.6 μM). Binding to ACE2 and in vitro inhibition of RdRp enzyme.Suppresses neuroinflammation, microglia and astrocyte activation, and oxidative stress.In silico
In vitro
[186,238,239]
HesperidinCitrus aurantiumInhibits Mpro, PLpro, and RBD-ACE2 binding (100 μM). Blocks the cellular entry of pseudo-particles of SARS-CoV2.Protects against apoptosis, oxidative stress, and inflammation in AD and PD models. Prevents brain damage.In silico
In vitro
Clinical trial
[240,241,242,243,244,245,246]
QuercetinGinkgo bilobaBinding to Mpro (Km: 11 μM) and RBD.Decreases oxidative stress, neuroinflammation, and neurodegeneration.In silico
In vitro
Clinical trial
[172,247,248,249,250,251,252]
AlkaloidsPiperinePiper longumPossible inhibitor of viral proteases. Combination with curcumin promotes symptomatic recovery in COVID-19 patients. Decreases inflammatory markers IL-1β, TNF-α, and reduces apoptosis. In silico
Clinical trial
[253,254,255,256]
BerberineTinospora
cordifolia
Antiviral activity in vitro (EC50: 9.1 μM). Inhibits Mpro and Nsp15. Reduces the inflammation associated with viral replication in the lungs.Anti-inflammatory, anti-apoptotic, anti-cholinesterase, and anti-amyloid activities.
Protects against subarachnoid hemorrhage by inhibiting the HMGB1/NF-κB pathway.
In silico
In vitro
Clinical trial
[205,213,219,220,257,258]
Tetrandrine Stephaniae tetrandraeAntiviral activity in vitro (IC50: 284 nM). Binding to Two-Pore Channels (TPCs) affecting the viral endosomal entry pathway.Reduces neuroinflammation and apoptosis. Neuroprotection in vascular dementia.In vitro
Clinical trial
[259,260,261,262,263,264,265]
TerpenoidsGlycyrrhizic
acid
Glycyrrhiza
glabra
Antiviral activity in vitro (EC50: 0.44 mg/mL). Binding to NSP-15 and Mpro inhibition during viral replication in vitro. Inhibition of viral replication in one patient.HMGB1 inhibitor. Prevents neuroinflammation, epileptogenesis, and cognitive impairmentIn vitro
Clinical trial
[266,267,268,269,270]
CelastrolTripterygium
wilfordii Hook F
Antiviral activity in vitro (EC50: 2.34 nM). Binding to Mpro and RBD. Inhibits viral replication and decreases IL-6 in vitro.Prevents oxidative stress and inflammation in models of cerebral ischemia, AD, and PD.In vitro[271,272,273,274]
AD: Alzheimer’s disease; PD: Parkinson’s disease; HMGB1: high mobility group box 1 protein.
Resveratrol, a stilbenoid widely used as an antioxidant, has neuroprotective activity in ischemic stroke [275,276]. Such an effect might be related to the activation of AMPK and the NAD + dependent deacetylase SIRT1, which participates in the adaptation to conditions of energy depletion [277].
Curcumin, the major polyphenolic compound extracted from Curcuma longa plants, is an herbal medicine with antitumor, anti-inflammatory, immunomodulatory, antioxidant, antimicrobial, and antiviral activities. Using in vitro models, the antiviral/anti-inflammatory properties of curcumin against SARS-CoV-2 have been evaluated in peripheral blood mononuclear cells (PBMCs), showing an antiviral effect against the DG614 strain and Delta variant. It was also found that pro-inflammatory cytokines (IL-1β, IL-6, and IL-8) released by PBMCs decrease after treatment with curcumin. The results suggest that curcumin affects the replication cycle of SARS-CoV-2 replicative cycle and exhibits virucidal activity with a variant/strain-independent antiviral effect and immunomodulatory properties [278].
Curcumin has several desirable properties as a neuroprotective drug, including anti-inflammatory, antioxidant, and anti-protein aggregation activities, with potential for the prevention of neurological diseases such as AD, PD, Huntington’s, head trauma, aging, and stroke [279]. Curcumin decreases the production of inflammatory mediators such as cytokines, chemokines, and adhesion molecules in the brain of cerebral ischemic patients [280]. Since anti-inflammatory molecules are employed to protect COVID-19 patients from neurological disorders and severe organ-level damage, curcumin treatment could play an important role as a neuroprotector.
Although the bioactive component of turmeric derived from curcuma has a variety of pharmacological activities, its use has been limited by its low solubility, poor bioavailability, rapid metabolism, physicochemical instability, and poor pharmacokinetics [281]. However, the encapsulation of curcumin into nanoformulations has been used to improve its pharmacokinetics, systemic bioavailability, and biological activity. Many nanoformulations have been approved for therapeutic use following the conclusion of preclinical and human clinical trials [282].

7. Conclusions and Perspectives

At present there are multiple therapeutic alternatives for COVID-19 treatment. However, the precise effects of those therapies in the neurological effects of the disease are unclear or have not been studied yet, especially in the long-term onset. The evidence discussed in this review shows that molecules of natural origin with antioxidant, anti-inflammatory and/or cytoprotective activities reduce neuronal damage and improve cognitive function. Thus, those molecules are attractive candidates to be further studied in the management of COVID-19 patients with neurological symptoms. Importantly, the molecules discussed here have key characteristics that support clinical analysis: (i) they are cheap and easily accessible; (ii) they have good safety profiles and biodistribution to the CNS; and (iii) they have shown efficacy in models of other neurodegenerative diseases, in some cases, validating their use in traditional medicine. Still, there are multiple areas of opportunity in the field. For example, clinical trials designed to test the efficacy of neuroprotective compounds in COVID-19 patients are required. In those studies, pharmacokinetic analysis should include evaluation of the bioavailability to show that concentrations effective for neuroprotection are reached after oral administration. However, those trials may be difficult to run given that worldwide vaccination has reduced the number of patients with relevant clinical pictures.
A subset of neuroprotective compounds also inhibits SARS-CoV-2 replication or virus–host interaction. Studies analyzing the structure–activity relationship would allow the design of new molecules with selective or enhanced activities and potential clinical translation. Molecules showing anti-SARS-CoV-2 and neuroprotective activities may improve the prevention and/or mitigation of damage to the CNS induced directly or indirectly by SARS-CoV-2, offering additional benefits to COVID-19 patients.

Author Contributions

Conceptualization, C.A.Z. and M.A.V.-V.; methodology, C.A.Z., C.E., K.L.R.-B. and M.A.V.-V.; investigation C.A.Z. and K.L.R.-B.; data curation C.A.Z., C.E., K.L.R.-B. and M.A.V.-V.; writing—original draft preparation C.A.Z., C.E., K.L.R.-B. and M.A.V.-V.; writing—review and editing, C.A.Z., Z.A. and M.A.V.-V.; supervision M.A.V.-V.; funding acquisition, C.E., Z.A. and M.A.V.-V. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Programa de Apoyo a Proyectos de Investigación e Innovación Tecnológica (PAPIIT) UNAM, grant numbers grant numbers IN206622 (M.A.V.-V.) and IV200121 (C.E. and M.A.V.-V.), by the Research Institutional program “NUATEI” from Instituto de Investigaciones Biomédicas UNAM (C.E.) and by the Welch Foundation AU-0042-20030616 (Z.A).

Acknowledgments

We appreciate the support of Anahi Valdes-Rives in the early compilation of information employed in this review and of Marisol De La Fuente and Karen Escutia-Solis in the preparation of figures. We are grateful to the Dirección General de Asuntos del Personal Académico, UNAM for the postdoctoral fellowship (FOLIO 083723) granted to K.L.R.-B.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

6-hydroxydopamine (6-OHDA); Acute kidney injury: AKI; Alzheimer’s disease: AD; Amyloid beta: Aβ; angiotensin II: Ang II; angiotensin-converting enzyme 2: ACE2; β-secretase 1: BACE-1; blood–brain barrier: BBB; central nervous system: catalase: CAT;CNS; cerebral venous sinus thrombosis: CVT; Coronavirus: CoV; diffuse alveolar damage: DAD; glutathione: GSH; Huntington’s disease: HD; inducible nitric oxide synthase: iNOS; interleukin-1β: IL-1β; Middle East respiratory syndrome: MERS; multiple sclerosis: MS; N-methyl-D-aspartate: NMDA; nonstructural proteins: NSPs; nuclear factor kappa B: NF-kB; olfactory sensory neurons: OSNs; open reading frame: ORF; Parkinson’s disease: PD; peripheral blood mononuclear cells: PBMCs; peripheral nervous system: PNS; polyproteins: pp; reactive oxygen species: ROS; receptor-binding domain: RBD; renin-angiotensin-aldosterone system: RAAS; RNA-dependent RNA polymerase: RdRp; Severe Acute Respiratory Syndrome: SARS; superoxide dismutase: SOD; synaptic vesicle transporter 2A: SV2A; transmembrane protease serine 2: TMPRSS2; variants of concern: VOC; variants of interest: VOI; von Willebrand factor: VWF.

