Abstract
With the progression of investigations on the pathogenesis of severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2), neurological complications have emerged as a critical aspect of the ongoing coronavirus disease 2019 (Covid‐19) pandemic. Besides the well‐known respiratory symptoms, many neurological manifestations such as anosmia/ageusia, headaches, dizziness, seizures, and strokes have been documented in hospitalised patients. The neurotropism background of coronaviruses has led to speculation that the neurological complications are caused by the direct invasion of SARS‐CoV‐2 into the nervous system. This invasion is proposed to occur through the infection of peripheral nerves or via systemic blood circulation, termed neuronal and haematogenous routes of invasion, respectively. On the other hand, aberrant immune responses and respiratory insufficiency associated with Covid‐19 are suggested to affect the nervous system indirectly. Deleterious roles of cytokine storm and hypoxic conditions in blood‐brain barrier disruption, coagulation abnormalities, and autoimmune neuropathies are well investigated in coronavirus infections, as well as Covid‐19. Here, we review the latest discoveries focussing on possible molecular mechanisms of direct and indirect impacts of SARS‐CoV‐2 on the nervous system and try to elucidate the link between some potential therapeutic strategies and the molecular pathways.
Keywords: central nervous system, Covid‐19, post‐COVID‐19 syndrome, SARS‐CoV‐2
Abbreviations
- ADAM17
a disintegrin and metalloproteinase domain 17
- ALT
alanine aminotransferase
- Ang II
Angiotensin II
- APC
antigen presenting cell
- ARDS
acute respiratory distress syndrome
- BBB
blood‐brain barrier
- BLAST
basic local alignment search tool
- BSG
basigin
- CD147
cluster of differentiation 147
- CLR
c‐type lectin receptor
- CNS
central nervous system
- Covid‐19
coronavirus disease of 2019
- COX
cyclooxygenase
- CSF
cerebrospinal fluid
- CVD
cardiovascular disease
- CXCL
(C‐X‐C motif) ligand
- DAMPs
damage‐associated molecular patterns
- DIC
disseminated intravascular coagulation
- DMF
dimethyl fumarate
- DMTs
disease modifying therapies
- EC
endothelial cell
- ECM
extracellular matrix
- eNOS
endothelial nitric oxide synthase
- ENS
enteric nervous system
- FAT
fast axonal transport
- GBS
Guillain‐Barre syndrome
- G‐CSF
granulocyte colony‐stimulating factor
- GRP78
Glucose‐Regulated Protein 78
- hACE2
human angiotensin converting enzyme 2
- HBC
horizontal basal cell
- hCoVs
human coronaviruses
- HIF‐1α
hypoxia inducible factor‐1α
- HIV
human immunodeficiency virus
- HPA axis
hypothalamic‐pituitary‐adrenal axis
- hsCRP
high‐sensitivity C‐reactive protein
- ICAM
intercellular adhesion molecule
- IFN
interferon
- Ig
immunoglobulin
- IL
interleukin
- IP‐10
interferon‐gamma‐induced protein‐10
- IVIg
intravenous immunoglobulin
- JAK‐STAT
Janus kinase‐signal transducer and activator of transcription
- LDH
lactate dehydrogenase
- LTs
leukotrienes
- MCP‐1
monocyte chemoattractant protein‐1
- MERS‐CoV
middle east respiratory syndrome
- MMP
matrix metalloproteinases
- MRI
magnetic resonance imaging
- MS
multiple sclerosis
- NETs
neutrophil extracellular traps
- NF‐κB
nuclear factor kappa‐light‐chain‐enhancer of activated B cells
- NLR
nod‐like receptor
- NLRP3
NLR family pyrin domain containing 3
- NRP1
neuropilin 1
- NVU
neurovascular unit
- OB
olfactory bulb
- OE
olfactory epithelium
- OSN
olfactory sensory neuron
- PAMPs
pathogen‐associated molecular patterns
- PCF
pro‐protein convertase furin
- PGE
prostaglandin E
- PGs
prostaglandins
- PNS
peripheral nervous system
- PRR
pattern recognition receptor
- PT
prothrombin time
- ROS
reactive oxygen species
- RSV
respiratory syncytial virus
- SARS‐CoV‐1
severe acute respiratory syndrome coronavirus 1
- SARS‐CoV‐2
severe acute respiratory syndrome coronavirus 2
- spO2
oxygen saturation
- T2D
type 2 diabetes
- TAG‐1
transient axonal glycoprotein‐1
- TFPI
tissue factor pathway inhibitor
- Th2
T helper 2
- THBD
thrombomodulin
- TJ
tight junction
- TLR
toll‐like receptor
- TMPRSS2
Transmembrane Serine Protease 2
- TNF
tumour necrosis factor
- VCAM
vascular cell adhesion molecule
- VEGF
vascular endothelial growth factor
- WHO
World Health Organization
- WNV
west Nile virus
- ZO‐1
zonula occludens‐1
1. INTRODUCTION
