Next Article in Journal
The Generation and Characterization of Monoclonal Antibodies against the MPXV A29L Protein
Previous Article in Journal
Special Issue “Arbovirus Diagnostics”
Previous Article in Special Issue
Suspected Human-to-Cat Spillover of SARS-CoV-2 Omicron Variant in South Korea
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Neurological Complications of COVID-19: Unraveling the Pathophysiological Underpinnings and Therapeutic Implications

1
Department of Pathology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
2
Lincoln Memorial DeBusk College of Osteopathic Medicine, Lincoln Memorial University, Knoxville, TN 37902, USA
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Viruses 2024, 16(8), 1183; https://doi.org/10.3390/v16081183
Submission received: 11 June 2024 / Revised: 11 July 2024 / Accepted: 23 July 2024 / Published: 24 July 2024
(This article belongs to the Special Issue Molecular Epidemiology of SARS-CoV-2, 3rd Edition)

Abstract

:
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease (COVID-19), induced a global pandemic with a diverse array of clinical manifestations. While the acute phase of the pandemic may be waning, the intricacies of COVID-19′s impact on neurological health remain a crucial area of investigation. Early recognition of the spectrum of COVID-19 symptoms, ranging from mild fever and cough to life-threatening respiratory distress and multi-organ failure, underscored the significance of neurological complications, including anosmia, seizures, stroke, disorientation, encephalopathy, and paralysis. Notably, patients requiring intensive care unit (ICU) admission due to neurological challenges or due to them exhibiting neurological abnormalities in the ICU have shown increased mortality rates. COVID-19 can lead to a range of neurological complications such as anosmia, stroke, paralysis, cranial nerve deficits, encephalopathy, delirium, meningitis, seizures, etc., in affected patients. This review elucidates the burgeoning landscape of neurological sequelae associated with SARS-CoV-2 infection and explores the underlying neurobiological mechanisms driving these diverse manifestations. A meticulous examination of potential neuroinvasion routes by SARS-CoV-2 underscores the intricate interplay between the virus and the nervous system. Moreover, we dissect the diverse neurological manifestations emphasizing the necessity of a multifaceted approach to understanding the disease’s neurological footprint. In addition to elucidating the pathophysiological underpinnings, this review surveys current therapeutic modalities and delineates prospective avenues for neuro-COVID research. By integrating epidemiological, clinical, and diagnostic parameters, we endeavor to foster a comprehensive analysis of the nexus between COVID-19 and neurological health, thereby laying the groundwork for targeted therapeutic interventions and long-term management strategies.

1. Introduction

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a novel beta coronavirus that causes a variety of symptoms collectively referred to as coronavirus disease (COVID-19) [1]. Though the devastating COVID-19 pandemic as we experienced it seems to be behind us, much needs to be comprehended about clinical manifestations of COVID-19 infection [2,3]. Early on in the pandemic, it became increasingly clear that clinical manifestations of SARS-CoV-2 infection could range from minor symptoms like fever, cough, and shortness of breath to serious life-threatening respiratory distress and multi-organ failure requiring hospitalization and intensive care unit (ICU) admission.
Frequently reported among these symptoms were neurological problems such as anosmia, seizures, stroke, disorientation, encephalopathy, and total paralysis [4,5]. Of specific concern is the increased likelihood of mortality observed in COVID-19 patients who require ICU admission secondary to neurological challenges, as well as COVID-19 patients in ICU with neurological abnormalities [5,6]. Additionally, the patients who leave the ICU and recover from their infection are at an increased risk of developing long-term neuropsychiatric and neurocognitive problems such as depression, post-traumatic stress disorder, psychosis, anxiety, and memory impairment [7,8]. The neurological manifestations observed in COVID-19 patients may be due to the direct SARS-CoV-2 mediated injury to the central nervous system or the sequelae of inflammatory cytokines in the storm triggered by COVID-19 infection or secondary to therapeutic interventions. It is also likely that a pre-existing neurological condition may become unmasked or worsen [9].
As we transition into the post-pandemic phase of COVID-19, it is crucial to recognize that we identify the neurological complications associated with the virus and gain a comprehensive understanding of its underlying mechanisms. This knowledge will serve as the basis for effectively managing patients clinically and creating specific therapeutic interventions for acute neurological conditions. In this review, we have focused on the neurological manifestations of COVID-19 diseases, potential pathogenic processes, and biomarkers relevant to the diagnosis of neuro-covid. Finally, we have attempted to make a better understanding of this important component of COVID-19 disease, which will hopefully provide some direction in charting the course to more effective management with regard to its nervous system involvement.

2. Neuroinvasion of SARS-CoV-2 via Potential Brain Routes

One of the potential mechanisms of SARS-CoV-2 entry into the nervous system could be direct entry via angiotensin-converting enzyme 2 (ACE2), an established receptor for entry of SARS-CoV-2. ACE2 has been found in human brain vessels, an observation that has since been attributed to its expression in vascular wall pericytes and smooth muscle cells but not in the endothelium lining cerebral capillaries [10]. Although the number of positive neurons was limited, data mining of human brain single-nuclear RNA sequencing (RNA-seq) data revealed expression in the choroid plexus and neocortical neurons (~2% or less) [11]. Also, lower expression of ACE2 was observed in the microglia, endothelial cells, or pericytes in the human brain as compared to the mouse brain. CD147, a surface molecule that is widely expressed in epithelial, neuronal, myeloid, and lymphoid cells, was identified as a SARS-CoV-2 receptor, hence increasing the likelihood of infection in multiple organs including the brain [12]. Cathepsin L activates SARS-CoV-2 Spike in endosomes and can compensate for viral entry into cells that lack TMPRSS2. Neuropilin (NRP1; transmembrane receptors that function as mediators of neuronal guidance and angiogenesis) can also mediate the transport of virus-sized particles from the intranasal region into the brain in a murine study [13]. The findings imply that vascular wall cells in the human brain may express ACE2 at low levels but other non-canonical SARS-CoV-2 receptors such as Cathepsin L and NRP1, which are present in various brain cells, can make them sensitive to the virus.
Additionally, transmission electron microscopy samples from the frontal lobes of COVID-19 patients revealed the presence of 80–110 nm viral particles with beta coronavirus features [14]. Similarly, coronavirus particles have been found in the neurons of prior viral epidemic victims [15,16]. SARS-CoV-2 may have significant neuroinvasive potential based on the expression of ACE2 in neurons, endothelial cells, and astrocytes [17,18]. Indeed, human brain organoid investigations and in vivo studies in mice overexpressing human ACE2 demonstrated that SARS-CoV-2 can infect neuronal cells [19,20,21]. However, SARS-CoV-2 RNA identification in the brain and cerebrospinal fluid (CSF) yielded inconsistent results [22,23]. As seen in monkeys infected with coronavirus, the endothelium can become infected and serve as an entry site into the brain parenchyma [24]. Investigating how the virus could infiltrate the nervous system could aid in determining the infection’s virulence and the possibility of direct invasion. Several putative entry pathways for SARS-CoV-2 have been hypothesized below based on investigations with previous coronaviruses.

2.1. Blood–Brain Barrier

Crossing the BBB (blood–brain barrier) is a typical pathway for blood-borne viruses to enter the brain. The virus was found to spread into the circulation in COVID-19 patients, though at varying rates (1% to 41%), and may enter the brain by crossing the BBB [25,26]. Internalization and transport of the virus through the cerebral endothelium, where the expression of SARS-CoV-2 docking proteins is unknown, would be required to cross the intact BBB. Immunoreactivity to ACE2 was found in the brain arteries of a patient who died from repeated ischemic infarctions but the precise cellular location remains unknown [27]. Entry via additional putative SARS-CoV-2 receptors, such as NRP1 and BSG, which are more broadly expressed in the cerebral vasculature, cannot be ruled out. SARS-CoV-2-associated cytokines, such as interleukin (IL)-6, IL-1b, tumor necrosis factor (TNF), and IL-17, damage the BBB, perhaps allowing the virus to enter [28]. SARS-CoV-2 has been suggested to cause endothelial infection and inflammation in peripheral vessels but no direct evidence has been found in brain endothelial cells. Rather, multiple autopsy studies have revealed a lack of florid cerebrovascular inflammation [29,30,31,32]. COVID-19 co-morbidities, such as cardiovascular risk factors or pre-existing neurological disorders, may increase BBB permeability alone or in combination with cytokines. For example, electron imaging detected viral particles in frontal lobe microvessels and neurons in a COVID-19 patient with Parkinson’s disease, implying trans-endothelial entry [33]. SARS-CoV-2 might potentially enter the brain through the hypothalamic median eminence and other circumventricular organs, as well as other brain areas that have a permeable BBB due to fenestrated capillary walls. Although the viral particle is bigger than endothelial fenestrae, preliminary results show that capillaries in the median eminence express ACE2 and TMPRSS, which might facilitate virus passage into the hypothalamus (Figure 1a) [34], which, under its extensive connections, might act as a portal to the whole brain.