References

  1. Zhu, N.; Zhang, D.; Wang, W.; Li, X.; Yang, B.; Song, J.; Zhao, X.; Huang, B.; Shi, W.; Lu, R.; et al. A novel coronavirus from patients with pneumonia in China, 2019. N. Engl. J. Med. 2020, 382, 727–733. [Google Scholar] [CrossRef]
  2. Lu, H.; Stratton, C.W.; Tang, Y.W. Outbreak of pneumonia of unknown etiology in Wuhan, China: The mystery and the miracle. J. Med. Virol. 2020, 92, 401–402. [Google Scholar] [CrossRef] [PubMed]
  3. Hu, B.; Guo, H.; Zhou, P.; Shi, Z.L. Characteristics of SARS-CoV-2 and COVID-19. Nat. Rev. Microbiol. 2021, 19, 141–154. [Google Scholar] [CrossRef] [PubMed]
  4. Cucinotta, D.; Vanelli, M. WHO declares COVID-19 a pandemic. Acta. Biomed. 2020, 91, 157–160. [Google Scholar] [CrossRef] [PubMed]
  5. WHO. Coronavirus Disease (COVID-19) Pandemic. Available online: https://www.who.int/emergencies/diseases/novel-coronavirus-2019. (accessed on 1 August 2023).
  6. Mistry, P.; Barmania, F.; Mellet, J.; Peta, K.; Strydom, A.; Viljoen, I.M.; James, W.; Gordon, S.; Pepper, M.S. SARS-CoV-2 variants, vaccines, and host immunity. Front. Immunol. 2022, 12, 809244. [Google Scholar] [CrossRef]
  7. Choi, J.Y.; Smith, D.M. SARS-CoV-2 variants of concern. Yonsei Med. J. 2021, 62, 961–968. [Google Scholar] [CrossRef]
  8. Mahilkar, S.; Agrawal, S.; Chaudhary, S.; Parikh, S.; Sonkar, S.C.; Verma, D.K.; Chitalia, V.; Mehta, D.; Koner, B.C.; Vijay, N.; et al. SARS-CoV-2 variants: Impact on biological and clinical outcome. Front. Med. 2022, 9, 1–20. [Google Scholar] [CrossRef]
  9. Aleem, A.; Akbar Samad, A.B.; Vaqar, S. Emerging Variants of SARS-CoV-2 and Novel Therapeutics Against Coronavirus (COVID-19). In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2023. [Google Scholar]
  10. World Health Organization. Coronavirus (COVID-19) Data. Available online: https://www.who.int/data#dashboards (accessed on 1 August 2023).
  11. Cui, J.; Li, F.; Shi, Z.L. Origin and evolution of pathogenic coronaviruses. Nat. Rev. Microbiol. 2019, 17, 181–192. [Google Scholar] [CrossRef]
  12. Biswas, N.; Kumar, K.; Mallick, P.; Das, S.; Kamal, I.M.; Bose, S.; Choudhury, A.; Chakrabarti, S. Structural and drug screening analysis of the non-structural proteins of severe acute respiratory syndrome coronavirus 2 virus extracted from indian coronavirus disease 2019 patients. Front. Genet. 2021, 12, 626642. [Google Scholar] [CrossRef]
  13. Yadav, R.; Chaudhary, J.K.; Jain, N.; Chaudhary, P.K.; Khanra, S.; Dhamija, P.; Sharma, A.; Kumar, A.; Handu, S. Role of structural and non-structural proteins and therapeutic targets of SARS-CoV-2 for COVID-19. Cells 2021, 10, 821. [Google Scholar] [CrossRef]
  14. Almehdi, A.M.; Khoder, G.; Alchakee, A.S.; Alsayyid, A.T.; Sarg, N.H.; Soliman, S.S.M. SARS-CoV-2 spike protein: Pathogenesis, vaccines, and potential therapies. Infection 2021, 49, 855–876. [Google Scholar] [CrossRef] [PubMed]
  15. Shang, J.; Wan, Y.; Luo, C.; Ye, G.; Geng, Q.; Auerbach, A.; Li, F. Cell entry mechanisms of SARS-CoV-2. Proc. Natl. Acad. Sci. USA 2020, 117, 11727–11734. [Google Scholar] [CrossRef] [PubMed]
  16. Ali, A.; Vijayan, R. Dynamics of the ACE2–SARS-CoV-2/SARS-CoV spike protein interface reveal unique mechanisms. Sci. Rep. 2020, 10, 14214. [Google Scholar] [CrossRef] [PubMed]
  17. Ma, W.; Fu, H.; Jian, F.; Cao, Y.; Li, M. Immune evasion and ACE2 binding affinity contribute to SARS-CoV-2 evolution. Nat. Ecol. Evol. 2023, 7, 1457–1466. [Google Scholar] [CrossRef] [PubMed]
  18. Jackson, C.B.; Farzan, M.; Chen, B.; Choe, H. Mechanisms of SARS-CoV-2 entry into cells. Nat. Rev. Mol. Cell Biol. 2022, 23, 3–20. [Google Scholar] [CrossRef]
  19. Devaux, C.A.; Rolain, J.M.; Raoult, D. ACE2 receptor polymorphism: Susceptibility to SARS-CoV-2, hypertension, multi-organ failure, and COVID-19 disease outcome. J. Microbiol. Immunol. Infect. 2020, 53, 425–435. [Google Scholar] [CrossRef]
  20. Berni Canani, R.; Comegna, M.; Paparo, L.; Cernera, G.; Bruno, C.; Strisciuglio, C.; Zollo, I.; Gravina, A.G.; Miele, E.; Cantone, E.; et al. Age-related differences in the expression of most relevant mediators of SARS-CoV-2 infection in human respiratory and gastrointestinal tract. Front. Pediatr. 2021, 9, 697390. [Google Scholar] [CrossRef]
  21. Chen, L.; Li, X.; Chen, M.; Feng, Y.; Xiong, C. The ACE2 expression in human heart indicates new potential mechanism of heart injury among patients infected with SARS-CoV-2. Cardiovasc. Res. 2020, 116, 1097–1100. [Google Scholar] [CrossRef]
  22. Gkogkou, E.; Barnasas, G.; Vougas, K.; Trougakos, I.P. Expression profiling meta-analysis of ACE2 and TMPRSS2, the putative anti-inflammatory receptor and priming protease of SARS-CoV-2 in human cells, and identification of putative modulators. Redox. Biol. 2020, 36, 101615. [Google Scholar] [CrossRef]
  23. Hoffmann, M.; Pöhlmann, S. How SARS-CoV-2 makes the cut. Nat. Microbiol. 2021, 6, 828–829. [Google Scholar] [CrossRef]
  24. Hoffmann, M.; Kleine-Weber, H.; Schroeder, S.; Krüger, N.; Herrler, T.; Erichsen, S.; Schiergens, T.S.; Herrler, G.; Wu, N.H.; Nitsche, A.; et al. SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor. Cell 2020, 181, 271–280.e278. [Google Scholar] [CrossRef] [PubMed]
  25. Thunders, M.; Delahunt, B. Gene of the month: TMPRSS2 (transmembrane serine protease 2). J. Clin. Pathol. 2020, 73, 773–776. [Google Scholar] [CrossRef] [PubMed]
  26. Gomes, C.P.; Fernandes, D.E.; Casimiro, F.; da Mata, G.F.; Passos, M.T.; Varela, P.; Mastroianni-Kirsztajn, G.; Pesquero, J.B. Cathepsin L in COVID-19: From pharmacological evidences to genetics. Front. Cell Infect. Microbiol. 2020, 10, 589505. [Google Scholar] [CrossRef] [PubMed]
  27. Zhou, H.; Fang, Y.; Xu, T.; Ni, W.J.; Shen, A.Z.; Meng, X.M. Potential therapeutic targets and promising drugs for combating SARS-CoV-2. Br. J. Pharmacol. 2020, 177, 3147–3161. [Google Scholar] [CrossRef]
  28. Hardenbrook, N.J.; Zhang, P. A structural view of the SARS-CoV-2 virus and its assembly. Curr. Opin. Virol. 2022, 52, 123–134. [Google Scholar] [CrossRef]
  29. Yan, W.; Zheng, Y.; Zeng, X.; He, B.; Cheng, W. Structural biology of SARS-CoV-2: Open the door for novel therapies. Sig. Transduct. Target. Ther. 2022, 7, 26. [Google Scholar] [CrossRef]
  30. Synowiec, A.; Szczepa, A.; Barreto-Duran, E.; Lie, L.K.; Pyrc, K. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2): A systemic infection. Clin. Microbiol. Rev. 2021, 34, 10. [Google Scholar] [CrossRef]
  31. El Jamal, S.M.; Pujadas, E.; Ramos, I.; Bryce, C.; Grimes, Z.M.; Amanat, F.; Tsankova, N.M.; Mussa, Z.; Olson, S.; Salem, F.; et al. Tissue-based SARS-CoV-2 detection in fatal COVID-19 infections: Sustained direct viral-induced damage is not necessary to drive disease progression. Hum. Pathol. 2021, 114, 110–119. [Google Scholar] [CrossRef]
  32. Petersen, E.; Koopmans, M.; Go, U.; Hamer, D.H.; Petrosillo, N.; Castelli, F.; Storgaard, M.; Al Khalili, S.; Simonsen, L. Comparing SARS-CoV-2 with SARS-CoV and influenza pandemics. Lancet Infect. Dis. 2020, 20, e238–e244. [Google Scholar] [CrossRef]
  33. Li, H.; Wang, Y.; Ji, M.; Pei, F.; Zhao, Q.; Zhou, Y.; Hong, Y.; Han, S.; Wang, J.; Wang, Q.; et al. Transmission routes analysis of SARS-CoV-2: A systematic review and case report. Front. Cell Dev. Biol. 2020, 8, 618. [Google Scholar] [CrossRef]
  34. Zhao, Y.; Zhao, Z.; Wang, Y.; Zhou, Y.; Ma, Y.; Zuo, W. Single-Cell RNA expression profiling of ACE2, the receptor of SARS-CoV-2. Am. J. Respir. Crit. Care. Med. 2021, 202, 756–759. [Google Scholar] [CrossRef] [PubMed]
  35. Lee, I.T.; Nakayama, T.; Wu, C.T.; Goltsev, Y.; Jiang, S.; Gall, P.A.; Liao, C.K.; Shih, L.C.; Schürch, C.M.; McIlwain, D.R.; et al. ACE2 localizes to the respiratory cilia and is not increased by ACE inhibitors or ARBs. Nat. Commun. 2020, 11, 5453. [Google Scholar] [CrossRef]
  36. Silva, M.G.; Falcoff, N.L.; Corradi, G.R.; Di Camillo, N.; Seguel, R.F.; Tabaj, G.C.; Guman, G.R.; de Matteo, E.; Nuñez, M.; Gironacci, M.M. Effect of age on human ACE2 and ACE2-expressing alveolar type II cells levels. Pediatr. Res. 2022, 93, 948–952. [Google Scholar] [CrossRef]
  37. Sungnak, W.; Huang, N.; Bécavin, C.; Berg, M.; Queen, R.; Litvinukova, M.; Talavera-López, C.; Maatz, H.; Reichart, D.; Sampaziotis, F.; et al. SARS-CoV-2 entry factors are highly expressed in nasal epithelial cells together with innate immune genes. Nat. Med. 2020, 26, 681–687. [Google Scholar] [CrossRef] [PubMed]
  38. Stenmark, K.R.; Frid, M.G.; Gerasimovskaya, E.; Zhang, H.; McCarthy, M.K.; Thurman, J.M.; Morrison, T.E. Mechanisms of SARS-CoV-2-induced lung vascular disease: Potential role of complement. Pulm. Circ. 2021, 11, 20458940211015799. [Google Scholar] [CrossRef] [PubMed]
  39. Bösmüller, H.; Matter, M.; Fend, F.; Tzankov, A. The pulmonary pathology of COVID-19. Virchows. Arch. 2021, 478, 137–150. [Google Scholar] [CrossRef] [PubMed]
  40. Harrison, A.G.; Lin, T.; Wang, P. Mechanisms of SARS-CoV-2 transmission and pathogenesis. Trends Immunol. 2020, 41, 1100–1115. [Google Scholar] [CrossRef]
  41. Lamers, M.M.; Haagmans, B.L. SARS-CoV-2 pathogenesis. Nat. Rev. Microbiol. 2022, 20, 270–284. [Google Scholar] [CrossRef]
  42. Attaway, A.H.; Scheraga, R.G.; Bhimraj, A.; Biehl, M.; Hatipoğ Lu, U. Severe COVID-19 pneumonia: Pathogenesis and clinical management. BMJ 2021, 372, n436. [Google Scholar] [CrossRef]
  43. Budinger, G.R.S.; Misharin, A.V.; Ridge, K.M.; Singer, B.D.; Wunderink, R.G. Distinctive features of severe SARS-CoV-2 pneumonia. J. Clin. Investig. 2021, 131, e149412. [Google Scholar] [CrossRef]
  44. Cheng, Y.; Luo, R.; Wang, K.; Zhang, M.; Wang, Z.; Dong, L.; Li, J.; Yao, Y.; Ge, S.; Xu, G. Kidney disease is associated with in-hospital death of patients with COVID-19. Kidney Int. 2020, 97, 829–838. [Google Scholar] [CrossRef] [PubMed]
  45. Hirsch, J.S.; Ng, J.H.; Ross, D.W.; Sharma, P.; Shah, H.H.; Barnett, R.L.; Hazzan, A.D.; Fishbane, S.; Jhaveri, K.D.; Abate, M.; et al. Acute kidney injury in patients hospitalized with COVID-19. Kidney Int. 2020, 98, 209–218. [Google Scholar] [CrossRef] [PubMed]
  46. Hassler, L.; Reyes, F.; Sparks, M.A.; Welling, P.; Batlle, D. Evidence for and against direct kidney infection by SARS-CoV-2 in patients with COVID-19. Clin. J. Am. Soc. Nephrol. 2021, 16, 1755–1765. [Google Scholar] [CrossRef] [PubMed]
  47. Nadim, M.K.; Forni, L.G.; Mehta, R.L.; Connor, M.J.; Liu, K.D.; Ostermann, M.; Rimmelé, T.; Zarbock, A.; Bell, S.; Bihorac, A.; et al. COVID-19-associated acute kidney injury: Consensus report of the 25th Acute Disease Quality Initiative (ADQI) Workgroup. Nat. Rev. Nephrol. 2020, 16, 747–764. [Google Scholar] [CrossRef]