The most recent life‐threatening outbreak is caused by severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2) with a high transmission rate, which has turned into a worldwide challenge with the first breakout reported in the Hubei province Wuhan, December 2019. 1 , 2 , 3 , 4 This ongoing outbreak was first known as an epidemic by the World Health Organization (WHO). Then, considering the rapid spread worldwide, the WHO declared the current outbreak a pandemic on 11 March 2020. 5 Other epidemics were also caused in the past two decades by other coronaviruses (CoVs) such as SARS‐CoV and MERS‐CoV in 2003 and 2012, 5 , 6 respectively, both inducing severe viral pneumonia with respiratory failure 1 and neurological manifestations. 2 , 7 , 8 Common respiratory viruses, including CoVs, influenza, and respiratory syncytial virus (RSV), can be associated with various neurological impairments. 5 , 9 , 10 , 11 , 12 CoVs are responsible for various respiratory, gastrointestinal, hepatic, and neurological diseases with different severity levels. 2 , 3 The presence of SARS‐CoV‐2 RNA in the cerebrospinal fluid (CSF) of some coronavirus disease 2019 (Covid‐19) patients and abnormal brain magnetic resonance imaging (MRI) findings might be convincing evidence supporting Covid‐19 neuroinvasion and neurovirulence. 13 , 14 , 15 , 16 Considering central nervous system (CNS) and peripheral nervous system (PNS) susceptibility to the SARS‐CoV‐2 infection, chronic or permanent changes to several parts of the nervous system could lead to multiple neurological manifestations, including encephalopathy, 17 encephalitis, 18 seizures, 19 headache, 5 , 7 anosmia and ageusia, 20 demyelination 21 , 22 and neuropsychiatric disorders, 23 which needs to be precisely investigated and treated. 24
2. MECHANISM OF CELLULAR VIRAL INFECTION
Coronaviruses mainly use their Spike (S) protein to enter the host cell. SARS‐CoV and SARS‐CoV‐2 share approximately 70% of their sequence identity in the Spike protein, and both utilise the human angiotensin converting enzyme 2 (hACE2) receptor of the host cell for cell entry. 25 , 26 , 27 Noteworthy, the binding affinity of SARS‐CoV‐2 S protein to ACE2 receptor is 10–20 times higher than that of SARS‐CoV due to some structural differences in the receptor binding domain of S protein. 28 , 29 In addition, the production of angiotensin II (Ang II) is counterbalanced by ACE2. The primary function of ACE2 is the degradation of Ang II and formation of Ang 1‐7 to neutralise the vasoconstrictor effect of Ang II and maintain blood pressure. 26 , 27 , 29 , 30 , 31 ACE2 is highly expressed in the small intestine, kidney, heart, adipose tissue, thyroid, testis, and pancreas, whereas the muscles, brain, spleen, and blood vessels have the lowest ACE2 expression. 32 , 33 Besides, lungs, liver, bladder, and colon have shown a medium ACE2 expression. 33 , 34 , 35 Moreover, ACE2 receptor is expressed in higher levels in the lungs of smokers and type 2 diabetes (T2D) patients and in the heart of patients with cardiovascular disease (CVD), which makes them more susceptible to be infected by SARS‐CoV‐2. 33 In addition, an increase in mRNA levels of ACE2 in Covid‐19 patients has been reported. 33 Low expression of ACE2 in respiratory tissues and high rates of infection in these tissues, lead to the speculation of plausible alternative receptors. Several experiments support the role of co‐receptors and attachment factors such as transmembrane serine protease 2 (TMPRSS2), basigin (BSG) (also known as CD147), GRP78, some toll‐like receptors (TLRs) and c‐type lectin receptors (CLRs), heparan sulfate, and sialic acids, which facilitate and enhance the entry of the virus in the presence of ACE2. 26 , 33 , 36 Nevertheless, these co‐receptors may not be sufficient for virus entry into some specific cells not expressing ACE2 although deletion of co‐receptors may reduce infection. 26 , 33 , 37 , 38 , 39 Furthermore, Neuropilin 1 (NRP1) mainly facilitates the regulation of angiogenesis, gangliogenesis, and vascular permeability, and similarly, enhances viral infectivity and acts as a co‐receptor for cell entry. 26 , 40 Noteworthy, NRP1 and BSG are found more than ACE2 and TMPRSS2 in the brain, including the olfactory bulb (OB). 39