2.2. Immune Deregulation

Viruses can reach the brain via infected immune cells, which can serve as their reservoir [35]. The vasculature, meninges, and choroid plexus all carry monocytes, neutrophils, and T cells into the brain and these locations might represent entry routes for infected immune cells [36,37]. Until recently, there has been no conclusive proof of the SARS-CoV-2 infection of immune cells. Single-cell RNA seq data revealed viral RNA in macrophages in bronchoalveolar lavage of COVID-19 patients, whereas SARS-CoV-2 nucleocapsid protein (NP) immunoreactivity was detected in CD68+ cells in lymphoid organs [38,39]. It is unclear as to whether this is due to virus replication in macrophages or if it is due to the phagocytic uptake of virus-infected cells or extracellular virions. A notable absence of immune cell infiltration has also been documented in numerous autopsy series [40,41,42].

2.3. Retrograde Axonal Transport

For viral transmission along the axon into the perikaryon, retrograde transport machinery can be used (Figure 1b) [43]. Colchicine and vinblastine disrupt microtubules, which impair neural transport and inhibit the reproduction of the murine hepatitis virus (MHV) [44]. Several respiratory viruses, including H5N1, have been found to move from the respiratory system to the brain via the vagus nerve in animal studies [45]. The perikaryon of vagal sensory neurons is located in the solitary nucleus of the medulla, linked to the medulla’s respiratory center and the pons. If the infection reaches the solitary nucleus, the respiratory center may be at risk, leading to respiratory failure. In addition to retrograde transport, SARS-CoV-2, like other coronaviruses, may use the axonal endoplasmic reticulum of infected neurons to spread to the brain [46].

2.4. Via Gastrointestinal Tract

The virus may be carried from the gastrointestinal tract into the brain because the former is lined by ACE2-expressing cells, contains numerous neurons, and is widely innervated by the vagus nerve [47]. ACE2 cleaves angiotensin (Ang) into Ang (1–7) that binds to Mas receptors and activates anti-inflammatory and anti-fibrosis pathways in the renin–angiotensin system (RAS) in vivo [48,49]. SARS-CoV-2 activates the host immune system by interacting with resident lymphocytes in the intestinal epithelium and lamina propria. The phosphorylation of p38 mitogen kinase and nuclear factor-kB pathways occurs as a result of this activation and the imbalance of RAS-ACE2, resulting in the production of inflammatory factors such as tumor necrosis factor-a, interleukin-1, interleukin-6, and interleukin-8 [50]. As a result, the shedding and death of intestinal epithelial cells is stimulated, increasing the intestinal mucosa’s permeability. On the other hand, the BBB’s tight junction proteins may be damaged, allowing SARS-CoV-2 to enter the brain and cause lesions in the central nervous system in COVID-19 patients. Furthermore, SARS-CoV-2 RNA has been found in rectal swabs and feces samples and it stays detectable for a longer period, even when PCR findings from nasopharyngeal swab testing are negative [51].