  48. Peiris, S.; Mesa, H.; Aysola, A.; Manivel, J.; Toledo, J.; Borges-Sa, M.; Aldighieri, S.; Reveiz, L. Pathological findings in organs and tissues of patients with COVID-19: A systematic review. PLoS ONE 2021, 16, e0250708. [Google Scholar] [CrossRef]
  49. Román, G.C.; Spencer, P.S.; Reis, J.; Buguet, A.; Faris, M.E.A.; Katrak, S.M.; Láinez, M.; Medina, M.T.; Meshram, C.; Mizusawa, H.; et al. The neurology of COVID-19 revisited: A proposal from the environmental neurology specialty group of the world federation of neurology to implement international neurological registries. J. Neurol. Sci. 2020, 414, 116884. [Google Scholar] [CrossRef]
  50. Mao, L.; Jin, H.; Wang, M.; Hu, Y.; Chen, S.; He, Q.; Chang, J.; Hong, C.; Zhou, Y.; Wang, D.; et al. Neurologic manifestations of hospitalized patients with coronavirus disease 2019 in Wuhan, China. JAMA Neurol. 2020, 77, 683–690. [Google Scholar] [CrossRef]
  51. Cooper, K.W.; Brann, D.H.; Farruggia, M.C.; Bhutani, S.; Pellegrino, R.; Tsukahara, T.; Weinreb, C.; Joseph, P.V.; Larson, E.D.; Parma, V.; et al. COVID-19 and the chemical senses: Supporting players take center stage. Neuron 2020, 107, 219–233. [Google Scholar] [CrossRef]
  52. Favas, T.T.; Dev, P.; Chaurasia, R.N.; Chakravarty, K.; Mishra, R.; Joshi, D.; Mishra, V.N.; Kumar, A.; Singh, V.K.; Pandey, M.; et al. Neurological manifestations of COVID-19: A systematic review and meta-analysis of proportions. Neurol. Sci. 2020, 41, 3437–3470. [Google Scholar] [CrossRef]
  53. Butt, I.; Sawlani, V.; Geberhiwot, T. Prolonged confusional state as first manifestation of COVID-19. Ann. Clin. Transl. Neurol. 2020, 7, 1450–1452. [Google Scholar] [CrossRef]
  54. Pinzon, R.T.; Wijaya, V.O.; Buana, R.B.; Al Jody, A.; Nunsio, P.N. Neurologic characteristics in coronavirus disease 2019 (COVID-19): A systematic review and meta-analysis. Front. Neurol. 2020, 11, 565. [Google Scholar] [CrossRef] [PubMed]
  55. Li, Y.; Li, M.; Wang, M.; Zhou, Y.; Chang, J.; Xian, Y.; Wang, D.; Mao, L.; Jin, H.; Hu, B. Acute cerebrovascular disease following COVID-19: A single center, retrospective, observational study. Stroke Vasc. Neurol. 2020, 5, 279–284. [Google Scholar] [CrossRef] [PubMed]
  56. Divani, A.A.; Andalib, S.; Di Napoli, M.; Lattanzi, S.; Hussain, M.S.; Biller, J.; McCullough, L.D.; Azarpazhooh, M.R.; Seletska, A.; Mayer, S.A.; et al. Coronavirus disease 2019 and stroke: Clinical manifestations and pathophysiological insights. J. Stroke Cerebrovasc. Dis. 2020, 29, 104941. [Google Scholar] [CrossRef]
  57. Carod-Artal, F.J. Neurological complications of coronavirus and COVID-19. Rev. Neurol. 2020, 70, 311–322. [Google Scholar] [CrossRef] [PubMed]
  58. Beyrouti, R.; Adams, M.E.; Benjamin, L.; Cohen, H.; Farmer, S.F.; Goh, Y.Y.; Humphries, F.; Jager, H.R.; Losseff, N.A.; Perry, R.J.; et al. Characteristics of ischaemic stroke associated with COVID-19. J. Neurol. Neurosurg. Psychiatry 2020, 91, 889–891. [Google Scholar] [CrossRef] [PubMed]
  59. Somani, S.; Pati, S.; Gaston, T.; Chitlangia, A.; Agnihotri, S. De novo status epilepticus in patients with COVID-19. Ann. Clin. Transl. Neurol. 2020, 7, 1240–1244. [Google Scholar] [CrossRef]
  60. Moriguchi, T.; Harii, N.; Goto, J.; Harada, D.; Sugawara, H.; Takamino, J.; Ueno, M.; Sakata, H.; Kondo, K.; Myose, N.; et al. A first case of meningitis/encephalitis associated with SARS-Coronavirus-2. Int. J. Infect. Dis. 2020, 94, 55–58. [Google Scholar] [CrossRef]
  61. Pellinen, J.; Holmes, M.G. Evaluation and treatment of seizures and epilepsy during the COVID-19 pandemic. Curr. Neurol. Neurosci. Rep. 2022, 22, 11–17. [Google Scholar] [CrossRef]
  62. Lundstrom, K.; Hromić-Jahjefendić, A.; Bilajac, E.; Aljabali, A.; Baralić, K.; Sabri, N.; Shehata, E.; Raslan, M.; Ferreira, A.; Orlandi, L.; et al. COVID-19 signalome: Pathways for SARS-CoV-2 infection and impact on COVID-19 associated comorbidity. Cell Signal. 2023, 1, 110495. [Google Scholar] [CrossRef]
  63. Siow, I.; Lee, K.S.; Zhang, J.J.Y.; Saffari, S.E.; Ng, A. Encephalitis as a neurological complication of COVID-19: A systematic review and meta-analysis of incidence, outcomes, and predictors. Eur. J. Neurol. 2021, 28, 3491–3502. [Google Scholar] [CrossRef]
  64. Rahimi, K. Guillain-Barre syndrome during COVID-19 pandemic: An overview of the reports. Neurol. Sci. 2020, 41, 3149–3156. [Google Scholar] [CrossRef]
  65. Caress, J.B.; Castoro, R.J.; Simmons, Z.; Scelsa, S.N.; Lewis, R.A.; Ahlawat, A.; Narayanaswami, P. COVID-19–associated Guillain-Barré syndrome: The early pandemic experience. Muscle Nerve 2020, 62, 485–491. [Google Scholar] [CrossRef] [PubMed]
  66. Alberti, P.; Beretta, S.; Piatti, M.; Karantzoulis, A.; Piatti, M.L.; Santoro, P.; Viganò, M.; Giovannelli, G.; Pirro, F.; Montisano, D.A.; et al. Guillain-Barré syndrome related to COVID-19 infection. Neurol. Neuroimmunol. Neuroinflam. 2020, 7, e741. [Google Scholar] [CrossRef]
  67. Virani, A.; Rabold, E.; Hanson, T.; Haag, A.; Elrufay, R.; Cheema, T.; Balaan, M.; Bhanot, N. Guillain-Barré Syndrome associated with SARS-CoV-2 infection. IDCases 2020, 20, e00771. [Google Scholar] [CrossRef] [PubMed]
  68. Burdick, K.E.; Millett, C.E. The impact of COVID-19 on cognition in severe cases highlights the need for comprehensive neuropsychological evaluations in all survivors. Neuropsychopharmacology 2021, 46, 2225. [Google Scholar] [CrossRef] [PubMed]
  69. Miskowiak, K.W.; Johnsen, S.; Sattler, S.M.; Nielsen, S.; Kunalan, K.; Rungby, J.; Lapperre, T.; Porsberg, C.M. Cognitive impairments four months after COVID-19 hospital discharge: Pattern, severity and association with illness variables. Eur. Neuropsychopharmacol. 2021, 46, 39–48. [Google Scholar] [CrossRef] [PubMed]
  70. Becker, J.H.; Lin, J.J.; Doernberg, M.; Stone, K.; Navis, A.; Festa, J.R.; Wisnivesky, J.P. Assessment of cognitive function in patients after COVID-19 infection. JAMA Netw. Open 2021, 4, e2130645. [Google Scholar] [CrossRef] [PubMed]
  71. Tavares-Junior, J.W.L.; de Souza, A.C.C.; Borges, J.W.P.; Oliveira, D.N.; Siqueira-Neto, J.I.; Sobreira-Neto, M.A.; Braga-Neto, P. COVID-19 associated cognitive impairment: A systematic review. Cortex 2022, 152, 77–97. [Google Scholar] [CrossRef]
  72. 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]
  73. Guerrero, J.I.; Barragán, L.A.; Martínez, J.D.; Montoya, J.P.; Peña, A.; Sobrino, F.E.; Tovar-Spinoza, Z.; Ghotme, K.A. Central and peripheral nervous system involvement by COVID-19: A systematic review of the pathophysiology, clinical manifestations, neuropathology, neuroimaging, electrophysiology, and cerebrospinal fluid findings. BMC Infect. Dis. 2021, 21, 1–15. [Google Scholar] [CrossRef]
  74. Butowt, R.; von Bartheld, C.S. The route of SARS-CoV-2 to brain infection: Have we been barking up the wrong tree? Mol. Neurodegener. 2022, 17, 1–4. [Google Scholar] [CrossRef] [PubMed]
  75. Bilinska, K.; Jakubowska, P.; Von Bartheld, C.S.; Butowt, R. Expression of the SARS-CoV-2 entry proteins, ACE2 and TMPRSS2, in cells of the olfactory epithelium: Identification of cell types and trends with age. ACS Chem. Neurosci. 2020, 11, 1555–1562. [Google Scholar] [CrossRef] [PubMed]
  76. Butowt, R.; von Bartheld, C.S. Anosmia in COVID-19: Underlying Mechanisms and Assessment of an Olfactory Route to Brain Infection. Neuroscientist 2021, 27, 582–603. [Google Scholar] [CrossRef] [PubMed]
  77. Finsterer, J.; Scorza, F.A.; Scorza, C.A.; Fiorini, A.C. COVID-19 associated cranial nerve neuropathy: A systematic review. Bosn. J. Basic. Med. Sci. 2022, 22, 39–45. [Google Scholar] [CrossRef]
  78. Doblan, A.; Kaplama, M.E.; Ak, S.; Basmacı, N.; Tarini, E.Z.; Göktaş, Ş.E.; Güler, S.; Müderris, T. Cranial nerve involvement in COVID-19. Am. J. Otolaryngol. 2021, 42, 102999. [Google Scholar] [CrossRef]
  79. Colosio, M.; Brocca, L.; Gatti, M.; Neri, M.; Crea, E.; Cadile, F.; Canepari, M.; Pel-legrino, M.; Polla, B.; Porcelli, S.; et al. Structural and functional impairments of skeletal muscle in patients with post-acute sequelae of SARS-CoV-2 infection. J. Appl. Physiol. 2023, 135, 902–917. [Google Scholar] [CrossRef]
  80. Mehan, W.A.; Yoon, B.C.; Lang, M.; Li, M.D.; Rincon, S.; Buch, K. Paraspinal myositis in patients with COVID-19 infection. AJNR Am. J. Neuroradiol. 2020, 41, 1949–1952. [Google Scholar] [CrossRef]
  81. Wang, Z.Z.; Li, K.; Maskey, A.R.; Huang, W.; Toutov, A.A.; Yang, N.; Srivastava, K.; Geliebter, J.; Tiwari, R.; Miao, M.; et al. A small molecule compound berberine as an orally active therapeutic candidate against COVID-19 and SARS: A computational and mechanistic study. FASEB J. 2021, 35, e21360. [Google Scholar] [CrossRef]
  82. Jin, M.; Tong, Q. Rhabdomyolysis as potential late complication associated with COVID-19. Emerg. Infect. Dis. 2020, 26, 1618–1620. [Google Scholar] [CrossRef]
  83. Fotuhi, M.; Mian, A.; Meysami, S.; Raji, C.A. Neurobiology of COVID-19. J. Alzheimer’s Dis. 2020, 76, 3–19. [Google Scholar] [CrossRef]
  84. Li, Y.C.; Bai, W.Z.; Hashikawa, T. The neuroinvasive potential of SARS-CoV2 may play a role in the respiratory failure of COVID-19 patients. J. Med. Virol. 2020, 92, 552–555. [Google Scholar] [CrossRef] [PubMed]
  85. Bryche, B.; St Albin, A.; Murri, S.; Lacôte, S.; Pulido, C.; Ar Gouilh, M.; Lesellier, S.; Servat, A.; Wasniewski, M.; Picard-Meyer, E.; et al. Massive transient damage of the olfactory epithelium associated with infection of sustentacular cells by SARS-CoV-2 in golden Syrian hamsters. Brain Behav. Immun. 2020, 89, 579–586. [Google Scholar] [CrossRef] [PubMed]
  86. Fabbri, V.P.; Foschini, M.P.; Lazzarotto, T.; Gabrielli, L.; Cenacchi, G.; Gallo, C.; Aspide, R.; Frascaroli, G.; Cortelli, P.; Riefolo, M.; et al. Brain ischemic injury in COVID-19-infected patients: A series of 10 post-mortem cases. Brain Pathol. 2021, 31, 205–210. [Google Scholar] [CrossRef] [PubMed]
  87. Brann, D.H.; Tsukahara, T.; Weinreb, C.; Lipovsek, M.; Van den Berge, K.; Gong, B.; Chance, R.; Macaulay, I.C.; Chou, H.J.; Fletcher, R.B.; et al. Non-neuronal expression of SARS-CoV-2 entry genes in the olfactory system suggests mechanisms underlying COVID-19-associated anosmia. Sci. Adv. 2020, 6, eabc5801. [Google Scholar] [CrossRef]