The pathogenesis of SARS‐CoV‐2 is mediated via the interaction between receptors and the spike protein of the virus for viral attachment, then utilising diverse endocytic pathways for entry. 41 , 42 To elaborate, after binding the receptor binding domain of S protein to the peptidase domain of ACE2, the SARS‐CoV‐2/ACE2 complex is formed. Subsequently, TMPRSS2, which activates and cleaves S protein, 25 , 28 , 29 is activated for S priming, and then the SARS‐CoV‐2/ACE2 complex undergoes endocytosis and forms an endosome. After acidification of the endosome and fusion of viral and lysosomal membranes, 41 encapsidated viral RNA is released to the cytoplasm for replication and transcription. 26 , 42 , 43 Complementary to this, SARS‐CoV‐2 may utilise CD147‐mediated endocytosis for cell entry, 44 and pro‐protein convertase furin (PCF) might also play a role in endocytic pathways.PCF is essential for the propagation of numerous viruses by cleavage of viral envelope glycoproteins, which may also involve the endocytosis of SARS‐CoV‐2. 45 Due to the expression of ACE2 and CD147 and other plausible receptors in the circumventricular organs of CNS, glial cells, and neurons, 30 , 31 , 46 , 47 SARS‐CoV‐2 can potentially invade the nervous system, which leads to neurological manifestations. Neurological impairments are not limited to direct infection by the virus. Other indirect effects of infection, including the immune response and cytokine storm, can also damage the nervous system, as explained in subsequent sections of this review. 48 Considering the high pathophysiological pathway similarity between SARS‐CoV and SARS‐CoV‐2, they might share putative routes for CNS invasion, which are elaborated below.
3. POTENTIOAL ROUTES OF DIRECT CNS INVASIONS
3.1. The neuronal route of invasion
The main transmission factor of SARS‐CoV‐2 is the droplets of Covid‐19 patients who cough and sneeze and release the droplets into the air. 49 Thus, clarifying the potential intranasal and oral routes of SARS‐CoV‐2 CNS invasion is required to understand the development of anosmia, ageusia, and other nervous system dysfunctions. After entering via droplets containing SARS‐CoV‐2 in the nasal cavity, viruses can either reach the lung through the airway or land on the nasal mucosa and infect susceptible cells. 50 The olfactory epithelium (OE) of the nasal cavity contains olfactory sensory neurons (OSNs), basal cells, epithelial cilia, and Bowman's gland for mucus secretion and homoeostatic electrolyte balance 51 (Figure 1a). Horizontal basal cells (HBCs) of the OE are directly attached to the basal lamina and are progenitors of OSNs. It is believed that OSNs do not express ACE2; it is expressed in HBCs, which then mature into OSNs. Infected HBCs mature into bipolar unmyelinated OSNs and then penetrate the cribriform plate and access the OB by a synaptic path 39 , 52 , 53 , 54 (Figure 1b). Subsequently, the virus could infect mitral cells of the OB, which are connected to several parts of the brain, and facilitate infection of other susceptible regions of the nervous system, including the cortex, the mesolimbic cortex, hippocampus, amygdala, and eventually brainstem and spinal cord via a trans‐synaptic pathway, using endocytosis and exocytosis 39 , 48 (Figure 1d). Similar animal experiments using MERS‐CoV, SARS‐CoV, and SARS‐CoV‐2 support HBC and OSN infection as a precurser to reaching the CNS via olfactory nerves. 55 , 56 , 57 The majority of Covid‐19 patients experience smell or taste disorders 58 , 59 , 60 , 61 ; this neurotoxic effect of SARS‐CoV‐2 might be due to changes in phosphorylation pattern of proteins associated with axons and synapses in olfactory/gustatory neurons or injuries to any of VII, IX, X cranial nerves and the nucleus of solitary tract. 50 , 62 Moreover, the molecular mechanism of virus transportation inside PNS and brain parenchyma neurons is almost identified in the neuronal route. Due to the high length of peripheral nerve axons, the migration of the virus via diffusion could not be possible. 62 Thus, experimental results suggest another propagation mechanism named fast axonal transport (FAT), which is mainly used by hCoVs to spread along neuronal cells. 63 After the endocytosis of the virus to peripheral neurons and formation of the endosomes, the endosome lysis occurs, and the virus undergoes retrograde trafficking to the cell body and nucleus via axonal microtubules, utilising the microtubule‐dependent motor proteins kinesin for anterograde and dynein for retrograde axonal transport. 48 , 62 , 64 , 65 Therefore, the envelope of SARS‐CoV‐2 should be stable during neuronal transport 66 (Figure 1c).