3. SARS-CoV-2 Variants and Their Neurological Manifestations

Throughout the COVID-19 pandemic, the genome of SARS-CoV-2 has undergone evolutionary changes, giving rise to new variants that have spread globally. By the end of 2020, three prominent variants had emerged: Alpha (B.1.1.7), Beta (B.1.351), and Gamma (P.1). Subsequently, in May 2021, the Delta variant (B.1.617.2) emerged, followed by the Omicron variant in October (B.1.1.529) [52]. These variants differ in their ability to spread, evade immunity, and affect the severity of COVID-19. Importantly, various studies have reported differences in the incidence of neurological disorders associated with these different variants. For instance, initial research indicated that 8.4% of patients likely infected with the original Wuhan-Hu-1 strain developed neurological issues. This proportion decreased to 5.0% during the prevalence of the Alpha variant and further declined to 3.3% during the Delta variant surge [53]. Similarly, animal studies, such as the one conducted with hamsters, have shown that the Wuhan strain induces anosmia more frequently than the Gamma variant, with no instances of anosmia observed in hamsters infected with the Delta or Omicron variants [54].
In a separate investigation conducted by Taquet et al., an extensive cohort analysis scrutinized neurological and psychiatric conditions among patients afflicted with COVID-19 before the onset of the Alpha variant, alongside those infected with Delta, or Omicron variants. The results notably demonstrated an elevated susceptibility among Delta-infected individuals to develop ischemic stroke, epilepsy, seizures, and cognitive impairments [55]. Distinct clinical manifestations correlate with differential susceptibility of nervous system cells. Recently, Proust et al. published a study comparing the infectivity and replication efficiency of the Wuhan strain, Alpha, Beta, Delta, Eta, and Omicron (BA.1) within blood–brain barrier (BBB) cells [56]. Their findings revealed varying susceptibilities, with Alpha, Beta, and Omicron showing productive infection in astrocytes; Wuhan, Beta, and Omicron in epithelial cells; Omicron exclusively in pericytes; and Wuhan, Delta, and Omicron variants in microglial cells.
Furthermore, the delta variant (B.1.617.2) of SARS-CoV-2 exhibited the highest risk among COVID-19 variants for neuropsychiatric complications and mortality. Specifically, there was a notable increase in cognitive deficits, epilepsy, and ischemic stroke incidences compounded by an elevated mortality risk [57]. During the periods dominated by the alpha variant (B.1.1.7) and the omicron variant (B.1.1.529), the neurological and psychiatric complication profiles closely mirrored those observed during the delta variant pandemic phase. There were no significant differences in the risks of cognitive deficits, epilepsy, ischemic stroke, psychotic disorders, or mood disorders among these variants. Despite similar rates of neurological complications, the omicron variant showed a reduced composite risk of hospitalization and death, likely due to a less severe course of SARS-CoV-2 infection [55,57].
Additionally, COVID-19 patients have had a wide range of neurological symptoms (Table 1). In mild cases, hyposmia and hypogeusia are seen, while severe cases of COVID-19 show agitation, diffuse corticospinal tract symptoms, and myoclonus [58,59,60]. Female patients are more affected than male ones but the severity is much less than in males [61,62]. Other data indicated that neurological symptoms such as a loss of smell and taste occurred in 19% of individuals and some reported ataxia, seizures, vision impairment, and other symptoms as low as 5% [63,64]. Because the symptoms are linked to a milder clinical course or cases without respiratory symptoms, the source of the disparity might be related to anamnesis techniques and a variation in the degree of illness severity [65]. An objective investigation employing a scent-identification (40 odorants) test indicated that 98 percent of individuals with COVID-19 had impaired smell function. In total, 58 percent of them were anosmic or severely microsmic [66]. In comparison to the real number of COVID-19 cases, the number of reported participants in such trials is still rather modest and additional data are needed. Nonetheless, anosmia and ageusia have been added to the list of symptoms and risk factors linked with COVID-19, according to the most recent COVID-19 clinical treatment guidelines [67]. Symptoms of muscle involvement, such as fatigue and limb pain or soreness are also linked. Dizziness and headache, decreased awareness, seizures, psychiatric symptoms, diffuse corticospinal tract indications, ataxia, and acute cerebrovascular events, such as ischemic stroke and cerebral hemorrhage, have all been reported as CNS involvement [68,69].
Also, there is evidence that viral proteins may survive in people for months and can affect various systems even if they do not infect cells. The SARS-CoV-2 N protein was identified in intestinal enterocytes in 5 of 14 people after an average of 4 months (range of 2.8 to 5.7 months) post-COVID-19 diagnosis, which may imply its participation in extended COVID [70]. Various neurological disorders have been reported to occur weeks, months, or even years after acute viral infection in survivors of previous pandemics. Between three and four weeks after SARS-CoV and MERS-CoV infection, neurological problems such as limb weakness, hyporeflexia, paresthesia, and hypesthesia were described [71,72]. Indeed, mice who survived HCoV-induced acute encephalitis had lower locomotor activity, a smaller hippocampus, and neuronal loss in the CA1 and CA3 layers, all of which might lead to learning and memory deficits [73]. Immune responses to viral infection have been linked to the onset of neurological autoimmune disorders such as multiple sclerosis (MS) [74]. The causes, which include persistent inflammation caused by prolonged IFN production, have recently been explored [75]. Furthermore, HCoV-OC43 was found in the brain tissues of 48 percent of MS patients [76]. The demyelinating effect of HCoV-229E in MS patients has been confirmed by investigating mouse hepatitis virus (MHV)-infected mice [77]. More research on how the immune system responds to viral infection and how this relates to the prevention of virus-related autoimmune disorders is essential. The involvement of the gastrointestinal tract in SARS-CoV-2 infection poses an additional hazard to the neurological system, since it may enhance the induction and transit of abnormal proteins such as α-synuclein, which is linked to neurodegenerative disorders [78]. Inflammation-induced bacterial leakage from the gastrointestinal system may transport bacteria and metabolites into the brain parenchyma, causing CNS disease [79].
At the molecular level, severe COVID-19 infection can induce epigenetic changes in the frontal cortex that resemble those of natural aging, such as DNA methylation and histone modifications [80]. COVID-19 infection can also trigger synapse elimination in microglia cells and upregulate cytokines. The single cell profiling of SAR-SoV-2-infected patients has observed upregulation of synapse engulfment and migration along with other immune-related genes such as TNF-α and IL-6. The disruption of normal brain circuitry can promote a cognitive decline in patients after recovery from COVID and stress-related responses such as mood alterations [81]. Furthermore, SARS-CoV-2 can affect normal mitochondrial physiology, which is essential for maintaining cellular energy reserve and homeostasis. Mitochondrial dysfunctionality contributes to the buildup of oxidative stress, inflammation, impaired synaptic plasticity, and inflammation. Mitochondrial malfunction is linked to several degenerative disorders, including Alzheimer’s, Parkinson’s, and Huntington’s. Furthermore, aging is also associated with mitochondrial anomalies and the dysregulation of cellular bioenergetics [82]. Furthermore, mitochondria are hijacked by SARS-CoV-2 of the immune cells and utilize these as replicative niches [83]. Additional factors such as diabetes, cardiovascular diseases, and age-associated vulnerabilities further complicate clinical etiological diseases [84,85].
Also, cytokines have been shown to play a critical role in mood disorders. For example, the upregulation of TNF-α and IL-6, two major cytokines in COVID-19, directly affects brain physiology and may negatively promote stress-related responses such as mood alterations [86].
Table 1. Neurological symptoms of COVID-19 according to the patients’ pathophysiological background.
Table 1. Neurological symptoms of COVID-19 according to the patients’ pathophysiological background.
S. No.Patient SizeSample TypeType of StudyStudy DesignNeurological ManifestationAltered Markers Potential Associated VariantAssay ProcedureRef.
111 COVID-19 patientsSerum and CSFClinicalRetrospectiveMyoclonus, oculomotor disturbance, delirium, dystonia, and epileptic seizuresNeurofilament light chain (NfL) levels in CSF were increased in all tested patients.
one patient showed Yo antibodies in serum and CSF and two patients myelin antibodies in serum. One patient had high-level serum IgG NMDA receptor antibodies
SARS-CoV-2 Beta Indirect immunofluorescence assay[87]
218 COVID-19 subjects, 14 healthy controls, and 68 non-COVID-19 neurological disease controlsCSFClinicalRetrospectiveStroke, encephalopathy, and headacheAnti-SARS-CoV2 antibodies were found in 77% of COVID-19 subjects’ CSF. Increase in pro-inflammatory cytokines (IL-6, TNFα, and IL-12p70) and IL-10 in the CSF of COVID-19 patients. CSF-hsCRP was present exclusively in COVID-19 casesSARS-CoV-2 BetaRT-PCR and ELISA[88]
335 COVID-19 patientsSerum/CSFClinicalRetrospectiveEncephalitis and strokeCSF tumor necrosis factor-alpha (TNFα) and IL6 levels were higher in patients presenting pronounced neuroimaging alterations compared to those who did not SARS-CoV-2 BetaCytokines analysis using magnetic beads, ELISA, and LC-MS/MS[89]
4844 COVID-19 patientsBloodClinicalRetrospectiveStroke and intracranial hemorrhageAnticardiolipin antibodies were observed in all the patientsSARS-CoV-2 BetaBiochemical analysis[90]
512 COVID-19 patients and 19 healthy controlsPlasmaClinicalLongitudinal analysis/ProspectiveDifficulty with memory or concentration, increased anxiety, and depressionIL-1β was significantly increased in COVID patients, both brain-derived neurotrophic factor and cortisol were significantly elevated in COVID patients SARS-CoV-2 Epsilon, Alpha, Delta, and OmicronGenotyping analysis, cytokine analysis, and ELISA[91]
6400 COVID-19 patientsPlasmaClinical/AutopsyRetrospectiveischemic stroke, dementia, epilepsy, and hemorrhagic strokeLDH, ferritin, hsTnI, and IL-6 were found to be higher in deceased patients. A concentration of hsTnI > 64 ng/L appeared to constitute a strong predictor of an unfavorable prognosisSARS-CoV-2 Epsilon, Alpha, and DeltaRT-PCR, MR, CT, Doppler US imaging, and biochemical analysis[92]
772 COVID-19 PatientsBloodClinicalProspectiveFatigue, anxiety, and depressionElevated levels of TNF-α and IL-1β were associated with decreased episodic memory, working memory, and inhibitory controlSARS-CoV-2 Beta and DeltaBiochemical analysis[93]

4. Therapeutic Strategies

At this time, there is no specific proven therapeutic regimen for neurological complications associated with COVID-19, regardless of the underlying mechanism. While acute stroke therapies and seizures have not been specifically tested in the COVID-19 population, there are no data to suggest that the risk–benefit ratio of these interventions differs for patients with COVID-19. Therefore, the management of ischemic and hemorrhagic stroke in COVID-19 should follow the same standards of care as for patients without COVID-19 (Table 2). This includes a timely evaluation of candidacy for acute medical and interventional stroke therapies, such as thrombolysis or thrombectomy in acute ischemic stroke [94,95]. For encephalopathy, reported options include monotherapy or combinations of corticosteroids (for example, methylprednisolone 1 g daily for 5–10 days), intravenous immunoglobulin, plasma exchange, and rituximab [96]. Furthermore, in GBS, it should only be considered for COVID-19 if respiratory insufficiency seems disproportionate to pulmonary findings. Also, with other causes of GBS, both intravenous immunoglobulin and plasma exchange are reported therapies that could be explored in COVID-19 [97].
The autonomic nervous system’s immunomodulatory actions may be useful in preventing the hyper-inflammatory state that leads to severe COVID-19 symptoms. Maintaining a small but efficient immune response may allow infections to be eliminated while minimizing tissue damage, including CNS involvement. Some neurons, such as those in the olfactory bulb of mice, can withstand coronavirus infection. Differential gene expression in those neurons may yield useful information that may be used to develop neuroprotection methods for acute neurotropic virus infection. IL-10 therapy in MHV-infected mice has been shown to cause astrocytes to produce glial scars that restrict demyelination regions [98]. Remyelination may occur after myelin breakdown, as evidenced by overexpression of genes involved in oligodendrocyte maturation, such as Oncostatin M, in mice infected with MHV [99]. It is still unclear if the degree of remyelination has an impact on the disease’s prognosis. The inhibition of CD147, one of the SARS-CoV-2 receptors, has been shown in mouse models of ischemic stroke to offer further benefits in preserving oligodendrocytes and white matter [100]. Antibodies against CD147, on the other hand, permeabilize BBB by binding to CD147 in brain endothelial cells, therefore increasing brain inflammation. This double-edged sword impact might thus be beneficial or harmful. Previous research has revealed the possibility of neurological problems associated with immunization. Following vaccination for influenza, polio, rabies, meningococcus, measles, mumps, and tetanus, acute inflammatory demyelinating polyneuropathy (AIDP) is the most frequent neurologic condition, as documented in case reports [101]. Despite these limitations, the currently available data show extremely modest rates of neurologic sequelae following immunization, especially when compared to the severity of infection-related morbidity and death in populations of individuals with neurological disease.