  88. Lechien, J.R.; Radulesco, T.; Calvo-Henriquez, C.; Chiesa-Estomba, C.M.; Hans, S.; Barillari, M.R.; Cammaroto, G.; Descamps, G.; Hsieh, J.; Vaira, L.; et al. ACE2 & TMPRSS2 Expressions in Head & Neck Tissues: A Systematic Review. Head Neck Pathol. 2021, 15, 225–235. [Google Scholar] [CrossRef]
  89. Al-olama, M.; Rashid, A.; Garozzo, D. COVID-19-associated meningoencephalitis complicated with intracranial hemorrhage: A case report. Acta. Neurochir. 2020, 162, 1495–1499. [Google Scholar] [CrossRef]
  90. Brechbühl, J.; Lopes, A.C.; Wood, D.; Bouteiller, S.; de Vallière, A.; Verdumo, C.; Broillet, M.C. Age-dependent appearance of SARS-CoV-2 entry sites in mouse chemosensory systems reflects COVID-19 anosmia-ageusia symptoms. Commun. Biol. 2021, 4, 880. [Google Scholar] [CrossRef]
  91. Proust, A.; Queval, C.J.; Harvey, R.; Adams, L.; Bennett, M.; Wilkinson, R. Differential effects of SARS-CoV-2 variants on central nerv-ous system cells and blood–brain barrier functions. J. Neuroinflam. 2023, 20, 1–17. [Google Scholar] [CrossRef]
  92. Reynolds, J.L.; Mahajan, S.D. SARS-CoV2 alters blood brain barrier integrity contributing to neuro-inflammation. J. Neuroimmune Pharmacol. 2021, 16, 4–6. [Google Scholar] [CrossRef]
  93. Dahm, T.; Rudolph, H.; Schwerk, C.; Schroten, H.; Tenenbaum, T. Neuroinvasion and inflammation in viral central nervous system infections. Mediat. Inflamm. 2016, 2016, 8562805. [Google Scholar] [CrossRef]
  94. Gu, J.; Gong, E.; Zhang, B.; Zheng, J.; Gao, Z.; Zhong, Y.; Zou, W.; Zhan, J.; Wang, S.; Xie, Z.; et al. Multiple organ infection and the pathogenesis of SARS. J. Exp. Med. 2005, 202, 415–424. [Google Scholar] [CrossRef] [PubMed]
  95. 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.; et al. Dysregulation of brain and choroid plexus cell types in severe COVID-19. Nature 2021, 595, 565–571. [Google Scholar] [CrossRef] [PubMed]
  96. Zhang, B.Y.; Chen, M.; Chen, X.C.; Cao, K.; You, Y.; Qian, Y.J.; Yu, W.K. Berberine reduces circulating inflammatory mediators in patients with severe COVID-19. Br. J. Surg. 2021, 108, E9–E11. [Google Scholar] [CrossRef] [PubMed]
  97. Conde, J.N.; Schutt, W.R.; Gorbunova, E.E.; Mackow, E.R. Recombinant ACE2 expression is required for SARS-CoV-2 to infect primary human endothelial cells and induce inflammatory and procoagulative responses. mBio 2020, 11, e03185-20. [Google Scholar] [CrossRef]
  98. Erickson, M.A.; Rhea, E.M.; Knopp, R.C.; Banks, W.A. Interactions of SARS-CoV-2 with the blood–brain barrier. Int. J. Mol. Sci. 2021, 22, 2681. [Google Scholar] [CrossRef]
  99. Buzhdygan, T.P.; DeOre, B.J.; Baldwin-Leclair, A.; Bullock, T.A.; McGary, H.M.; Khan, J.A.; Razmpour, R.; Hale, J.F.; Galie, P.A.; Potula, R.; et al. The SARS-CoV-2 spike protein alters barrier function in 2D static and 3D microfluidic in-vitro models of the human blood–brain barrier. Neurobiol. Dis. 2020, 146, 105131. [Google Scholar] [CrossRef]
  100. Ahmad, I.; Rathore, F.A. Neurological manifestations and complications of COVID-19: A literature review. J. Clin. Neurosci. 2020, 77, 8–12. [Google Scholar] [CrossRef]
  101. Song, E.; Zhang, C.; Israelow, B.; Lu-Culligan, A.; Prado, A.V.; 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. [Google Scholar] [CrossRef]
  102. Ghasemi, M.; Umeton, R.P.; Keyhanian, K.; Mohit, B.; Rahimian, N.; Eshaghhosseiny, N.; Davoudi, V. SARS-CoV-2 and acute cerebrovascular events: An overview. J. Clin. Med. 2021, 10, e20202135. [Google Scholar] [CrossRef]
  103. 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]
  104. Guo, Y.R.; Cao, Q.D.; Hong, Z.S.; Tan, Y.Y.; Chen, S.D.; Jin, H.J.; Tan, K.S.; Wang, D.Y.; Yan, Y. The origin, transmission and clinical therapies on coronavirus disease 2019 (COVID-19) outbreak—An update on the status. Mil. Med. Res. 2020, 7, 1–10. [Google Scholar] [CrossRef] [PubMed]
  105. Huang, C.; Wang, Y.; Li, X.; Ren, L.; Zhao, J.; Hu, Y.; Zhang, L.; Fan, G.; Xu, J.; Gu, X.; et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet 2020, 395, 497–506. [Google Scholar] [CrossRef] [PubMed]
  106. Conti, P.; Ronconi, G.; Caraffa, A.; Gallenga, C.E.; Ross, R.; Frydas, I.; Kritas, S.K. Induction of pro-inflammatory cytokines (IL-1 and IL-6) and lung inflammation by Coronavirus-19 (COVI-19 or SARS-CoV-2): Anti-inflammatory strategies. J. Biol. Regul. Homeost. Agents 2020, 34, 327–331. [Google Scholar]
  107. Hasan, L.K.; Deadwiler, B.; Haratian, A.; Bolia, I.K.; Weber, A.E.; Petrigliano, F.A. Effects of COVID-19 on the musculoskeletal system: Clinician’s guide. Orthop. Res. Rev. 2021, 13, 141–150. [Google Scholar] [CrossRef] [PubMed]
  108. Disser, N.P.; De Micheli, A.J.; Schonk, M.M.; Konnaris, M.A.; Piacentini, A.N.; Edon, D.L.; Toresdahl, B.G.; Rodeo, S.A.; Casey, E.K.; Mendias, C.L. Musculoskeletal Consequences of COVID-19. J. Bone Jt. Surg. Am. 2020, 102, 1197–1204. [Google Scholar] [CrossRef] [PubMed]
  109. Iadecola, C.; Anrather, J.; Kamel, H. Effects of COVID-19 on the nervous system. Cell 2020, 183, 16–27.e11. [Google Scholar] [CrossRef]
  110. Vogrig, A.; Gigli, G.L.; Bnà, C.; Morassi, M. Stroke in patients with COVID-19: Clinical and neuroimaging characteristics. Neurosci. Lett. 2021, 743, 135564. [Google Scholar] [CrossRef]
  111. Altable, M.; de la Serna, J.M. Cerebrovascular disease in COVID-19: Is there a higher risk of stroke? Brain Behav. Immun. Health 2020, 6, 100092. [Google Scholar] [CrossRef]
  112. Leonard-Lorant, I.; Delabranche, X.; Severac, F.; Helms, J.; Pauzet, C.; Collange, O.; Schneider, F.; Labani, A.; Bilbault, P.; Moliere, S.; et al. Acute pulmonary embolism in patients with COVID-19 at CT angiography and relationship to d-dimer levels. Radiology 2020, 296, E189–E191. [Google Scholar] [CrossRef]
  113. Zhang, Y.; Xiao, M.; Zhang, S.; Xia, P.; Cao, W.; Jiang, W.; Chen, H.; Ding, X.; Zhao, H.; Zhang, H.; et al. Coagulopathy and Antiphospholipid Antibodies in Patients with COVID-19. N. Engl. J. Med. 2020, 382, e38. [Google Scholar] [CrossRef]
  114. Pavoni, V.; Gianesello, L.; Pazzi, M.; Stera, C.; Meconi, T.; Frigieri, F.C. Evaluation of coagulation function by rotation thromboelastometry in critically ill patients with severe COVID-19 pneumonia. J. Thromb. Thrombolysis 2020, 50, 281–286. [Google Scholar] [CrossRef] [PubMed]
  115. Cavalcanti, D.D.; Raz, E.; Shapiro, M.; Dehkharghani, S.; Yaghi, S.; Lillemoe, K.; Nossek, E.; Torres, J.; Jain, R.; Riina, H.A.; et al. Cerebral venous thrombosis associated with COVID-19. AJNR Am. J. Neuroradiol. 2020, 41, 1370–1376. [Google Scholar] [CrossRef] [PubMed]
  116. Ghosh, R.; Roy, D.; Mandal, A.; Pal, S.K.; Chandra Swaika, B.; Naga, D.; Pandit, A.; Ray, B.K.; Benito-León, J. Cerebral venous thrombosis in COVID-19. Diabetes Metab. Syndr. 2021, 15, 1039–1045. [Google Scholar] [CrossRef] [PubMed]
  117. Capecchi, M.; Abbattista, M.; Martinelli, I. Cerebral venous sinus thrombosis. J. Thromb. Haemost. 2018, 16, 1918–1931. [Google Scholar] [CrossRef]
  118. Christopher, H.; Tom, N.; Martin, P.; Christian, S.; Salah, E. Cerebral venous sinus thrombosis as a presentation of COVID-19. Eur. J. Case. Rep. Intern. Med. 2020, 7, 001691. [Google Scholar] [CrossRef]
  119. Essajee, F.; Solomons, R.; Goussard, P.; Van Toorn, R. Child with tuberculous meningitis and COVID-19 coinfection complicated by extensive cerebral sinus venous thrombosis. BMJ Case Rep. 2020, 13, e238597. [Google Scholar] [CrossRef]
  120. Zanza, C.; Tassi, M.F.; Romenskaya, T.; Piccolella, F.; Abenavoli, L.; Franceschi, F.; Piccioni, A.; Ojetti, V.; Saviano, A.; Canonico, B.; et al. Lock, stock and barrel: Role of renin-angiotensin-aldosterone system in coronavirus disease 2019. Cells 2021, 10, 1752. [Google Scholar] [CrossRef]
  121. Panariello, F.; Cellini, L.; Speciani, M.; De Ronchi, D.; Atti, A.R. How does SARS-CoV-2 affect the central nervous system? A working hypothesis. Front. Psychiatry 2020, 11, 582345. [Google Scholar] [CrossRef]
  122. Christian, W.; Julian, K.-B. Epidemiology, prognosis and prevention of non-traumatic intracerebral hemorrhage. Curr. Pharm. Des. 2017, 15, 2193–2219. [Google Scholar]
  123. Benger, M.; Williams, O.; Siddiqui, J.; Sztriha, L. Intracerebral haemorrhage and COVID-19: Clinical characteristics from a case series. Brain Behav. Immun. 2020, 88, 940–944. [Google Scholar] [CrossRef]
  124. Delgado-Roche, L.; Mesta, F. Oxidative stress as key player in severe acute respiratory syndrome coronavirus (SARS-CoV) infection. Arch. Med. Res. 2020, 51, 384–387. [Google Scholar] [CrossRef] [PubMed]
  125. Lin, C.W.; Lin, K.H.; Hsieh, T.H.; Shiu, S.Y.; Li, J.Y. Severe acute respiratory syndrome coronavirus 3C-like protease-induced apoptosis. FEMS Microbiol. Immunol. 2006, 46, 375–380. [Google Scholar] [CrossRef]
  126. Forcados, G.E.; Muhammad, A.; Oladipo, O.O.; Makama, S.; Meseko, C.A. Metabolic implications of oxidative stress and inflammatory process in SARS-CoV-2 pathogenesis: Therapeutic potential of natural antioxidants. Front. Cell Infect. Microbiol. 2021, 11, 654813. [Google Scholar] [CrossRef] [PubMed]
  127. Mingoti, M.E.D.; Bertollo, A.G.; Simões, J.L.B.; Francisco, G.R.; Bagatini, M.D.; Ignácio, Z.M. COVID-19, oxidative stress, and neuroinflammation in the depression route. J. Mol. Neurosci. 2022, 72, 1166–1181. [Google Scholar] [CrossRef] [PubMed]
  128. Matschke, J.; Lütgehetmann, M.; Hagel, C.; Sperhake, J.P.; Schröder, 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]
  129. 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]
  130. Liu, S.; Hossinger, A.; Heumüller, S.E.; Hornberger, A.; Buravlova, O.; Konstantoulea, K.; Müller, S.A.; Paulsen, L.; Rousseau, F.; Schymkowitz, J.; et al. Highly efficient intercellular spreading of protein misfolding mediated by viral ligand-receptor interactions. Nat. Commun. 2021, 12, 5739. [Google Scholar] [CrossRef]
  131. Idrees, D.; Kumar, V. SARS-CoV-2 spike protein interactions with amyloidogenic proteins: Potential clues to neurodegeneration. Biochem. Biophys. Res. Commun. 2021, 554, 94–98. [Google Scholar] [CrossRef]
  132. Dechant, G.; Neumann, H. Neurotrophins. Adv. Exp. Med. 2002, 513, 303–334. [Google Scholar] [CrossRef] [PubMed]
  133. Hallböök, F. Evolution of the vertebrate neurotrophin and Trk receptor gene families. Curr. Opin. Neurobiol. 1999, 9, 616–621. [Google Scholar] [CrossRef]
  134. Chao, M.V. Neurotrophins and their receptors: A convergence point for many signalling pathways. Nat. Rev. Neurosci. 2003, 4, 299–309. [Google Scholar] [CrossRef] [PubMed]