FIGURE 1.

Potential route of central nervous system invasion of severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2) via nasal cavity and axonal transport propagation. (a) Olfactory epithelium is located in the roof of the nasal cavity and its distinct cell types; (b) Proposed mechanism of olfactory sensory neuron (OSN) infection process by infection and differentiation of horizontal basal cells (HBCs) to OSNs and propagation of viruses to the olfactory bulb (OB) through the cribriform plate; (c) Axonal transport of viruses via retrograde and anterograde dissemination utilising Dynein for retrograde and Kinesin for anterograde transport to facilitate the infection of neuronal cells; (d) Trans‐synaptic pathway of virus propagation in the OB to infiltrate in the brain and infect more cells by exocytosis and endocytosis
In addition to the olfactory nerve, the virus could utilise other peripheral nerves to reach the CNS and brainstem, including the pulmonary network and enteric nervous system (ENS) via the vagus nerve. 50 , 53 , 66 , 67 NRP1 and ACE2 are highly expressed in the gastrointestinal tract; meanwhile, intestinal neurons and glia highly express ACE2 and TMPRSS2 and are susceptible to being infected by SARS‐CoV‐2. 39 , 49 , 64 In addition, viral nucleic acid has been detected in the stool of Covid‐19 patients, 68 , 69 , 70 , 71 which may be due to the infection of intestinal cells or self‐ingestion of mucus from the airways. 72 The enteric neuronal network is directly connected to the CNS through the parasympathetic vagus nerve arising from the hindbrain, and the sympathetic nerve fibres arising from the spine. 66 Hence, transferring the infection from the intestine to the CNS is possible in animal models. At the same time, there is not sufficient evidence in humans and requires attaining more data of vagal complex ACE2 expression and the ability of trans‐neuronal spread of SARS‐CoV‐2 in the gut‐brain axis. 39 In brief, considering the anatomically close distance of the olfactory nerves to the CNS, it can be suggested as the main pathway of neuronal dissemination of SARS‐CoV‐2 to reach the CNS in the early stages of the infection rather than the gut‐brain axis or other plausible neuronal routes.
3.2. The haematogenous route of invasion
As the second possible infectivity route, haematogenous dissemination of viral particles could provide entry into the CNS for SARS‐CoV‐2 via overcoming the barriers of CNS or through circumventricular organs. 73 , 74 , 75 Despite the wide range of frequency in results of different studies, dissemination of SARS‐CoV‐2 into the blood has been reported in up to 40% of patients with Covid‐19. 76 Circulating viral particles could cross the blood‐brain barrier (BBB) and invade the brain parenchyma, facilitating ACE2 receptors that are expressed by brain endothelial cells (ECs) and pericytes 74 , 77 , 78 (Figure 2a). In vitro, human vessel organoids are susceptible to SARS‐CoV‐2 infection in an ACE2‐dependent manner. 79 In line with this, clinical studies have observed the presence of viral elements in ECs of multiple organs in Covid‐19 patients; more specifically, an autopsy study in which electron micrographs indicated the presence of SARS‐CoV‐2 viral‐like proteins inside ECs of frontal lobe tissue of the brain. 80 , 81 Of note, there is evidence of cerebral vasculature wide expression of some SARS‐CoV‐2 alternative receptors such as NRP1 and BSG, which could be considered a synergistic factor for this entry route. 39 , 76 , 82 Moreover, increased secretion of pro‐inflammatory cytokines and chemokines and pneumonia‐induced hypoxia associated with Covid‐19 compromise the BBB integrity, expedite virus entry and contribute to CNS invasion by SARS‐CoV‐2. 74 , 83 , 84
FIGURE 2.