5. Future Perspective

The field of neuro-COVID research is still in its early stages and much is yet to be learned about the long-term effects of COVID-19 on the brain and nervous system. However, several potential future perspectives could emerge from the ongoing research.
  • Understanding the mechanisms of COVID-19’s impact on the brain: More studies should be performed to enhance the understanding of the mechanisms by which the SARS-CoV-2 impacts the neuro system. As more time passes since the beginning of the pandemic, researchers will be able to study the long-term effects of COVID-19 on the brain and nervous system. This could provide important insights into the potential long-term consequences of COVID-19, such as increased risk of dementia or other cognitive impairments;
  • Development of new diagnostic tools: Neuro-COVID research could lead to the development of new diagnostic tools to detect neurological complications of COVID-19. For example, the use of MRI and other imaging techniques to detect brain changes in COVID-19 patients should be explored;
  • Treatment and management of neurological symptoms: Finally, research into the neurological effects of COVID-19 could lead to new treatments and management strategies for patients experiencing these symptoms. A study published in the Journal of Neuroimmunology in 2021 found that a combination of two monoclonal antibodies, casirivimab and imdevimab, improved neurological symptoms in hospitalized COVID-19 patients [102]. Another study published in the Journal of Clinical Investigation in 2021 found that a monoclonal antibody called CT-P59 was effective in reducing COVID-19 symptoms, including neurological symptoms, in non-hospitalized patients [103]. Also, there is a growing interest in non-pharmacological interventions for the treatment of NeuroCOVID. These interventions may include therapies such as physical rehabilitation, cognitive–behavioral therapy, and neuropsychological assessments. A study published in the Journal of Neurologic Physical Therapy in 2021 found that physical therapy was effective in improving balance and gait in COVID-19 patients with neurological complications [104].
Overall, the future perspectives on the therapeutics of NeuroCOVID are promising, with a range of potential treatments and approaches being explored. As our understanding of the neurological complications of COVID-19 continues to evolve, new therapies and interventions will likely emerge to help those affected by this condition.