  135. Asgarzadeh, A.; Fouladi, N.; Asghariazar, V.; Sarabi, S.F.; Khiavi, H.A.; Mahmoudi, M.; Safarzadeh, E. Serum brain-derived neurotrophic factor (BDNF) in COVID-19 patients and its association with the COVID-19 manifestations. J. Mol. Neurosci. 2022, 72, 1820–1830. [Google Scholar] [CrossRef] [PubMed]
  136. Biamonte, F.; Re, A.; Balzamino, B.O.; Ciasca, G.; Santucci, D.; Napodano, C.; Nocca, G.; Fiorita, A.; Marino, M.; Basile, U.; et al. Circulating and salivary NGF and BDNF levels in SARS-CoV-2 infection: Potential predictor biomarkers of COVID-19 disease-preliminary data. J. Pers. Med. 2022, 12, 1877. [Google Scholar] [CrossRef] [PubMed]
  137. Minuzzi, L.G.; Seelaender, M.; Silva, B.S.A.; Cunha, E.D.B.B.; Deus, M.C.; Vasconcellos, F.T.F.; Marqueze, L.F.B.; Gadotti, A.C.; Baena, C.P.; Pereira, T.; et al. COVID-19 outcome relates with circulating bdnf, according to patient adiposity and age. Front. Nutr. 2021, 10, 784429. [Google Scholar] [CrossRef]
  138. Azoulay, D.; Shehadeh, M.; Chepa, S.; Shaoul, E.; Baroum, M.; Horowitz, N.A.; Kaykov, E. Recovery from SARS-CoV-2 infection is associated with serum BDNF restoration. J. Infect. 2020, 81, e79–e81. [Google Scholar] [CrossRef]
  139. Demir, B.; Beyazyüz, E.; Beyazyüz, M.; Çelikkol, A.; Albayrak, Y. Long-lasting cognitive effects of COVID-19: Is there a role of BDNF? Eur. Arch. Psychiatry. Clin. Neurosci. 2023, 273, 1339–1347. [Google Scholar] [CrossRef]
  140. Santiago, J.A.; Bottero, V.; Potashkin, J.A. Dissecting the molecular mechanisms of neurodegenerative diseases through network biology. Front. Aging. Neurosci. 2017, 9, 166. [Google Scholar] [CrossRef]
  141. Kim, G.H.; Kim, J.E.; Rhie, S.J.; Yoon, S. The role of oxidative stress in neurodegenerative diseases. Exp. Neurobiol. 2015, 24, 325–340. [Google Scholar] [CrossRef]
  142. Solleiro-Villavicencio, H.; Rivas-Arancibia, S. Effect of chronic oxidative stress on neuroinflammatory response mediated by CD4+T cells in neurodegenerative diseases. Front. Cell Neurosci. 2018, 12, 114. [Google Scholar] [CrossRef]
  143. Jellinger, K.A. Basic mechanisms of neurodegeneration: A critical update. J. Cell Mol. Med. 2010, 14, 457–487. [Google Scholar] [CrossRef]
  144. Mohd Sairazi, N.S.; Sirajudeen, K.N.S. Natural products and their bioactive compounds: Neuroprotective potentials against neurodegenerative diseases. Evid Based Complement Altern. Med. 2020, 2020, 6565396. [Google Scholar] [CrossRef] [PubMed]
  145. Rahman, M.H.; Bajgai, J.; Fadriquela, A.; Sharma, S.; Trinh, T.T.; Akter, R.; Jeong, Y.J.; Goh, S.H.; Kim, C.S.; Lee, K.J. Therapeutic potential of natural products in treating neurodegenerative disorders and their future prospects and challenges. Molecules 2021, 26, 5327. [Google Scholar] [CrossRef] [PubMed]
  146. Stephenson, J.; Nutma, E.; van der Valk, P.; Amor, S. Inflammation in CNS neurodegenerative diseases. Immunology 2018, 154, 204–219. [Google Scholar] [CrossRef] [PubMed]
  147. Chitnis, T.; Weiner, H.L. CNS inflammation and neurodegeneration. J. Clin. Investig. 2017, 127, 3577–3587. [Google Scholar] [CrossRef] [PubMed]
  148. Moujalled, D.; Strasser, A.; Liddell, J.R. Molecular mechanisms of cell death in neurological diseases. Cell Death Differ 2021, 28, 2029–2044. [Google Scholar] [CrossRef]
  149. Cui, J.; Zhao, S.; Li, Y.; Zhang, D.; Wang, B.; Xie, J.; Wang, J. Regulated cell death: Discovery, features and implications for neurodegenerative diseases. Cell Commun. Signal 2021, 19, 1–29. [Google Scholar] [CrossRef]
  150. Gorman, A.M. Neuronal cell death in neurodegenerative diseases: Recurring themes around protein handling: Apoptosis Review Series. J. Cell Mol. Med. 2008, 12, 2263–2280. [Google Scholar] [CrossRef]
  151. Dong, X.X.; Wang, Y.; Qin, Z.H. Molecular mechanisms of excitotoxicity and their relevance to pathogenesis of neurodegenerative diseases. Acta Pharmacol. Sin. 2009, 30, 379–387. [Google Scholar] [CrossRef]
  152. Soto, C.; Pritzkow, S. Protein misfolding, aggregation, and conformational strains in neurodegenerative diseases. Nat. Neurosci. 2018, 21, 1332–1340. [Google Scholar] [CrossRef]
  153. Nguyen, K.; Hoffman, H.; Chakkamparambil, B.; Grossberg, G.T. Evaluation of rivastigmine in Alzheimer’s disease. Neurodegener. Dis. Manag. 2021, 11, 35–48. [Google Scholar] [CrossRef]
  154. Yiannopoulou, K.G.; Papageorgiou, S.G. Current and future treatments in Alzheimer disease: An update. J. Cent. Nerv. Syst. Dis. 2020, 12, 117957352090739. [Google Scholar] [CrossRef]
  155. Folch, J.; Busquets, O.; Ettcheto, M.; Sánchez-López, E.; Castro-Torres, R.D.; Verdaguer, E.; Garcia, M.L.; Olloquequi, J.; Casadesús, G.; Beas-Zarate, C.; et al. Memantine for the treatment of dementia: A review on its current and future applications. J. Alzheimer’s Dis. 2018, 62, 1223–1240. [Google Scholar] [CrossRef]
  156. González-Fuentes, J.; Selva, J.; Moya, C.; Castro-Vázquez, L.; Lozano, M.V.; Marcos, P.; Plaza-Oliver, M.; Rodríguez-Robledo, V.; Santander-Ortega, M.J.; Villaseca-González, N.; et al. Neuroprotective natural molecules, from food to brain. Front. Neurosci. 2018, 12, 721. [Google Scholar] [CrossRef] [PubMed]
  157. Limanaqi, F.; Biagioni, F.; Mastroiacovo, F.; Polzella, M.; Lazzeri, G.; Fornai, F. Merging the multi-target effects of phytochemicals in neurodegeneration: From oxidative stress to protein aggregation and inflammation. Antioxidants 2020, 9, 1022. [Google Scholar] [CrossRef] [PubMed]
  158. Sahebnasagh, A.; Eghbali, S.; Saghafi, F.; Sureda, A.; Avan, R. Neurohormetic phytochemicals in the pathogenesis of neurodegenerative diseases. Immun. Ageing. 2022, 19, 1–16. [Google Scholar] [CrossRef] [PubMed]
  159. Yadav, D.K. Potential therapeutic strategies of phytochemicals in neurodegenerative disorders. Curr. Top Med. Chem. 2021, 21, 2814–2838. [Google Scholar] [CrossRef] [PubMed]
  160. Krüger, N.; Kronenberger, T.; Xie, H.; Rocha, C.; Pöhlmann, S.; Su, H.; Xu, Y.; Laufer, S.A.; Pillaiyar, T. Discovery of Polyphenolic Natural Products as SARS-CoV-2 Mpro Inhibitors for COVID-19. Pharm 2023, 16, 190. [Google Scholar] [CrossRef]
  161. Wang, Z.; Wang, N.; Yang, L.; Song, X.Q. Bioactive natural products in COVID-19 therapy. Front. Pharmacol. 2022, 19, 926507. [Google Scholar] [CrossRef]
  162. Song, J.; Zhang, L.; Xu, Y.; Yang, D.; Zhang, L.; Yang, S.; Zhang, W.; Wang, J.; Tian, S.; Yang, S.; et al. The comprehensive study on the therapeutic effects of baicalein for the treatment of COVID-19 in vivo and in vitro. Biochem. Pharmacol. 2021, 183, 114302. [Google Scholar] [CrossRef]
  163. Maher, P. The potential of flavonoids for the treatment of neurodegenerative diseases. Int. J. Mol. Sci. 2019, 20, 3056. [Google Scholar] [CrossRef]
  164. Cui, X.; Lin, Q.; Liang, Y. Plant-derived antioxidants protect the nervous system from aging by inhibiting oxidative stress. Front. Aging Neurosci. 2020, 12, 209. [Google Scholar] [CrossRef] [PubMed]
  165. Matias, I.; Buosi, A.S.; Gomes, F.C.A. Functions of flavonoids in the central nervous system: Astrocytes as targets for natural compounds. Neurochem. Int. 2016, 95, 85–91. [Google Scholar] [CrossRef] [PubMed]
  166. Velásquez, D.; Corella, D.; Zuñiga, B.; Domínguez, A.; Montiel, M.; Salazar, N.; Rodrigo-Garcia, J.; Villegas-Ochoa, M.A.; González-Aguilar, G.A. Phenolic compounds that cross the blood-brain barrier exert positive health effects as central nervous system antioxidants. Food Funct. 2021, 12, 10356–10369. [Google Scholar] [CrossRef]
  167. Zhao, L.; Wang, J.L.; Wang, Y.R.; Fa, X.Z. Apigenin attenuates copper-mediated β-amyloid neurotoxicity through antioxidation, mitochondrion protection and MAPK signal inactivation in an AD cell model. Brain Res. 2013, 1492, 33–45. [Google Scholar] [CrossRef] [PubMed]
  168. Choi, A.Y.; Choi, J.H.; Lee, J.Y.; Yoon, K.S.; Choe, W.; Ha, J.; Yeo, E.J.; Kang, I. Apigenin protects HT22 murine hippocampal neuronal cells against endoplasmic reticulum stress-induced apoptosis. Neurochem. Int. 2010, 57, 143–152. [Google Scholar] [CrossRef] [PubMed]
  169. Balez, R.; Steiner, N.; Engel, M.; Muñoz, S.S.; Lum, J.S.; Wu, Y.; Wang, D.; Vallotton, P.; Sachdev, P.; O’Connor, M.; et al. Neuroprotective effects of apigenin against inflammation, neuronal excitability and apoptosis in an induced pluripotent stem cell model of Alzheimer’s disease. Sci. Rep. 2016, 6, 31450. [Google Scholar] [CrossRef]
  170. Rezai-Zadeh, K.; Ehrhart, J.; Bai, Y.; Sanberg, P.R.; Bickford, P.; Tan, J.; Douglas, R.D. Apigenin and luteolin modulate microglial activation via inhibition of STAT1-induced CD40 expression. J. Neuroinflam. 2008, 5, 1–10. [Google Scholar] [CrossRef]
  171. Anusha, C.; Sumathi, T.; Joseph, L.D. Protective role of apigenin on rotenone induced rat model of Parkinson’s disease: Suppression of neuroinflammation and oxidative stress mediated apoptosis. Chem. Biol. Interact. 2017, 269, 67–79. [Google Scholar] [CrossRef]
  172. Khan, H.; Ullah, H.; Aschner, M.; Cheang, W.S.; Akkol, E.K. Neuroprotective effects of quercetin in Alzheimer’s disease. Biomolecules 2020, 10, 59. [Google Scholar] [CrossRef]
  173. Abd El Mohsen, M.M.; Kuhnle, G.; Rechner, A.R.; Schroeter, H.; Rose, S.; Jenner, P.; Rice-Evans, C.A. Uptake and metabolism of epicatechin and its access to the brain after oral ingestion. Free Radic. Biol. Med. 2002, 33, 1693–1702. [Google Scholar] [CrossRef]
  174. Van Praag, H.; Lucero, M.J.; Yeo, G.W.; Stecker, K.; Heivand, N.; Zhao, C.; Yip, E.; Afanador, M.; Schroeter, H.; Hammerstone, J.; et al. Plant-derived flavanol (-)epicatechin enhances angiogenesis and retention of spatial memory in mice. J. Neurosci. 2007, 27, 5869–5878. [Google Scholar] [CrossRef] [PubMed]
  175. Nichols, M.; Zhang, J.; Polster, B.M.; Elustondo, P.A.; Thirumaran, A.; Pavlov, E.V.; Robertson, G.S. Synergistic neuroprotection by epicatechin and quercetin: Activation of convergent mitochondrial signaling pathways. Neuroscience 2015, 308, 75–94. [Google Scholar] [CrossRef] [PubMed]
  176. Oztanir, M.N.; Ciftci, O.; Cetin, A.; Aladag, M.A. Hesperidin attenuates oxidative and neuronal damage caused by global cerebral ischemia/reperfusion in a C57BL/J6 mouse model. Neurol. Sci. 2014, 35, 1393–1399. [Google Scholar] [CrossRef] [PubMed]
  177. Wang, J.J.; Cui, P. Neohesperidin attenuates cerebral ischemia-reperfusion injury via inhibiting the apoptotic pathway and activating the Akt/Nrf2/HO-1 pathway. J. Asian Nat. Prod. Res. 2013, 15, 1023–1037. [Google Scholar] [CrossRef] [PubMed]