Possible mechanisms of severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2) haematogenous route of neuroinvasion. (a) Blood‐brain barrier (BBB). Brain endothelial cells (ECs) and pericytes are observed to express angiotensin converting enzyme 2 (ACE2) and other SARS‐CoV‐2 alternative receptors. This could facilitate the SARS‐CoV‐2 invasion to the brain tissue through the paracellular passage of the viral particles across the BBB; (b) choroid plexus. The barrier at the interface between the blood and the cerebrospinal fluid (CSF) consists of a more permeable endothelium due to the fenestrated structure of the ECs. Moreover, the choroid plexus epithelium cells at the apical side of the blood‐CSF barrier express ACE2. With these properties, the blood‐CSF barrier could serve as a SARS‐CoV‐2 entry gate to the CSF and then brain parenchyma; (c) circumventricular organs. Capillaries of the median eminence and other circumventricular organs lack the tightly coordinated BBB structure and consist of a continuous, fenestrated endothelium permeable to polypeptides and hormone molecules. Due to this extensive permeability, circumventricular organs could act as possible gateways to the brain tissue for SARS‐CoV‐2; (d) Trojan horse mechanism. SARS‐CoV‐2 could infect the leucocytes. Dissemination of infected leucocytes into the cerebral blood circulation and later extravasation of infected cells could facilitate SARS‐CoV‐2 entry to the brain parenchyma by the so‐called Trojan horse mechanism
The choroid plexus and circumventricular organs are other regions that could possibly act as entry gates to the brain for circulating SARS‐CoV‐2 particles. Termed as the blood‐CSF barrier, the choroid plexus is the selectively permeable structure that restricts the free diffusion of molecules at the blood‐CSF interface and contributes to CSF production. 64 , 78 ACE2 and TMPRSS2 are expressed by human choroid plexus cells, 85 and in vitro experiments that modelled the human choroid plexus by organoids demonstrated a high susceptibility of this tissue to SARS‐CoV‐2 infection. 64 , 86 Thus, the presence of SARS‐CoV‐2 in CSF, which has been reported by few case reports, 14 , 87 , 88 is likely to occur through the infection of choroid plexus; however, the indirect effects of SARS‐CoV‐2 on the CNS by disruption of the blood‐CSF barrier due to infection of choroid plexus may play a more critical role in the exhibition of neurological manifestation in Covid‐19. 86 On the other hand, circumventricular organs are highly vascularised structures adjacent to the third and fourth ventricles, characterised by their continuous fenestrated and extensively permeable vessels. 74 , 89 Preliminary data on the median eminence of the hypothalamus, one of the circumventricular organs, suggest the expression of ACE2 and TMPRSS2 in this tissue. 90 This could facilitate SARS‐CoV‐2 entry to the hypothalamus tissue and further spread of the virus to the entire brain, owing to the widespread connection of the hypothalamus to other centres of the brain 76 (Figure 2b,c).
Dissemination of virus‐infected leucocytes into the’ blood circulation and subsequent extravasation of the immune cells into the brain parenchyma could serve as another gateway for the virus to the CNS. The so‐called ‘Trojan horse’ mechanism has been well investigated previously in some of the neurotropic viruses such as HIV and West Nile Virus (WNV). 91 , 92 Infected leucocytes could infiltrate into the brain through the vasculature, the meninges, and the choroid plexus. These sites have been observed as entry points for monocytes, neutrophils, and T cells. 77 There are indications that SARS‐CoV could infect lymphocytes, granulocytes, monocytes, and monocyte derivatives 93 ; thus, it is likely that SARS‐CoV‐2 also utilises this mechanism in order to invade the CNS by infecting ACE2‐expressing leucocytes. 45 , 94 SARS‐CoV‐2 is shown to abortively infect dendritic cells and macrophages. 95 This evidence, in conjunction with systemic inflammation and hypoxic condition that increase the infiltration of leucocytes through the BBB during the infection, 45 , 96 strengthens the feasibility of SARS‐CoV‐2 neuroinvasion by this route (Figure 2d).