Author Contributions

Conceptualization, A.V. and R.K.; Formal analysis, A.V., V.V., H.S. and P.A.; Writing—original draft preparation, A.V., V.V., H.S. and P.A.; Writing—review and editing, A.K.M., C.W., J.F. and J.W.; Supervision, R.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Synowiec, A.; Szczepański, 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, e00133-20. [Google Scholar] [CrossRef] [PubMed]
  2. Chakraborty, C.; Bhattacharya, M.; Sharma, A.R. Present variants of concern and variants of interest of severe acute respiratory syndrome coronavirus 2: Their significant mutations in S-glycoprotein, infectivity, re-infectivity, immune escape and vaccines activity. Rev. Med. Virol. 2021, 32, e2270. [Google Scholar] [CrossRef]
  3. Scheepers, C.; Everatt, J.; Amoako, D.G.; Mnguni, A.; Ismail, A.; Mahlangu, B.; Wibmer, C.K.; Wilkinson, E.; Tegally, H.; San, J.E.; et al. The continuous evolution of SARS-CoV-2 in South Africa: A new lineage with rapid accumulation of mutations of concern and global detection. medRxiv 2021. [Google Scholar] [CrossRef]
  4. 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]
  5. Li, Z.; Liu, T.; Yang, N.; Han, D.; Mi, X.; Li, Y.; Liu, K.; Vuylsteke, A.; Xiang, H.; Guo, X. Neurological manifestations of patients with COVID-19: Potential routes of SARS-CoV-2 neuroinvasion from the periphery to the brain. Front. Med. 2020, 14, 1–9. [Google Scholar] [CrossRef] [PubMed]
  6. Chen, T.; Wu, D.I.; Chen, H.; Yan, W.; Yang, D.; Chen, G.; Ma, K.; Xu, D.; Yu, H.; Wang, H.; et al. Clinical characteristics of 113 deceased patients with coronavirus disease 2019: Retrospective study. BMJ 2020, 368, m1091. [Google Scholar] [CrossRef]
  7. Frontera, J.A.; Simon, N.M. Bridging Knowledge Gaps in the Diagnosis and Management of Neuropsychiatric Sequelae of COVID-19. JAMA Psychiatry 2022, 79, 811–817. [Google Scholar] [CrossRef] [PubMed]
  8. Rogers, J.P.; Chesney, E.; Oliver, D.; Pollak, T.A.; McGuire, P.; Fusar-Poli, P.; Zandi, M.S.; Lewis, G.; David, A.S. Psychiatric and neuropsychiatric presentations associated with severe coronavirus infections: A systematic review and meta-analysis with comparison to the COVID-19 pandemic. Lancet Psychiatry 2020, 7, 611–627. [Google Scholar] [CrossRef]
  9. Słyk, S.; Domitrz, I. Neurological manifestations of SARS-CoV-2—A systematic review. Neurol. I Neurochir. Pol. 2020, 54, 378–383. [Google Scholar] [CrossRef]
  10. He, L.; Mäe, M.A.; Sun, Y.; Muhl, L.; Nahar, K.; Liébanas, E.V.; Fagerlund, M.J.; Oldner, A.; Liu, J.; Genové, G.; et al. Pericyte-specific vascular expression of SARS-CoV-2 receptor ACE2–implications for microvascular inflammation and hypercoagulopathy in COVID-19 patients. bioRxiv 2020. [Google Scholar] [CrossRef]
  11. Chen, R.; Wang, K.; Yu, J.; Howard, D.; French, L.; Chen, Z.; Wen, C.; Xu, Z. The spatial and cell-type distribution of SARS-CoV-2 receptor ACE2 in the human and mouse brains. Front. Neurol. 2021, 11, 1860. [Google Scholar] [CrossRef] [PubMed]
  12. Wang, K.; Chen, W.; Zhou, Y.S.; Lian, J.Q.; Zhang, Z.; Du, P.; Gong, L.; Zhang, Y.; Cui, H.Y.; Geng, J.J.; et al. SARS-CoV-2 invades host cells via a novel route: CD147-spike protein. bioRxiv 2020. [Google Scholar] [CrossRef]
  13. Cantuti-Castelvetri, L.; Ojha, R.; Pedro, L.D.; Djannatian, M.; Franz, J.; Kuivanen, S.; Kallio, K.; Kaya, T.; Anastasina, M.; Smura, T.; et al. Neuropilin-1 facilitates SARS-CoV-2 cell entry and provides a possible pathway into the central nervous system. bioRxiv 2020. [Google Scholar] [CrossRef]
  14. Paniz-Mondolfi, A.; Bryce, C.; Grimes, Z.; Gordon, R.E.; Reidy, J.; Lednicky, J.; Sordillo, E.M.; Fowkes, M. Central nervous system involvement by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). J. Med. Virol. 2020, 92, 699–702. [Google Scholar] [CrossRef]
  15. Ding, Y.; He, L.I.; Zhang, Q.; Huang, Z.; Che, X.; Hou, J.; Wang, H.; Shen, H.; Qiu, L.; Li, Z.; et al. Organ distribution of severe acute respiratory syndrome (SARS) associated coronavirus (SARS-CoV) in SARS patients: Implications for pathogenesis and virus transmission pathways. J. Pathol. A J. Pathol. Soc. Great Br. Irel. 2004, 203, 622–630. [Google Scholar] [CrossRef] [PubMed]
  16. 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]
  17. Gowrisankar, Y.V.; Clark, M.A. Angiotensin II regulation of angiotensin-converting enzymes in spontaneously hypertensive rat primary astrocyte cultures. J. Neurochem. 2016, 138, 74–85. [Google Scholar] [CrossRef] [PubMed]
  18. Hamming, I.; Timens, W.; Bulthuis, M.L.C.; Lely, A.T.; Navis, G.V.; Van Goor, H. Tissue distribution of ACE2 protein, the functional receptor for SARS coronavirus. A first step in understanding SARS pathogenesis. J. Pathol. A J. Pathol. Soc. Great Br. Irel. 2004, 203, 631–637. [Google Scholar] [CrossRef] [PubMed]
  19. Mesci, P.; Macia, A.; Saleh, A.; Martin-Sancho, L.; Yin, X.; Snethlage, C.; Avansini, S.; Chanda, S.K.; Muotri, A. Sofosbuvir protects human brain organoids against SARS-CoV-2. bioRxiv 2020. [Google Scholar] [CrossRef]
  20. Ramani, A.; Müller, L.; Ostermann, P.N.; Gabriel, E.; Abida-Islam, P.; Müller-Schiffmann, A.; Mariappan, A.; Goureau, O.; Gruell, H.; Walker, A.; et al. SARS-CoV-2 targets neurons of 3D human brain organoids. EMBO J. 2020, 39, e106230. [Google Scholar] [CrossRef]
  21. Song, E.; Zhang, C.; Israelow, B.; Lu-Culligan, A.; Sprado, A.; Skriabine, S.; Lu, P.; Weizman, O.E.; Liu, F.; Dai, Y.; et al. Neuroinvasive potential of SARS-CoV-2 revealed in a human brain. J. Exp. Med. 2021, 218, e20202135. [Google Scholar] [CrossRef]
  22. 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] [PubMed]
  23. Schaller, T.; Hirschbühl, K.; Burkhardt, K.; Braun, G.; Trepel, M.; Märkl, B.; Claus, R. Postmortem examination of patients with COVID-19. JAMA 2020, 323, 2518–2520. [Google Scholar] [CrossRef] [PubMed]
  24. Cabirac, G.F.; Soike, K.F.; Butunoi, C.; Hoel, K.; Johnson, S.; Cai, G.Y.; Murray, R.S. Coronavirus JHM OMP1 pathogenesis in owl monkey CNS and coronavirus infection of owl monkey CNS via peripheral routes. In Coronaviruses; Springer: Boston, MA, USA, 1994; pp. 347–352. [Google Scholar]
  25. Wang, W.; Xu, Y.; Gao, R.; Lu, R.; Han, K.; Wu, G.; Tan, W. Detection of SARS-CoV-2 in different types of clinical specimens. JAMA 2020, 323, 1843–1844. [Google Scholar] [CrossRef]
  26. Zheng, S.; Fan, J.; Yu, F.; Feng, B.; Lou, B.; Zou, Q.; Xie, G.; Lin, S.; Wang, R.; Yang, X.; et al. Viral load dynamics and disease severity in patients infected with SARS-CoV-2 in Zhejiang province, China, January-March 2020: Retrospective cohort study. BMJ 2020, 369, m1443. [Google Scholar] [CrossRef] [PubMed]
  27. Bryce, C.; Grimes, Z.; Pujadas, E.; Ahuja, S.; Beasley, M.B.; Albrecht, R.; Hernandez, T.; Stock, A.; Zhao, Z.; Rasheed, M.A.; et al. Pathophysiology of SARS-CoV-2: Targeting of endothelial cells renders a complex disease with thrombotic microangiopathy and aberrant immune response. The Mount Sinai COVID-19 autopsy experience. medRxiv 2020. [Google Scholar] [CrossRef]
  28. Erickson, M.A.; Banks, W.A. Neuroimmune axes of the blood–brain barriers and blood–brain interfaces: Bases for physiological regulation, disease states, and pharmacological interventions. Pharmacol. Rev. 2018, 70, 278–314. [Google Scholar] [CrossRef]
  29. Teuwen, L.A.; Geldhof, V.; Pasut, A.; Carmeliet, P. COVID-19: The vasculature unleashed. Nat. Rev. Immunol. 2020, 20, 389–391. [Google Scholar] [CrossRef]
  30. Kantonen, J.; Mahzabin, S.; Mäyränpää, M.I.; Tynninen, O.; Paetau, A.; Andersson, N.; Sajantila, A.; Vapalahti, O.; Carpén, O.; Kekäläinen, E.; et al. Neuropathologic features of four autopsied COVID-19 patients. Brain Pathol. 2020, 30, 1012–1016. [Google Scholar] [CrossRef]
  31. Reichard, R.R.; Kashani, K.B.; Boire, N.A.; Constantopoulos, E.; Guo, Y.; Lucchinetti, C.F. Neuropathology of COVID-19: A spectrum of vascular and acute disseminated encephalomyelitis (ADEM)-like pathology. Acta Neuropathol. 2020, 140, 1–6. [Google Scholar] [CrossRef]
  32. Solomon, I.H.; Normandin, E.; Bhattacharyya, S.; Mukerji, S.S.; Keller, K.; Ali, A.S.; Adams, G.; Hornick, J.L.; Padera Jr, R.F. Neuropathological features of COVID-19. N. Engl. J. Med. 2020, 383, 989–992. [Google Scholar] [CrossRef] [PubMed]
  33. Kaur, C.; Ling, E.A. The circumventricular organs. Histol. Histopathol. 2017, 32, 879–892. [Google Scholar] [PubMed]
  34. Nampoothiri, S.; Sauve, F.; Ternier, G.; Fernandois, D.; Coelho, C.; Imbernon, M.; Deligia, E.; Perbet, R.; Florent, V.; Baroncini, M.; et al. The hypothalamus as a hub for putative SARS-CoV-2 brain infection. bioRxiv 2020. [Google Scholar] [CrossRef]
  35. Bergmann, C.C.; Lane, T.E.; Stohlman, S.A. Coronavirus infection of the central nervous system: Host–virus stand-off. Nat. Rev. Microbiol. 2006, 4, 121–132. [Google Scholar] [CrossRef] [PubMed]
  36. Engelhardt, B.; Vajkoczy, P.; Weller, R.O. The movers and shapers in immune privilege of the CNS. Nat. Immunol. 2017, 18, 123–131. [Google Scholar] [CrossRef] [PubMed]
  37. Merad, M.; Martin, J.C. Pathological inflammation in patients with COVID-19: A key role for monocytes and macrophages. Nat. Rev. Immunol. 2020, 20, 355–362. [Google Scholar] [CrossRef]
  38. Feng, Z.; Diao, B.; Wang, R.; Wang, G.; Wang, C.; Tan, Y.; Liu, L.; Wang, C.; Liu, Y.; Liu, Y.; et al. The novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) directly decimates human spleens and lymph nodes. medRxiv 2020. [Google Scholar] [CrossRef]
  39. Bost, P.; Giladi, A.; Liu, Y.; Bendjelal, Y.; Xu, G.; David, E.; Blecher-Gonen, R.; Cohen, M.; Medaglia, C.; Li, H.; et al. Host-viral infection maps reveal signatures of severe COVID-19 patients. Cell 2020, 181, 1475–1488. [Google Scholar] [CrossRef] [PubMed]