  178. Vazquez-Calvo, A.; Jimenez de Oya, N.; Martin-Acebes, M.A.; Garcia-Moruno, E.; Saiz, J.C. Antiviral properties of the natural polyphenols delphinidin and epigallocatechin gallate against the flaviviruses West Nile Virus, Zika Virus, and Dengue Virus. Front. Microbiol. 2017, 8, 1314. [Google Scholar] [CrossRef]
  179. Johari, J.; Kianmehr, A.; Mustafa, M.R.; Abubakar, S.; Zandi, K. Antiviral activity of baicalein and quercetin against the Japanese encephalitis virus. Int. J. Mol. Sci. 2012, 13, 16020–16045. [Google Scholar] [CrossRef] [PubMed]
  180. Zandi, K.; Teoh, B.T.; Sam, S.S.; Wong, P.F.; Mustafa, M.; Abubakar, S. Antiviral activity of four types of bioflavonoid against dengue virus type-2. Virol. J. 2011, 8, 1–11. [Google Scholar] [CrossRef]
  181. Wang, H.; Wang, H.; Cheng, H.; Che, Z. Ameliorating effect of luteolin on memory impairment in an Alzheimer’s disease model. Mol. Med. Rep. 2016, 13, 4215–4220. [Google Scholar] [CrossRef]
  182. Chen, H.Q.; Jin, Z.Y.; Wang, X.J.; Xu, X.M.; Deng, L.; Zhao, J.W. Luteolin protects dopaminergic neurons from inflammation-induced injury through inhibition of microglial activation. Neurosci. Lett. 2008, 448, 175–179. [Google Scholar] [CrossRef]
  183. Lee, J.K.; Kim, S.Y.; Kim, Y.S.; Lee, W.H.; Hwang, D.H.; Lee, J.Y. Suppression of the TRIF-dependent signaling pathway of Toll-like receptors by luteolin. Biochem. Pharmacol. 2009, 77, 1391–1400. [Google Scholar] [CrossRef]
  184. Paris, D.; Mathura, V.; Ait-Ghezala, G.; Beaulieu-Abdelahad, D.; Patel, N.; Bachmeier, C.; Mullan, M. Flavonoids lower Alzheimer’s Abeta production via an NFkappaB dependent mechanism. Bioinformation 2011, 6, 229–236. [Google Scholar] [CrossRef]
  185. Katalinić, M.; Rusak, G.; Domaćinović Barović, J.; Šinko, G.; Jelić, D.; Antolović, R.; Kovarik, Z. Structural aspects of flavonoids as inhibitors of human butyrylcholinesterase. Eur. J. Med. Chem. 2010, 45, 186–192. [Google Scholar] [CrossRef]
  186. Alvarado, W.; Perez, G.; Menéndez, C.; Byléhn, F.; de Pablo, J.J. Molecular characterization of COVID-19 therapeutics: Luteolin as an allosteric modulator of the spike protein of SARS-CoV-2. Mol. Syst. Des. Eng. 2022, 7, 58–66. [Google Scholar] [CrossRef]
  187. Yu, R.; Chen, L.; Lan, R.; Shen, R.; Li, P. Computational screening of antagonists against the SARS-CoV-2 (COVID-19) coronavirus by molecular docking. Int. J. Antimicrob. Agents. 2020, 56, 106012. [Google Scholar] [CrossRef]
  188. Khazdair, M.R.; Anaeigoudari, A.; Agbor, G.A. Anti-viral and anti-inflammatory effects of kaempferol and quercetin and COVID-2019: A scoping review. Asian. Pac. J. Trop. Biomed. 2021, 11, 327–334. [Google Scholar] [CrossRef]
  189. Hämäläinen, M.; Nieminen, R.; Vuorela, P.; Heinonen, M.; Moilanen, E. Anti-inflammatory effects of flavonoids: Genistein, kaempferol, quercetin, and daidzein inhibit STAT-1 and NF-κB activations, whereas flavone, isorhamnetin, naringenin, and pelargonidin inhibit only NF-κB activation along with their inhibitory effect on iNOS expression and NO production in activated macrophages. Mediat. Inflamm. 2007, 2007, 45673. [Google Scholar] [CrossRef]
  190. Rangel-Ordóñez, L.N.M.; Schubert-Zsilavecz, M.; Wurglics, M. Plasma levels and distribution of flavonoids in rat brain after single and repeated doses of standardized Ginkgo biloba extract EGb 761®. Planta. Med. 2010, 76, 1683–1690. [Google Scholar] [CrossRef]
  191. Chang, S.; Li, X.; Zheng, Y.; Shi, H.; Zhang, D.; Jing, B.; Chen, Z.; Qian, G.; Zhao, G. Kaempferol exerts a neuroprotective effect to reduce neuropathic pain through TLR4/NF-ĸB signaling pathway. Phytother. Res. 2022, 36, 1678–1691. [Google Scholar] [CrossRef]
  192. Ahmed, H.; Khan, M.A.; Ali Zaidi, S.A.; Muhammad, S. In silico and in vivo: Evaluating the Therapeutic Potential of Kaempferol, Quercetin, and Catechin to Treat Chronic Epilepsy in a Rat Model. Front. Bioeng. Biotechnol. 2021, 9, 754952. [Google Scholar] [CrossRef]
  193. Lee, S.; Seol, H.S.; Eom, S.; Lee, J.; Kim, C.; Park, J.H.; Kim, T.H.; Lee, J.H. Hydroxy Pentacyclic Triterpene Acid, Kaempferol, Inhibits the Human 5-Hydroxytryptamine Type 3A Receptor Activity. Int. J. Mol. Sci. 2022, 23, 544. [Google Scholar] [CrossRef]
  194. Balkis, A.; Tran, K.; Lee, Y.Z.; Balkis, K.N.; Ng, K. Screening Flavonoids for Inhibition of Acetylcholinesterase Identified Baicalein as the Most Potent Inhibitor. J. Agric. Sci. 2015, 7, 26–35. [Google Scholar] [CrossRef]
  195. Khan, A.; Heng, W.; Wang, Y.; Qiu, J.; Wei, X.; Peng, S.; Saleem, S.; Khan, M.; Ali, S.S.; Wei, D.Q. In silico and in vitro evaluation of kaempferol as a potential inhibitor of the SARS-CoV-2 main protease (3CLpro). Phytother. Res. 2021, 35, 2841–2845. [Google Scholar] [CrossRef] [PubMed]
  196. Shaldam, M.A.; Yahya, G.; Mohamed, N.H.; Abdel-Daim, M.M.; Al Naggar, Y. In silico screening of potent bioactive compounds from honeybee products against COVID-19 target enzymes. Env. Sci. Pollut. Res. Int. 2021, 28, 40507–40514. [Google Scholar] [CrossRef] [PubMed]
  197. Hussain, G.; Rasul, A.; Anwar, H.; Aziz, N.; Razzaq, A.; Wei, W.; Ali, M.; Li, J.; Li, X. Role of plant derived alkaloids and their mechanism in neurodegenerative disorders. Int. J. Biol. Sci. 2018, 14, 341–357. [Google Scholar] [CrossRef]
  198. Murray, A.P.; Faraoni, M.B.; Castro, M.J.; Alza, N.P.; Cavallaro, V. Natural AChE inhibitors from plants and their contribution to Alzheimer’s disease therapy. Curr. Neuropharmacol. 2013, 11, 388–413. [Google Scholar] [CrossRef] [PubMed]
  199. Nazifi, M.; Oryan, S.; Esfahani, D.E.; Ashrafpoor, M. The functional effects of piperine and piperine plus donepezil on hippocampal synaptic plasticity impairment in rat model of Alzheimer’s disease. Life Sci. 2021, 265, 118802. [Google Scholar] [CrossRef]
  200. Shrivastava, P.; Vaibhav, K.; Tabassum, R.; Khan, A.; Ishrat, T.; Khan, M.M.; Ahmad, A.; Islam, F.; Safhi, M.M.; Islam, F. Anti-apoptotic and anti-inflammatory effect of Piperine on 6-OHDA induced Parkinson’s rat model. J. Nutr. Biochem. 2013, 24, 680–687. [Google Scholar] [CrossRef] [PubMed]
  201. Mishra, A.; Punia, J.K.; Bladen, C.; Zamponi, G.W.; Goel, R.K. Anticonvulsant mechanisms of piperine, a piperidine alkaloid. Channels 2015, 9, 317–323. [Google Scholar] [CrossRef]
  202. Kong, Y.R.; Tay, K.C.; Su, Y.X.; Khaw, K.Y.; Wong, C.K.; Tan, W.N. Potential of naturally derived alkaloids as multi-targeted therapeutic agents for neurodegenerative diseases. Molecules 2021, 26, 728. [Google Scholar] [CrossRef]
  203. Ai, X.; Yu, P.; Peng, L.; Luo, L.; Liu, J.; Li, S.; Lai, X.; Luan, F.; Meng, X. Berberine: A Review of its Pharmacokinetics Properties and Therapeutic Potentials in Diverse Vascular Diseases. Front. Pharmacol. 2021, 12, 762654. [Google Scholar] [CrossRef]
  204. Jiang, W.; Li, S.; Li, X. Therapeutic potential of berberine against neurodegenerative diseases. Sci. China Life Sci. 2015, 58, 564–569. [Google Scholar] [CrossRef] [PubMed]
  205. Yuan, N.N.; Cai, C.Z.; Wu, M.Y.; Su, H.X.; Li, M.; Lu, J.H. Neuroprotective effects of berberine in animal models of Alzheimer’s disease: A systematic review of pre-clinical studies. BMC Complement Altern. Med. 2019, 19, 1–10. [Google Scholar] [CrossRef] [PubMed]
  206. Peng, W.H.; Wu, C.R.; Chen, C.S.; Chen, C.F.; Leu, Z.C.; Hsieh, M.T. Anxiolytic effect of berberine on exploratory activity of the mouse in two experimental anxiety models: Interaction with drugs acting at 5-HT receptors. Life Sci. 2004, 75, 2451–2462. [Google Scholar] [CrossRef] [PubMed]
  207. Lu, J.; Cao, Y.; Cheng, K.; Xu, B.; Wang, T.; Yang, Q.; Yang, Q.; Feng, X.; Xia, Q. Berberine regulates neurite outgrowth through AMPK-dependent pathways by lowering energy status. Exp. Cell Res. 2015, 334, 194–206. [Google Scholar] [CrossRef]
  208. Ma, X.; Jiang, Y.; Wu, A.; Chen, X.; Pi, R.; Liu, M.; Liu, Y. Berberine attenuates experimental autoimmune encephalomyelitis in C57 BL/6 mice. PLoS ONE 2010, 5, e13489. [Google Scholar] [CrossRef]
  209. De Oliveira, J.S.; Abdalla, F.H.; Dornelles, G.L.; Palma, T.V.; Signor, C.; da Silva Bernardi, J.; Baldissarelli, J.; Lenz, L.S.; de Oliveira, V.A.; Chitolina Schetinger, M.R.; et al. Neuroprotective effects of berberine on recognition memory impairment, oxidative stress, and damage to the purinergic system in rats submitted to intracerebroventricular injection of streptozotocin. Psychopharmacology 2019, 236, 641–655. [Google Scholar] [CrossRef]
  210. Hussien, H.M.; Abd-Elmegied, A.; Ghareeb, D.A.; Hafez, H.S.; Ahmed, H.E.A.; El-moneam, N.A. Neuroprotective effect of berberine against environmental heavy metals-induced neurotoxicity and Alzheimer’s-like disease in rats. Food Chem. Toxicol. 2018, 111, 432–444. [Google Scholar] [CrossRef]
  211. Tian, L.; Ri, H.; Qi, J.; Fu, P. Berberine elevates mitochondrial membrane potential and decreases reactive oxygen species by inhibiting the Rho/ROCK pathway in rats with diabetic encephalopathy. Mol. Pain 2021, 17, 1744806921996101. [Google Scholar] [CrossRef]
  212. Kassab, R.B.; Vasicek, O.; Ciz, M.; Lojek, A.; Perecko, T. The effects of berberine on reactive oxygen species production in human neutrophils and in cell-free assays. Interdiscip. Toxicol. 2017, 10, 61–65. [Google Scholar] [CrossRef]
  213. Imenshahidi, M.; Hosseinzadeh, H. Chapter 14—Berberine neuroprotection and antioxidant activity. In Oxidative Stress and Dietary Antioxidants in Neurological Diseases; Martin Colin, R., Preedy Victor, R., Eds.; Elsevier Inc.: Amsterdam, The Netherlands, 2020; pp. 199–216. [Google Scholar]
  214. Chu, M.; Chen, X.; Wang, J.; Guo, L.; Wang, Q.; Gao, Z.; Kang, J.; Zhang, M.; Feng, J.; Guo, Q.; et al. Polypharmacology of berberine based on multi-target binding motifs. Front. Pharmacol. 2018, 9, 801. [Google Scholar] [CrossRef]
  215. Pohanka, M. Inhibitors of acetylcholinesterase and butyrylcholinesterase meet immunity. Int. J. Mol. Sci. 2014, 15, 9809–9825. [Google Scholar] [CrossRef] [PubMed]
  216. Imenshahidi, M.; Qaredashi, R.; Hashemzaei, M.; Hosseinzadeh, H. Inhibitory effect of Berberis vulgaris aqueous extract on acquisition and reinstatement effects of morphine in conditioned place preferences (CPP) in mice. Jundishapur J. Nat. Pharm. Prod. 2014, 9, e16145. [Google Scholar] [CrossRef] [PubMed]
  217. Huang, M.; Jiang, X.; Liang, Y.; Liu, Q.; Chen, S.; Guo, Y. Berberine improves cognitive impairment by promoting autophagic clearance and inhibiting production of beta-amyloid in APP/tau/PS1 mouse model of Alzheimer’s disease. Exp. Gerontol. 2017, 91, 25–33. [Google Scholar] [CrossRef] [PubMed]