4. COVID‐19 ASSOCIATED CYTOKINE STORM
Considering various clinical observations, Covid‐19 infection can promote immune dysregulation, characterised by high levels of pro‐and anti‐inflammatory cytokines and chemokines. 97 , 98 Dysregulated immune response may exhibit as severe lymphopenia 99 with hyperactivated pro‐inflammatory T‐cells and decreased regulatory T‐cells, mostly in critically ill patients. 100 In contrast, no decrease of B‐cells has been seen in Covid‐19 patients. 99 Cytokines, the main indication of hyper‐inflammation in Covid‐19 patients, are a group of immunoregulatory cell‐cell communication molecules, including chemokines, interleukins, lymphokines, monokines, and interferons. 101 Chemokines such as CXCL8 and CXCL10 act as chemotaxis cytokines and bring leucocytes to the site of concern. 102 , 103 Additionally, the production of type I interferons is the fastest and first response of infected cells to slow down or stop viral replication and alert the presence of the pathogen to immune cells. 74 , 102 Although cytokines are essential for combating viral infections, overexpression and elevated levels of inflammatory cytokines, known as cytokine storm, could lead to immune cell infiltration to different organs, which subsequently causes multiple organ damage such as acute respiratory distress syndrome (ARDS) and CNS dysfunction or even death. 100 , 101 , 104 , 105 , 106 , 107 According to obtained data, the majority of severe Covid‐19 patients have exhibited a significant increase in pro‐and anti‐inflammatory cytokines, including IL‐2, IL‐6, IL‐7, IL‐1β, tumour necrosis factor‐α (TNF‐α), IFN‐γ, interferon‐gamma‐induced protein‐10 (IP‐10), monocyte chemoattractant protein‐1 (MCP‐1), granulocyte colony‐stimulating factor (G‐CSF), 108 , 109 , 110 , 111 , 112 and other molecules and inflammatory markers including paracalcitonin, alanine aminotransferase (ALT), lactate dehydrogenase (LDH), D‐dimer, high‐sensitivity C‐reactive protein (hsCRP), and ferritin 98 , 108 which are associated with Covid‐19 severity. 98 , 100 , 112 , 113 Interestingly, Th2 cell‐secreted cytokines such as IL‐4 and IL‐10 have also been elevated in Covid‐19 patients, which take part in inhibiting the inflammatory response. 112 Complement activation also plays a critical role in the disease severity of SARS‐CoV‐2 by promoting immune cell activation and pro‐inflammatory states. In the same way, Increased plasma complement levels were noted in moderate and severe Covid‐19 patients, making them susceptible to complement‐mediated injuries. 114 , 115
The generation of IFNs and other cytokines is mediated through several pattern‐recognition receptors (PRRs), including TLRs and NOD‐like receptors (NLRs), which are expressed in monocytes, neutrophils, macrophages, and dendritic cells. 116 PRRs detect pathogen‐associated molecular patterns (PAMPs) and damage‐associated molecular patterns (DAMPs), including viral components such as RNA and molecular complexes from damaged cells. 76 As an example, TLR3 expressed on ECs recognises viral RNA and consequently increases the release of IFNs. 74 Besides, by activating the PRRs, formation of inflammasomes is promoted, and procaspase‐1 converts to caspase‐1, leading to converting pro‐IL‐1β to the active IL‐1β. 106 The triggered signalling process leads to the expression or activation of nuclear factor kappa‐light‐chain‐enhancer of activated B cells (NF‐κB) as well as activation of interferon regulatory factors that mediate the type I interferon‐dependent anti‐viral response, which involves in the activation of innate immunity. 106 , 113 , 117 SARS‐CoV‐2 infection may also induce a massive release of ATP in the alveolar microenvironment that ATPs can act as DAMP and can activate P2X7R and NLRP3, which eventually results in the progression of inflammatory response and IL‐1β and IL‐18 release. 97 , 118 P2X7 receptors are widely expressed in immune, lung, and CNS cells, mainly in microglia and oligodendrocytes, and play a key role in inflammation. 119 NLRP3 inflammasome is an essential cause of activation of the innate immune response, 113 and unlike other PRRs, NLRP3 can react to other signals such as K+ efflux, production of reactive oxygen species (ROS), and Ca2+ mobilisation. 120 For instance, P2X7R activation triggers K+ efflux, which then stimulates the NLRP3 inflammasome and cytokine release. 120 Hence, upregulation of transcription of NLRP3 genes may help the recognition of PAMPs and DAMPs that may be induced by activation of purine sensing receptors such as P2X7R and results in cytokine release. 113 , 119
Another involved pathway in the activation of cytokines such as type I IFNs after viral infection is the Janus kinase‐signal transducer and activator of transcription (JAK‐STAT) signalling pathway. 121 This pathway mediates biologic activity for a large number of inflammatory cytokines, which demonstrates JAK‐STAT activation contribution to critical events such as inflammation and the development of the immune system, which may lead to facilitating the invasion of SARS‐CoV‐2 into the CNS. 104 , 122 Since multiple pathways control cytokine activation, 101 the inhibition of involved pathways looks to be a promising strategy to balance the immune response. 123 However, prolonged inhibition of plausible pathways associated with cytokine release or activation may lead to a compromised anti‐viral immune response, which could subsequently promote the proliferation of the SARS‐CoV‐2. 104 , 106 Therefore, clinical trials are in progress focussing on inhibiting associated inflammatory molecules or receptors and pathways to overcome the hyper‐inflammation and prevent harmful effects arising from the hyper‐inflammatory response of SARS‐CoV‐2 infection (Table 1).