  40. Li, Y.C.; Bai, W.Z.; Hirano, N.; Hayashida, T.; Taniguchi, T.; Sugita, Y.; Tohyama, K.; Hashikawa, T. Neurotropic virus tracing suggests a membranous-coating-mediated mechanism for transsynaptic communication. J. Comp. Neurol. 2013, 521, 203–212. [Google Scholar] [CrossRef]
  41. Biswas, K.; Das Sarma, J. Effect of microtubule disruption on neuronal spread and replication of demyelinating and nondemyelinating strains of mouse hepatitis virus in vitro. J. Virol. 2014, 88, 3043–3047. [Google Scholar] [CrossRef]
  42. Matsuda, K.; Park, C.H.; Sunden, Y.; Kimura, T.; Ochiai, K.; Kida, H.; Umemura, T. The vagus nerve is one route of transneural invasion for intranasally inoculated influenza a virus in mice. Vet. Pathol. 2004, 41, 101–107. [Google Scholar] [CrossRef] [PubMed]
  43. Fenrich, M.; Mrdenovic, S.; Balog, M.; Tomic, S.; Zjalic, M.; Roncevic, A.; Mandic, D.; Debeljak, Z.; Heffer, M. SARS-CoV-2 dissemination through peripheral nerves explains multiple organ injury. Front. Cell. Neurosci. 2020, 14, 229. [Google Scholar] [CrossRef] [PubMed]
  44. Papatsiros, V.G.; Stylianaki, I.; Papakonstantinou, G.; Papaioannou, N.; Christodoulopoulos, G. Case report of transmissible gastroenteritis coronavirus infection associated with small intestine and brain lesions in piglets. Viral Immunol. 2019, 32, 63–67. [Google Scholar] [CrossRef] [PubMed]
  45. Sanchis-Gomar, F.; Lavie, C.J.; Perez-Quilis, C.; Henry, B.M.; Lippi, G. Angiotensin-converting enzyme 2 and antihypertensives (angiotensin receptor blockers and angiotensin-converting enzyme inhibitors) in coronavirus disease 2019. Mayo Clin. Proc. 2020, 95, 1222–1230. [Google Scholar] [CrossRef] [PubMed]
  46. Zuckerkandl, E.; Linus, P. Evolutionary divergence and convergence in proteins. In Evolving Genes and Proteins; Academic Press: Cambridge, MA, US, 1965; pp. 97–166. [Google Scholar]
  47. Beacon, T.H.; Su, R.C.; Lakowski, T.M.; Delcuve, G.P.; Davie, J.R. SARS-CoV-2 multifaceted interaction with the human host. Part II: Innate immunity response, immunopathology, and epigenetics. IUBMB Life 2020, 72, 2331–2354. [Google Scholar] [CrossRef] [PubMed]
  48. Sundar, U.S.; Shah, M.H.; Merchant, S.A.; Asole, D.C. Acute ataxia and myoclonus in COVID-19: A case series. Ann. Indian Acad. Neurol. 2021, 24, 807–809. [Google Scholar] [CrossRef] [PubMed]
  49. 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]
  50. Liotta, E.M.; Batra, A.; Clark, J.R.; Shlobin, N.A.; Hoffman, S.C.; Orban, Z.S.; Koralnik, I.J. Frequent neurologic manifestations and encephalopathy-associated morbidity in COVID-19 patients. Ann. Clin. Transl. Neurol. 2020, 7, 2221–2230. [Google Scholar] [CrossRef] [PubMed]
  51. Wu, Y.; Guo, C.; Tang, L.; Hong, Z.; Zhou, J.; Dong, X.; Yin, H.; Xiao, Q.; Tang, Y.; Qu, X.; et al. Prolonged presence of SARS-CoV-2 viral RNA in fecal samples. Lancet Gastroenterol. Hepatol. 2020, 5, 434–435. [Google Scholar] [CrossRef]
  52. 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]
  53. Beretta, S.; Cristillo, V.; Camera, G.; Morotti Colleoni, C.; Pellitteri, G.; Viti, B.; Bianchi, E.; Gipponi, S.; Grimoldi, M.; Valente, M.; et al. Incidence and long-term functional outcome of neurologic disorders in hospitalized patients with COVID-19 infected with pre-omicron variants. Neurology 2023, 101, e892–e903. [Google Scholar] [CrossRef]
  54. de Melo, G.D.; Perraud, V.; Alvarez, F.; Vieites-Prado, A.; Kim, S.; Kergoat, L.; Coleon, A.; Trüeb, B.S.; Tichit, M.; Piazza, A.; et al. Neuroinvasion and anosmia are independent phenomena upon infection with SARS-CoV-2 and its variants. Nat. Commun. 2023, 14, 4485. [Google Scholar] [CrossRef] [PubMed]
  55. TTaquet, M.; Sillett, R.; Zhu, L.; Mendel, J.; Camplisson, I.; Dercon, Q.; Harrison, P.J. Neurological and psychiatric risk trajectories after SARS-CoV-2 infection: An analysis of 2-year retrospective cohort studies including 1,284,437 patients. Lancet Psychiatry 2022, 9, 815–827. [Google Scholar] [CrossRef] [PubMed]
  56. Proust, A.; Queval, C.J.; Harvey, R.; Adams, L.; Bennett, M.; Wilkinson, R.J. Differential effects of SARS-CoV-2 variants on central nervous system cells and blood–brain barrier functions. J. Neuroinflamm. 2023, 20, 184. [Google Scholar] [CrossRef] [PubMed]
  57. Nyberg, T.; Ferguson, N.M.; Nash, S.G.; Webster, H.H.; Flaxman, S.; Andrews, N.; Hinsley, W.; Bernal, J.L.; Kall, M.; Bhatt, S.; et al. Comparative analysis of the risks of hospitalisation and death associated with SARS-CoV-2 omicron (B. 1.1. 529) and delta (B. 1.617. 2) variants in England: A cohort study. Lancet 2022, 399, 1303–1312. [Google Scholar] [CrossRef] [PubMed]
  58. Paterson, R.W.; Brown, R.L.; Benjamin, L.; Nortley, R.; Wiethoff, S.; Bharucha, T.; Jayaseelan, D.L.; Kumar, G.; Raftopoulos, R.E.; Zambreanu, L.; et al. The emerging spectrum of COVID-19 neurology: Clinical, radiological and laboratory findings. Brain 2020, 143, 3104–3120. [Google Scholar] [CrossRef] [PubMed]
  59. Lechien, J.R.; Chiesa-Estomba, C.M.; De Siati, D.R.; Horoi, M.; Le Bon, S.D.; Rodriguez, A.; Dequanter, D.; Blecic, S.; El Afia, F.; Distinguin, L.; et al. Olfactory and gustatory dysfunctions as a clinical presentation of mild-to-moderate forms of the coronavirus disease (COVID-19): A multicenter European study. Eur. Arch. Oto-Rhino Laryngol. 2020, 277, 2251–2261. [Google Scholar] [CrossRef] [PubMed]
  60. Aggarwal, S.; Garcia-Telles, N.; Aggarwal, G.; Lavie, C.; Lippi, G.; Henry, B.M. Clinical features, laboratory characteristics, and outcomes of patients hospitalized with coronavirus disease 2019 (COVID-19): Early report from the United States. Diagnosis 2020, 7, 91–96. [Google Scholar] [CrossRef] [PubMed]
  61. Massion, S.P.; Howa, A.C.; Zhu, Y.; Kim, A.; Halasa, N.; Chappell, J.; McGonigle, T.; Mellis, A.M.; Deyoe, J.E.; Reed, C.; et al. Sex differences in COVID-19 symptom severity and trajectories among ambulatory adults. Influenza Other Respir. Viruses 2023, 17, e13235. [Google Scholar] [CrossRef] [PubMed]
  62. Vashisht, A.; Ahluwalia, P.; Mondal, A.K.; Singh, H.; Sahajpal, N.S.; Fulzele, S.; Kota, V.; Gahlay, G.K.; Kolhe, R. Immune Factors Drive Expression of SARS-CoV-2 Receptor Genes Amid Sexual Disparity. Viruses 2023, 15, 657. [Google Scholar] [CrossRef] [PubMed]
  63. Spinato, G.; Fabbris, C.; Polesel, J.; Cazzador, D.; Borsetto, D.; Hopkins, C.; Boscolo-Rizzo, P. Alterations in smell or taste in mildly symptomatic outpatients with SARS-CoV-2 infection. JAMA 2020, 323, 2089–2090. [Google Scholar] [CrossRef]
  64. Beltrán-Corbellini, Á.; Chico-García, J.L.; Martínez-Poles, J.; Rodríguez-Jorge, F.; Natera-Villalba, E.; Gómez-Corral, J.; Gómez-López, A.; Monreal, E.; Parra-Díaz, P.; Cortés-Cuevas, J.L.; et al. Acute-onset smell and taste disorders in the context of COVID-19: A pilot multicentre polymerase chain reaction based case–control study. Eur. J. Neurol. 2020, 27, 1738–1741. [Google Scholar] [CrossRef] [PubMed]
  65. Moein, S.T.; Hashemian, S.M.; Mansourafshar, B.; Khorram-Tousi, A.; Tabarsi, P.; Doty, R.L. Smell dysfunction: A biomarker for COVID-19. Int. Forum Allergy Rhinol. 2020, 10, 944–950. [Google Scholar] [CrossRef] [PubMed]
  66. Oxley, T.J.; Mocco, J.; Majidi, S.; Kellner, C.P.; Shoirah, H.; Singh, I.P.; De Leacy, R.A.; Shigematsu, T.; Ladner, T.R.; Yaeger, K.A.; et al. Large-vessel stroke as a presenting feature of COVID-19 in the young. N. Engl. J. Med. 2020, 382, e60. [Google Scholar] [CrossRef] [PubMed]
  67. Varghese, G.M.; John, R.; Manesh, A.; Karthik, R.; Abraham, O.C. Clinical management of COVID-19. Indian J. Med. Res. 2020, 151, 401. [Google Scholar] [CrossRef] [PubMed]
  68. 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] [PubMed]
  69. 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]
  70. Gaebler, C.; Wang, Z.; Lorenzi, J.C.; Muecksch, F.; Finkin, S.; Tokuyama, M.; Cho, A.; Jankovic, M.; Schaefer-Babajew, D.; Oliveira, T.Y.; et al. Evolution of antibody immunity to SARS-CoV-2. Nature 2021, 591, 639–644. [Google Scholar] [CrossRef] [PubMed]
  71. Kim, J.E.; Heo, J.H.; Kim, H.O.; Song, S.H.; Park, S.S.; Park, T.H.; Ahn, J.Y.; Kim, M.K.; Choi, J.P. Neurological complications during treatment of Middle East respiratory syndrome. J. Clin. Neurol. 2017, 13, 227–233. [Google Scholar] [CrossRef] [PubMed]
  72. Tsai, L.K.; Hsieh, S.T.; Chao, C.C.; Chen, Y.C.; Lin, Y.H.; Chang, S.C.; Chang, Y.C. Neuromuscular disorders in severe acute respiratory syndrome. Arch. Neurol. 2004, 61, 1669–1673. [Google Scholar] [CrossRef] [PubMed]
  73. Jacomy, H.; Fragoso, G.; Almazan, G.; Mushynski, W.E.; Talbot, P.J. Human coronavirus OC43 infection induces chronic encephalitis leading to disabilities in BALB/C mice. Virology 2006, 349, 335–346. [Google Scholar] [CrossRef]
  74. Fierz, W. Multiple sclerosis: An example of pathogenic viral interaction? Virol. J. 2017, 14, 1–5. [Google Scholar] [CrossRef] [PubMed]
  75. Crow, M.K.; Olferiev, M.; Kirou, K.A. Type I interferons in autoimmune disease. Annu. Rev. Pathol. Mech. Dis. 2019, 14, 369–393. [Google Scholar] [CrossRef] [PubMed]
  76. Arbour, N.; Day, R.; Newcombe, J.; Talbot, P.J. Neuroinvasion by human respiratory coronaviruses. J. Virol. 2000, 74, 8913–8921. [Google Scholar] [CrossRef] [PubMed]
  77. Boucher, A.; Desforges, M.; Duquette, P.; Talbot, P.J. Long-term human coronavirus-myelin cross-reactive T-cell clones derived from multiple sclerosis patients. Clin. Immunol. 2007, 123, 258–267. [Google Scholar] [CrossRef] [PubMed]
  78. Kim, S.; Kwon, S.H.; Kam, T.I.; Panicker, N.; Karuppagounder, S.S.; Lee, S.; Lee, J.H.; Kim, W.R.; Kook, M.; Foss, C.A.; et al. Transneuronal propagation of pathologic α-synuclein from the gut to the brain models Parkinson’s disease. Neuron 2019, 103, 627–641. [Google Scholar] [CrossRef] [PubMed]
  79. Srikantha, P.; Mohajeri, M.H. The possible role of the microbiota-gut-brain-axis in autism spectrum disorder. Int. J. Mol. Sci. 2019, 20, 2115. [Google Scholar] [CrossRef]