  218. Jiang, W.; Wei, W.; Gaertig, M.A.; Li, S.; Li, X.-J. Therapeutic Effect of Berberine on Huntington’s Disease Transgenic Mouse Model. PLoS ONE 2015, 10, e0134142. [Google Scholar] [CrossRef]
  219. Rodriguez-Rodriguez, B.A.; Noval, M.G.; Kaczmarek, M.E.; Jang, K.K.; Thannickal, S.A.; Kottkamp, A.C.; Brown, R.S.; Kielian, M.; Cadwell, K.; Stapleford, K.A. Atovaquone and berberine chloride reduce SARS-CoV-2 replication in vitro. Viruses 2021, 13, 2437. [Google Scholar] [CrossRef]
  220. Varghese, F.S.; van Woudenbergh, E.; Overheul, G.J.; Eleveld, M.J.; Kurver, L.; van Heerbeek, N.; van Laarhoven, A.; Miesen, P.; Den Hartog, G.; de Jonge, M.I.; et al. Berberine and obatoclax inhibit SARS-CoV-2 replication in primary human nasal epithelial cells in vitro. Viruses 2021, 13, 282. [Google Scholar] [CrossRef]
  221. Chowdhury, P. In silico investigation of phytoconstituents from Indian medicinal herb ‘Tinospora cordifolia (giloy)’ against SARS-CoV-2 (COVID-19) by molecular dynamics approach. J. Biomol. Struct. Dyn. 2021, 39, 6792–6809. [Google Scholar] [CrossRef]
  222. Narkhede, R.R.; Pise, A.V.; Cheke, R.S.; Shinde, S.D. Recognition of natural products as potential inhibitors of COVID-19 main protease (Mpro): In-silico evidences. Nat. Prod. Bioprospect. 2020, 10, 297–306. [Google Scholar] [CrossRef]
  223. Cao, J.; Li, L.; Xiong, L.; Wang, C.; Chen, Y.; Zhang, X. Research on the mechanism of berberine in the treatment of COVID-19 pneumonia pulmonary fibrosis using network pharmacology and molecular docking. Phytomed. Plus 2022, 2, 100252. [Google Scholar] [CrossRef]
  224. Guan, X.; Li, X.; Yang, X.; Yan, J.; Shi, P.; Ba, L.; Cao, Y.; Wang, P. The neuroprotective effects of carvacrol on ischemia/reperfusion-induced hippocampal neuronal impairment by ferroptosis mitigation. Life Sci. 2019, 235, 116795. [Google Scholar] [CrossRef]
  225. Li, Z.; Hua, C.; Pan, X.; Fu, X.; Wu, W. Carvacrol exerts neuroprotective effects via suppression of the inflammatory response in middle cerebral artery Occlusion Rats. Inflammation 2016, 39, 1566–1572. [Google Scholar] [CrossRef] [PubMed]
  226. Chen, W.; Xu, B.; Xiao, A.; Liu, L.; Fang, X.; Liu, R.; Turlova, E.; Barszczyk, A.; Zhong, X.; Sun, C.L.F.; et al. TRPM7 inhibitor carvacrol protects brain from neonatal hypoxic-ischemic injury. Mol. Brain 2015, 8, 1–13. [Google Scholar] [CrossRef] [PubMed]
  227. Li, Y.; He, D.; Zhang, X.; Liu, Z.; Zhang, X.; Dong, L.; Xing, Y.; Wang, C.; Qiao, H.; Zhu, C.; et al. Protective effect of celastrol in rat cerebral ischemia model: Down-regulating p-JNK, p-c-Jun and NF-κB. Brain Res. 2012, 1464, 8–13. [Google Scholar] [CrossRef] [PubMed]
  228. Choi, B.S.; Kim, H.; Lee, H.J.; Sapkota, K.; Park, S.E.; Kim, S.; Kim, S.J. Celastrol from ‘Thunder God Vine’ Protects SH-SY5Y cells through the preservation of mitochondrial function and inhibition of p38 MAPK in a rotenone model of parkinson’s disease. Neurochem. Res. 2014, 39, 84–96. [Google Scholar] [CrossRef]
  229. Szwajgier, D.; Borowiec, K.; Pustelniak, K. The neuroprotective effects of phenolic acids: Molecular mechanism of action. Nutrients 2017, 9, 477. [Google Scholar] [CrossRef]
  230. Caruso, G.; Godos, J.; Privitera, A.; Lanza, G.; Castellano, S.; Chillemi, A.; Bruni, O.; Ferri, R.; Caraci, F.; Grosso, G. Phenolic acids and prevention of cognitive decline: Polyphenols with a neuroprotective role in cognitive disorders and Alzheimer’s disease. Nutrients 2022, 14, 819. [Google Scholar] [CrossRef]
  231. Gay, N.H.; Phopin, K.; Suwanjang, W.; Songtawee, N.; Ruankham, W.; Wongchitrat, P.; Prachayasittikul, S.; Prachayasittikul, V. Neuroprotective Effects of Phenolic and Carboxylic Acids on Oxidative Stress-Induced Toxicity in Human Neuroblastoma SH-SY5Y Cells. Neurochem. Res. 2018, 43, 619–636. [Google Scholar] [CrossRef]
  232. Sun, J.; Li, Y.Z.; Ding, Y.H.; Wang, J.; Geng, J.; Yang, H.; Ren, J.; Tang, J.Y.; Gao, J. Neuroprotective effects of gallic acid against hypoxia/reoxygenation-induced mitochondrial dysfunctions in vitro and cerebral ischemia/reperfusion injury in vivo. Brain Res. 2014, 1589, 126–139. [Google Scholar] [CrossRef]
  233. Nabavi, S.F.; Habtemariam, S.; Di Lorenzo, A.; Sureda, A.; Khanjani, S.; Nabavi, S.M.; Daglia, M. Post-stroke depression modulation and in vivo antioxidant activity of gallic acid and its synthetic derivatives in a murine model system. Nutrients 2016, 8, 248. [Google Scholar] [CrossRef]
  234. Liang, G.; Shi, B.; Luo, W.; Yang, J. The protective effect of caffeic acid on global cerebral ischemia-reperfusion injury in rats. Behav. Brain Funct. 2015, 11, 1–10. [Google Scholar] [CrossRef]
  235. Zandi, K.; Musall, K.; Oo, A.; Cao, D.; Liang, B.; Hassandarvish, P.; Lan, S.; Slack, R.L.; Kirby, K.A.; Bassit, L.; et al. Baicalein and baicalin inhibit SARS-CoV-2 RNA-Dependent-RNA polymerase. Microorganisms 2021, 9, 893. [Google Scholar] [CrossRef] [PubMed]
  236. Li, Q.; Li, Q.Q.; Jia, J.N.; Sun, Q.Y.; Zhou, H.H.; Jin, W.L.; Mao, X.Y. Baicalein Exerts Neuroprotective Effects in FeCl(3)-Induced Posttraumatic Epileptic Seizures via Suppressing Ferroptosis. Front. Pharmacol. 2019, 10, 638. [Google Scholar] [CrossRef] [PubMed]
  237. CSPC ZhongQi Pharmaceutical Technology Co., Ltd. A Randomized, Double-Blind, Placebo-Controlled, Multicenter and Phase IIa Clinical Trial for the Effectiveness and Safety of Baicalein Tablets in the Treatment of Improve other Aspects of Healthy Adult with Influenza Fever. ClinicalTrials.gov (ID:NCT03830684). Available online: https://clinicaltrials.gov/study/NCT03830684?term=belcalein&rank=1 (accessed on 30 August 2023).
  238. Munafo, F.; Donati, E.; Brindani, N.; Ottonello, G.; Armirotti, A.; De Vivo, M. Quercetin and luteolin are single-digit micromolar inhibitors of the SARS-CoV-2 RNA-dependent RNA polymerase. Sci. Rep. 2022, 12, 10571. [Google Scholar] [CrossRef] [PubMed]
  239. Kempuraj, D.; Thangavel, R.; Kempuraj, D.D.; Ahmed, M.E.; Selvakumar, G.P.; Raikwar, S.P.; Zaheer, S.A.; Iyer, S.S.; Govindarajan, R.; Chandrasekaran, P.N.; et al. Neuroprotective effects of flavone luteolin in neuroinflammation and neurotrauma. Biofactors 2021, 47, 190–197. [Google Scholar] [CrossRef] [PubMed]
  240. Montreal-Heart-Institute. Study of Hesperidin Therapy on COVID-19 Symptoms (HESPERIDIN) (Hesperidin). ClinicalTrials.gov (ID:NCT04715932). Available online: https://classic.clinicaltrials.gov/ct2/show/NCT04715932?term=hesperidin&cond=COVID-19&draw=2&rank=1 (accessed on 30 August 2023).
  241. Das, S.; Sarmah, S.; Lyndem, S.; Singha Roy, A. An investigation into the identification of potential inhibitors of SARS-CoV-2 main protease using molecular docking study. J. Biomol. Struct. Dyn. 2021, 39, 3347–3357. [Google Scholar] [CrossRef]
  242. Cheng, F.J.; Huynh, T.K.; Yang, C.S.; Hu, D.W.; Shen, Y.C.; Tu, C.Y.; Wu, Y.C.; Tang, C.H.; Huang, W.C.; Chen, Y.; et al. Hesperidin is a Potential Inhibitor against SARS-CoV-2 Infection. Nutrients 2021, 13, 2800. [Google Scholar] [CrossRef]
  243. Kumar, S.; Paul, P.; Yadav, P.; Kaul, R.; Maitra, S.S.; Jha, S.K.; Chaari, A. A multi-targeted approach to identify potential flavonoids against three targets in the SARS-CoV-2 life cycle. Comput. Biol. Med. 2022, 142, 105231. [Google Scholar] [CrossRef]
  244. Welbat, J.U.; Naewla, S.; Pannangrong, W.; Sirichoat, A.; Aranarochana, A.; Wigmore, P. Neuroprotective effects of hesperidin against methotrexate-induced changes in neurogenesis and oxidative stress in the adult rat. Biochem. Pharmacol. 2020, 178, 114083. [Google Scholar] [CrossRef]
  245. Justin Thenmozhi, A.; Raja, T.R.; Janakiraman, U.; Manivasagam, T. Neuroprotective effect of hesperidin on aluminium chloride induced Alzheimer’s disease in Wistar rats. Neurochem. Res. 2015, 40, 767–776. [Google Scholar] [CrossRef]
  246. Hajialyani, M.; Hosein Farzaei, M.; Echeverria, J.; Nabavi, S.M.; Uriarte, E.; Sobarzo-Sanchez, E. Hesperidin as a neuroprotective agent: A review of animal and clinical evidence. Molecules 2019, 24, 648. [Google Scholar] [CrossRef]
  247. Abian, O.; Ortega-Alarcon, D.; Jimenez-Alesanco, A.; Ceballos-Laita, L.; Vega, S.; Reyburn, H.T.; Rizzuti, B.; Velazquez-Campoy, A. Structural stability of SARS-CoV-2 3CLpro and identification of quercetin as an inhibitor by experimental screening. Int. J. Biol. Macromol. 2020, 164, 1693–1703. [Google Scholar] [CrossRef]
  248. Manjunathan, R.; Periyaswami, V.; Mitra, K.; Rosita, A.S.; Pandya, M.; Selvaraj, J.; Ravi, L.; Devarajan, N.; Doble, M. Molecular docking analysis reveals the functional inhibitory effect of Genistein and Quercetin on TMPRSS2: SARS-CoV-2 cell entry facilitator spike protein. BMC Bioinform. 2022, 23, 180. [Google Scholar] [CrossRef] [PubMed]
  249. Corona, A.; Wycisk, K.; Talarico, C.; Manelfi, C.; Milia, J.; Cannalire, R.; Esposito, F.; Gribbon, P.; Zaliani, A.; Iaconis, D.; et al. Natural compounds inhibit SARS-CoV-2 nsp13 unwinding and ATPase enzyme activities. ACS Pharmacol. Transl. Sci. 2022, 5, 226–239. [Google Scholar] [CrossRef] [PubMed]
  250. Costa, L.G.; Garrick, J.M.; Roque, P.J.; Pellacani, C. Mechanisms of Neuroprotection by Quercetin: Counteracting Oxidative Stress and More. Oxid. Med. Cell Longev. 2016, 2016, 2986796. [Google Scholar] [CrossRef] [PubMed]
  251. Grewal, A.K.; Singh, T.G.; Sharma, D.; Sharma, V.; Singh, M.; Rahman, M.H.; Najda, A.; Walasek-Janusz, M.; Kamel, M.; Albadrani, G.M.; et al. Mechanistic insights and perspectives involved in neuroprotective action of quercetin. Biomed. Pharmacother. 2021, 140, 111729. [Google Scholar] [CrossRef]
  252. King-Edward-Medical-University. Nutritional Supplementation of Flavonoids Quercetin and Curcumin for Early Mild Symptoms of COVID-19. ClinicalTrials.gov (IDr: NCT05130671). Available online: https://classic.clinicaltrials.gov/ct2/show/NCT05130671?term=flavonoid&cond=COVID-19&draw=2&rank=1 (accessed on 30 August 2023).
  253. Pawar, K.S.; Mastud, R.N.; Pawar, S.K.; Pawar, S.S.; Bhoite, R.R.; Bhoite, R.R.; Kulkarni, M.V.; Deshpande, A.R. Oral curcumin with piperine as adjuvant therapy for the treatment of COVID-19: A randomized clinical trial. Front. Pharmacol. 2021, 12, 669362. [Google Scholar] [CrossRef]
  254. Nag, A.; Paul, S.; Banerjee, R.; Kundu, R. In silico study of some selective phytochemicals against a hypothetical SARS-CoV-2 spike RBD using molecular docking tools. Comput. Biol. Med. 2021, 137, 104818. [Google Scholar] [CrossRef]