  80. Mavrikaki, M.; Lee, J.D.; Solomon, I.H.; Slack, F.J. Severe COVID-19 induces molecular signatures of aging in the human brain. Nat. Aging 2022, 2, 1130–1137. [Google Scholar] [CrossRef] [PubMed]
  81. Samudyata, N.; Oliveira, A.O.; Malwade, S.; Rufino de Sousa, N.; Goparaju, S.K.; Gracias, J.; Orhan, F.; Steponaviciute, L.; Schalling, M.; Sheridan, S.D.; et al. SARS-CoV-2 promotes microglial synapse elimination in human brain organoids. Mol. Psychiatry 2022, 27, 3939–3950. [Google Scholar] [CrossRef]
  82. Wang, Y.; Xu, E.; Musich, P.R.; Lin, F. Mitochondrial dysfunction in neurodegenerative diseases and the potential countermeasure. CNS Neurosci. Ther. 2019, 25, 816–824. [Google Scholar] [CrossRef] [PubMed]
  83. Ganji, R.; Reddy, P.H. Impact of COVID-19 on mitochondrial-based immunity in aging and age-related diseases. Front. Aging Neurosci. 2021, 12, 614650. [Google Scholar] [CrossRef]
  84. Yang, J.; Zheng, Y.A.; Gou, X.; Pu, K.; Chen, Z.; Guo, Q.; Ji, R.; Wang, H.; Wang, Y.; Zhou, Y. Prevalence of comorbidities and its effects in patients infected with SARS-CoV-2: A systematic review and meta-analysis. Int. J. Infect. Dis. 2020, 94, 91–95. [Google Scholar] [CrossRef] [PubMed]
  85. Pei, H.; Wang, Y.; Zhang, X.; Luo, W.; Zhou, C. Association of frailty status with adverse clinical outcomes in patients with COVID-19: Protocol for a systematic review and dose–response meta-analysis. BMJ Open 2021, 11, e046980. [Google Scholar] [CrossRef] [PubMed]
  86. Williams, E.S.; Martins, T.B.; Shah, K.S.; Hill, H.R.; Coiras, M.; Spivak, A.M.; Planelles, V. Cytokine Deficiencies in Patients with Long-COVID. J. Clin. Cell. Immunol. 2022, 13, 672. [Google Scholar] [PubMed]
  87. Franke, C.; Ferse, C.; Kreye, J.; Reincke, S.M.; Sanchez-Sendin, E.; Rocco, A.; Steinbrenner, M.; Angermair, S.; Treskatsch, S.; Zickler, D.; et al. High frequency of cerebrospinal fluid autoantibodies in COVID-19 patients with neurological symptoms. Brain Behav. Immun. 2021, 93, 415–419. [Google Scholar] [CrossRef] [PubMed]
  88. GGarcia, M.A.; Barreras, P.V.; Lewis, A.; Pinilla, G.; Sokoll, L.J.; Kickler, T.; Mostafa, H.; Caturegli, M.; Moghekar, A.; Fitzgerald, K.C.; et al. Cerebrospinal fluid in COVID-19 neurological complications: No cytokine storm or neuroinflammation. medRxiv 2021. [Google Scholar] [CrossRef]
  89. Barros-Aragão, F.G.; Pinto, T.P.; Carregari, V.C.; Rezende, N.B.; Pinheiro, T.L.; Reis-de-Oliveira, G.; Cabral-Castro, M.J.; Queiroz, D.C.; Fonseca, P.L.; Gonçalves, A.L.; et al. Changes in neuroinflammatory biomarkers correlate with disease severity and neuroimaging alterations in patients with COVID-19 neurological complications. Brain Behav. Immun. Health 2024, 39, 100805. [Google Scholar] [CrossRef] [PubMed]
  90. Rothstein, A.; Oldridge, O.; Schwennesen, H.; Do, D.; Cucchiara, B.L. Acute cerebrovascular events in hospitalized COVID-19 patients. Stroke 2020, 51, e219–e222. [Google Scholar] [CrossRef] [PubMed]
  91. Tang, N.; Kido, T.; Shi, J.; McCafferty, E.; Ford, J.M.; Dal Bon, K.; Pulliam, L. Blood markers show neural consequences of longCOVID-19. Cells 2024, 13, 478. [Google Scholar] [CrossRef] [PubMed]
  92. Zielińska-Turek, J.; Turek, G.; Lyson, T.; Gajewski, J.; Ząbek, M.; Dorobek, M. Inflammatory proteins as strong predictors of death in COVID-19 patients with coexisting neurological diseases. medRxiv 2024. [Google Scholar] [CrossRef]
  93. Sudo, F.K.; Pinto, T.P.; Barros-Aragão, F.G.; Bramati, I.; Marins, T.F.; Monteiro, M.; Meireles, F.; Soares, R.; Erthal, P.; Calil, V.; et al. Cognitive, behavioral, neuroimaging and inflammatory biomarkers after hospitalization for COVID-19 in Brazil. Brain Behav. Immun. 2024, 115, 434–447. [Google Scholar] [CrossRef]
  94. Wang, A.; Mandigo, G.K.; Yim, P.D.; Meyers, P.M.; Lavine, S.D. Stroke and mechanical thrombectomy in patients with COVID-19: Technical observations and patient characteristics. J. Neurointerventional Surg. 2020, 12, 648–653. [Google Scholar] [CrossRef] [PubMed]
  95. Escalard, S.; Maïer, B.; Redjem, H.; Delvoye, F.; Hébert, S.; Smajda, S.; Ciccio, G.; Desilles, J.P.; Mazighi, M.; Blanc, R.; et al. Treatment of acute ischemic stroke due to large vessel occlusion with COVID-19: Experience from Paris. Stroke 2020, 51, 2540–2543. [Google Scholar] [CrossRef] [PubMed]
  96. Siow, I.; Lee, K.S.; Zhang, J.J.; Saffari, S.E.; Ng, A. Encephalitis as 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] [PubMed]
  97. Aladawi, M.; Elfil, M.; Abu-Esheh, B.; Jazar, D.A.; Armouti, A.; Bayoumi, A.; Piccione, E. Guillain Barre syndrome as a complication of COVID-19: A systematic review. Can. J. Neurol. Sci. 2022, 49, 38–48. [Google Scholar] [CrossRef] [PubMed]
  98. Puntambekar, S.S.; Hinton, D.R.; Yin, X.; Savarin, C.; Bergmann, C.C.; Trapp, B.D.; Stohlman, S.A. Interleukin-10 is a critical regulator of white matter lesion containment following viral induced demyelination. Glia 2015, 63, 2106–2120. [Google Scholar] [CrossRef] [PubMed]
  99. Glezer, I.; Rivest, S. Oncostatin M is a novel glucocorticoid-dependent neuroinflammatory factor that enhances oligodendrocyte precursor cell activity in demyelinated sites. Brain Behav. Immun. 2010, 24, 695–704. [Google Scholar] [CrossRef]
  100. Liu, S.; Jin, R.; Xiao, A.Y.; Zhong, W.; Li, G. Inhibition of CD147 improves oligodendrogenesis and promotes white matter integrity and functional recovery in mice after ischemic stroke. Brain Behav. Immun. 2019, 82, 13–24. [Google Scholar] [CrossRef]
  101. Haber, P.; Sejvar, J.; Mikaeloff, Y.; DeStefano, F. Vaccines and guillain-barre syndrome. Drug Saf. 2009, 32, 309–323. [Google Scholar] [CrossRef]
  102. O’Brien, M.P.; Forleo-Neto, E.; Sarkar, N.; Isa, F.; Hou, P.; Chan, K.C.; Musser, B.J.; Bar, K.J.; Barnabas, R.V.; Barouch, D.H.; et al. Effect of subcutaneous casirivimab and imdevimab antibody combination vs placebo on development of symptomatic COVID-19 in early asymptomatic SARS-CoV-2 infection: A randomized clinical trial. JAMA 2022, 327, 432–441. [Google Scholar] [CrossRef]
  103. Streinu-Cercel, A.; Săndulescu, O.; Preotescu, L.L.; Kim, J.Y.; Kim, Y.S.; Cheon, S.; Jang, Y.R.; Lee, S.J.; Kim, S.H.; Chang, I.; et al. Efficacy and safety of regdanvimab (CT-P59): A phase 2/3 randomized, double-blind, placebo-controlled trial in outpatients with mild-to-moderate coronavirus disease 2019. In Open Forum Infectious Diseases; Oxford University Press: Oxford, MA, USA, 2022; Volume 9, p. ofac053. [Google Scholar]
  104. Cuenca-Zaldivar, J.N.; Monroy Acevedo, Á.; Fernández-Carnero, J.; Sánchez-Romero, E.A.; Villafañe, J.H.; Barragán Carballar, C. Effects of a Multicomponent Exercise Program on Improving Frailty in Post-COVID-19 Older Adults after Intensive Care Units: A Single-Group Retrospective Cohort Study. Biology 2022, 11, 1084. [Google Scholar] [CrossRef]
Figure 1. Schematic representation of possible routes through which SARS-CoV-2 could invade the nervous system. (a) Blood–brain barrier: the virus may be able to breach this barrier and enter the brain, potentially causing inflammation and other neurological complications; (b) Retrograde axonal transport, which refers to the transport of the virus from peripheral nerves back to the brain. Once the virus enters the peripheral nervous system, it may be transported along nerve fibers back to the central nervous system.
Figure 1. Schematic representation of possible routes through which SARS-CoV-2 could invade the nervous system. (a) Blood–brain barrier: the virus may be able to breach this barrier and enter the brain, potentially causing inflammation and other neurological complications; (b) Retrograde axonal transport, which refers to the transport of the virus from peripheral nerves back to the brain. Once the virus enters the peripheral nervous system, it may be transported along nerve fibers back to the central nervous system.
Viruses 16 01183 g001aViruses 16 01183 g001b
Table 2. Management of neurological complications associated with COVID-19: therapeutic strategies and considerations.
Table 2. Management of neurological complications associated with COVID-19: therapeutic strategies and considerations.
Aspect of ManagementTreatment Details
Acute Stroke TherapiesAcute stroke therapies (thrombolysis and thrombectomy) not specifically tested in COVID-19 population; standard care applies as for non-COVID-19 patients. Evaluation for candidacy should be timely.
Seizure ManagementNo specific data on seizure management in COVID-19 patients; general risk–benefit ratio applies.
Encephalopathy Treatment OptionsReported options include monotherapy or combinations of corticosteroids (e.g., methylprednisolone 1 g daily for 5–10 days), intravenous immunoglobulin, plasma exchange, and rituximab.
Ischemic and Hemorrhagic Stroke ManagementFollow same standards of care as non-COVID-19 patients, including timely evaluation for acute medical and interventional stroke therapies.
Guillain-Barré Syndrome (GBS)Consider if respiratory insufficiency is disproportionate to pulmonary findings in COVID-19 patients. Intravenous immunoglobulin and plasma exchange are reported therapies.
Autonomic Nervous SystemImmunomodulatory actions may help prevent the hyper-inflammatory state, leading to severe COVID-19 symptoms. Maintaining an efficient immune response may minimize tissue damage, including CNS involvement.
IL-10 TherapyNo study comprising human samples so far but in MHV-infected mice, IL-10 therapy causes astrocytes to produce glial scars that restrict demyelination regions.
CD147 InhibitionIn mouse models of ischemic stroke, the inhibition of CD147 (a SARS-CoV-2 receptor) helps preserve oligodendrocytes and white matter. However, antibodies against CD147 can increase brain inflammation, presenting a double-edged sword effect.
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

Vashisht, A.; Vashisht, V.; Singh, H.; Ahluwalia, P.; Mondal, A.K.; Williams, C.; Farmaha, J.; Woodall, J.; Kolhe, R. Neurological Complications of COVID-19: Unraveling the Pathophysiological Underpinnings and Therapeutic Implications. Viruses 2024, 16, 1183. https://doi.org/10.3390/v16081183

AMA Style

Vashisht A, Vashisht V, Singh H, Ahluwalia P, Mondal AK, Williams C, Farmaha J, Woodall J, Kolhe R. Neurological Complications of COVID-19: Unraveling the Pathophysiological Underpinnings and Therapeutic Implications. Viruses. 2024; 16(8):1183. https://doi.org/10.3390/v16081183

Chicago/Turabian Style

Vashisht, Ashutosh, Vishakha Vashisht, Harmanpreet Singh, Pankaj Ahluwalia, Ashis K. Mondal, Colin Williams, Jaspreet Farmaha, Jana Woodall, and Ravindra Kolhe. 2024. "Neurological Complications of COVID-19: Unraveling the Pathophysiological Underpinnings and Therapeutic Implications" Viruses 16, no. 8: 1183. https://doi.org/10.3390/v16081183

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

Article Metrics

Article metric data becomes available approximately 24 hours after publication online.
Back to TopTop