  255. Amperayani, K.R.; Varadhi, G.; Oruganti, B.; Parimi, U.D. Molecular dynamics and absolute binding free energy studies of piperine derivatives as potential inhibitors of SARS-CoV-2 main protease. J. Biomol. Struct. Dyn. 2023. [Google Scholar] [CrossRef]
  256. Hua, S.; Liu, J.; Zhang, Y.; Li, J.; Zhang, X.; Dong, L.; Zhao, Y.; Fu, X. Piperine as a neuroprotective functional component in rats with cerebral ischemic injury. Food Sci. Nutr. 2019, 7, 3443–3451. [Google Scholar] [CrossRef]
  257. Kumar, S.; Kashyap, P.; Chowdhury, S.; Kumar, S.; Panwar, A.; Kumar, A. Identification of phytochemicals as potential therapeutic agents that binds to Nsp15 protein target of coronavirus (SARS-CoV-2) that are capable of inhibiting virus replication. Phytomedicine 2021, 85, 153317. [Google Scholar] [CrossRef]
  258. Zhang, X.H.; Peng, L.; Zhang, J.; Dong, Y.P.; Wang, C.J.; Liu, C.; Xia, D.Y.; Zhang, X.S. Berberine ameliorates subarachnoid hemorrhage injury via induction of sirtuin 1 and inhibiting HMGB1/Nf-kappaB pathway. Front. Pharmacol. 2020, 11, 1073. [Google Scholar] [CrossRef] [PubMed]
  259. Khadilkar, A.; Bunch, Z.L.; Wagoner, J.; Ravindran, V.; Oda, J.M.; Vidar, W.S.; Clark, T.N.; Manwill, P.K.; Todd, D.A.; Barr, S.A.; et al. Modulation of in Vitro SARS-CoV-2 Infection by Stephania tetrandra and its alkaloid constituents. J. Nat. Prod. 2023, 86, 1061–1073. [Google Scholar] [CrossRef] [PubMed]
  260. Heister, P.M.; Poston, R.N. Pharmacological hypothesis: TPC2 antagonist tetrandrine as a potential therapeutic agent for COVID-19. Pharmacol. Res. Perspect. 2020, 8, e00653. [Google Scholar] [CrossRef]
  261. Mamontov, E.; Cheng, Y.; Daemen, L.L.; Kolesnikov, A.I.; Ramirez-Cuesta, A.J.; Ryder, M.R.; Stone, M.B. Low rotational barriers for the most dynamically active methyl groups in the proposed antiviral drugs for treatment of SARS-CoV-2, apilimod and tetrandrine. Chem. Phys. Lett. 2021, 777, 138727. [Google Scholar] [CrossRef]
  262. Lv, Y.L.; Wu, Z.Z.; Chen, L.X.; Wu, B.X.; Chen, L.L.; Qin, G.C.; Gui, B.; Zhou, J.Y. Neuroprotective effects of tetrandrine against vascular dementia. Neural. Regen. Res. 2016, 11, 454–459. [Google Scholar] [CrossRef] [PubMed]
  263. He, F.Q.; Qiu, B.Y.; Zhang, X.H.; Li, T.K.; Xie, Q.; Cui, D.J.; Huang, X.L.; Gan, H.T. Tetrandrine attenuates spatial memory impairment and hippocampal neuroinflammation via inhibiting NF-kappaB activation in a rat model of Alzheimer’s disease induced by amyloid-beta(1-42). Brain Res. 2011, 1384, 89–96. [Google Scholar] [CrossRef]
  264. Ma, H.; Yao, L.; Pang, L.; Li, X.; Yao, Q. Tetrandrine ameliorates sevoflurane-induced cognitive impairment via the suppression of inflammation and apoptosis in aged rats. Mol. Med. Rep. 2016, 13, 4814–4820. [Google Scholar] [CrossRef]
  265. Henan-Provincial-People’s-Hospital. Tetrandrine Tablets used in the Treatment of COVID-19 (TT-NPC). ClinicalTrials.gov (ID:NCT04308317). Available online: https://classic.clinicaltrials.gov/ct2/show/NCT04308317 (accessed on 30 August 2023).
  266. Luo, P.; Liu, D.; Li, J. Pharmacological perspective: Glycyrrhizin may be an efficacious therapeutic agent for COVID-19. Int. J. Antimicrob. Agents 2020, 55, 105995. [Google Scholar] [CrossRef]
  267. Patil, R.; Chikhale, R.; Khanal, P.; Gurav, N.; Ayyanar, M.; Sinha, S.; Prasad, S.; Dey, Y.N.; Wanjari, M.; Gurav, S.S. Computational and network pharmacology analysis of bioflavonoids as possible natural antiviral compounds in COVID-19. Inf. Med. Unlocked 2021, 22, 100504. [Google Scholar] [CrossRef]
  268. van de Sand, L.; Bormann, M.; Alt, M.; Schipper, L.; Heilingloh, C.S.; Steinmann, E.; Todt, D.; Dittmer, U.; Elsner, C.; Witzke, O.; et al. Glycyrrhizin effectively inhibits SARS-CoV-2 replication by inhibiting the viral main protease. Viruses 2021, 13, 609. [Google Scholar] [CrossRef]
  269. Kim, S.W.; Lim, C.M.; Lee, H.K.; Lee, J.K. The use of Stronger Neo-Minophagen C, a glycyrrhizin-containing preparation, in robust neuroprotection in the postischemic brain. Anat. Cell Biol. 2011, 44, 304–313. [Google Scholar] [CrossRef]
  270. Egyptian-Biomedical-Research-Network. Complementary Intervention for COVID-19. ClinicalTrials.gov (ID:NCT04487964). Available online: https://classic.clinicaltrials.gov/ct2/show/NCT04487964?term=Glycyrrhizin&cond=COVID-19&draw=2&rank=1 (accessed on 30 August 2023).
  271. Fuzo, C.A.; Martins, R.B.; Fraga-Silva, T.F.C.; Amstalden, M.K.; Canassa De Leo, T.; Souza, J.P.; Lima, T.M.; Faccioli, L.H.; Okamoto, D.N.; Juliano, M.A.; et al. Celastrol: A lead compound that inhibits SARS-CoV-2 replication, the activity of viral and human cysteine proteases, and virus-induced IL-6 secretion. Drug Dev. Res. 2022, 83, 1623–1640. [Google Scholar] [CrossRef]
  272. Zhang, B.; Zhong, Q.; Chen, X.; Wu, X.; Sha, R.; Song, G.; Zhang, C.; Chen, X. Neuroprotective effects of celastrol on transient global cerebral ischemia rats via regulating HMGB1/NF-kappaB signaling pathway. Front. Neurosci. 2020, 14, 847. [Google Scholar] [CrossRef] [PubMed]
  273. Liu, D.D.; Luo, P.; Gu, L.; Zhang, Q.; Gao, P.; Zhu, Y.; Chen, X.; Guo, Q.; Zhang, J.; Ma, N.; et al. Celastrol exerts a neuroprotective effect by directly binding to HMGB1 protein in cerebral ischemia-reperfusion. J. Neuroinflam. 2021, 18, 174. [Google Scholar] [CrossRef]
  274. Schiavone, S.; Morgese, M.G.; Tucci, P.; Trabace, L. The therapeutic potential of celastrol in central nervous system disorders: Highlights from In vitro and In vivo approaches. Molecules 2021, 26, 4700. [Google Scholar] [CrossRef]
  275. Yang, H.N.; Zhang, A.; Zhang, Y.Q.; Ma, S.; Wang, C.L. Resveratrol pretreatment protected against cerebral ischemia/reperfusion injury in rats via expansion of T regulatory cells. J. Stroke. Cerebrovasc. Dis. 2016, 25, 1914–1921. [Google Scholar] [CrossRef]
  276. Wei, H.; Wang, S.; Zhen, L.; Yang, Q.; Wu, Z.; Lei, X.; Lv, J.; Xiong, L.; Xue, R. Resveratrol attenuates the blood-brain barrier dysfunction by regulation of the MMP-9/TIMP-1 balance after cerebral ischemia reperfusion in rats. J. Mol. Neurosci. 2015, 55, 872–879. [Google Scholar] [CrossRef]
  277. Khoury, N.; Xu, J.; Stegelmann, S.D.; Jackson, C.W.; Koronowski, K.B.; Dave, K.R.; Young, J.I.; Perez-Pinzon, M.A. Resveratrol preconditioning induces genomic and metabolic adaptations within the long-term window of cerebral ischemic tolerance leading to bioenergetic efficiency. Mol. Neurobiol. 2019, 56, 4549–4565. [Google Scholar] [CrossRef]
  278. Marin-Palma, D.; Tabares-Guevara, J.H.; Zapata-Cardona, M.I.; Florez-Alvarez, L.; Yepes, L.M.; Rugeles, M.T.; Zapata-Builes, W.; Hernandez, J.C.; Taborda, N.A. Curcumin inhibits in Vitro SARS-CoV-2 infection in Vero E6 cells through multiple antiviral mechanisms. Molecules 2021, 26, 6900. [Google Scholar] [CrossRef]
  279. Cole, G.M.; Teter, B.; Frautschy, S.A. Neuroprotective effects of curcumin. Adv. Exp. Med. Biol. 2007, 595, 197–212. [Google Scholar] [CrossRef]
  280. Sarkar, S.; Karmakar, S.; Basu, M.; Ghosh, P.; Ghosh, M.K. Neurological damages in COVID-19 patients: Mechanisms and preventive interventions. Med. Comm. 2023, 4, e247. [Google Scholar] [CrossRef] [PubMed]
  281. Sohn, S.I.; Priya, A.; Balasubramaniam, B.; Muthuramalingam, P.; Sivasankar, C.; Selvaraj, A.; Valliammai, A.; Jothi, R.; Pandian, S. Biomedical Applications and bioavailability of curcumin—An updated overview. Pharmaceutics 2021, 13, 2102. [Google Scholar] [CrossRef] [PubMed]
  282. Shojaei, M.; Foshati, S.; Abdi, M.; Askari, G.; Sukhorukov, V.N.; Bagherniya, M.; Sahebkar, A. The effectiveness of nano-curcumin on patients with COVID-19: A systematic review of clinical trials. Phytother. Res. 2023, 37, 1663–1677. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Neurological symptoms caused by SARS-CoV-2 infection and their reported frequencies in COVID-19 patients. Created with Biorender.Com.
Figure 1. Neurological symptoms caused by SARS-CoV-2 infection and their reported frequencies in COVID-19 patients. Created with Biorender.Com.
Biomolecules 13 01585 g001
Figure 2. COVID-19 and neurodegenerative diseases share common mechanisms of neuronal damage. RAAS: Renin-angiotensin-aldosterone system. Created with BioRender.com.
Figure 2. COVID-19 and neurodegenerative diseases share common mechanisms of neuronal damage. RAAS: Renin-angiotensin-aldosterone system. Created with BioRender.com.
Biomolecules 13 01585 g002
Figure 3. Structure of neuroprotective compounds and comparison with SARS-CoV-2 Mpro inhibitors. (AC) Chemical structure of the flavonoids (A), alkaloids (B) and terpenoids (C) listed in Table 1. (D) Ribbon representation of SARS-CoV-2 Main protease (Mpro) crystal structures with the flavonoids quercetin, baicalein, or myricetin bound to the catalytic site (insets). (E) Structural comparison by extended connectivity fingerprints of compounds from Table 1 and reported Mpro inhibitors. Compounds in blue are natural products with an Mpro IC50 < 10 μM [160], whereas compounds in green are Mpro inhibitors that have reached clinical use.
Figure 3. Structure of neuroprotective compounds and comparison with SARS-CoV-2 Mpro inhibitors. (AC) Chemical structure of the flavonoids (A), alkaloids (B) and terpenoids (C) listed in Table 1. (D) Ribbon representation of SARS-CoV-2 Main protease (Mpro) crystal structures with the flavonoids quercetin, baicalein, or myricetin bound to the catalytic site (insets). (E) Structural comparison by extended connectivity fingerprints of compounds from Table 1 and reported Mpro inhibitors. Compounds in blue are natural products with an Mpro IC50 < 10 μM [160], whereas compounds in green are Mpro inhibitors that have reached clinical use.
Biomolecules 13 01585 g003
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

Zaa, C.A.; Espitia, C.; Reyes-Barrera, K.L.; An, Z.; Velasco-Velázquez, M.A. Neuroprotective Agents with Therapeutic Potential for COVID-19. Biomolecules 2023, 13, 1585. https://doi.org/10.3390/biom13111585

AMA Style

Zaa CA, Espitia C, Reyes-Barrera KL, An Z, Velasco-Velázquez MA. Neuroprotective Agents with Therapeutic Potential for COVID-19. Biomolecules. 2023; 13(11):1585. https://doi.org/10.3390/biom13111585

Chicago/Turabian Style

Zaa, César A., Clara Espitia, Karen L. Reyes-Barrera, Zhiqiang An, and Marco A. Velasco-Velázquez. 2023. "Neuroprotective Agents with Therapeutic Potential for COVID-19" Biomolecules 13, no. 11: 1585. https://doi.org/10.3390/biom13111585

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