TABLE 1.
Clinical trials associated with Covid‐19 sequels and further neurological complications
| Trial identification | Drug used | Delivery route | Drug description | Phase | Status | |
|---|---|---|---|---|---|---|
| Inflammatory‐ and Autoinflammatory‐related trials | NCT04334044 | Ruxolitinib | Orally | Inhibits JAK1/2 and decreases IL‐6 production by macrophages | 1 and 2 | Completed |
| NCT04362137 | Ruxolitinib | Orally | 3 | Completed | ||
| NCT04348071 | Ruxolitinib | Orally | 2 and 3 | Withdrawn | ||
| NCT04354714 | Ruxolitinib | Orally | 2 | Withdrawn | ||
| NCT04377620 | Ruxolitinib | Orally | 3 | Terminated | ||
| NCT04338958 | Ruxolitinib | Orally | 2 | Completed | ||
| NCT04891133 | Baricitinib | Orally | Inhibits JAK1 and JAK2 which leads to dampen inflammatory immune responses | 2 and 3 | Recruiting | |
| NCT04320277 | Baricitinib | Orally | 2 and 3 | Not yet recruiting | ||
| NCT04321993 | Baricitinib | Orally | 2 | Recruiting | ||
| NCT04393051 | Baricitinib | Orally | 2 | Not yet recruiting | ||
| NCT04421027 | Baricitinib | Orally | 3 | Completed | ||
| NCT04358614 | Baricitinib | Orally | 2 and 3 | Completed | ||
| NCT04340232 | Baricitinib | Orally | 2 and 3 | Withdrawn | ||
| NCT04373044 | Baricitinib + Hydroxychloroquine | Orally | Hydroxychloroquine: reducing activated T cells and the production of cytokines by lymphocytes | 2 | Terminated | |
| NCT04412772 | Tocilizumab | Intravenously | Blocks the IL‐6 signalling pathways | 3 | Recruiting | |
| NCT04730323 | Tocilizumab | Intravenously | 4 | Completed | ||
| NCT04445272 | Tocilizumab | Intravenously | 2 | Completed | ||
| NCT04377750 | Tocilizumab | Intravenously | 4 | Recruiting | ||
| NCT04643678 | Anakinra | Subcutanous injection | A recombinant IL‐1 receptor antagonist | 2 and 3 | Recruiting | |
| NCT04603742 | Anakinra | Intravenously | 2 | Not yet recruiting | ||
| NCT04510493 | Canakinumab | Intravenously | Anti‐IL‐1β monoclonal antibody which leads to neutralisation of IL‐1β signalling | 3 | Completed | |
| NCT04362813 | Canakinumab | Intravenously | 3 | Completed | ||
| NCT04393311 | Ulinastatin | Intravenously | A serine protease inhibitor | 1 and 2 | Not yet recruiting | |
| NCT04795583 | Prednisone | Orally | An immunomodulatory drug | 3 | Not yet recruiting | |
| NCT04355247 | MethylPREDNISolone | Intravenously | A potent anti‐inflammatory drug | 2 | Recruiting | |
| NCT04329650 | Siltuximab and Methylprednisolone | Intravenously | Siltuximab: IL‐6 inhibitor | 2 | Recruiting | |
| NCT04355637 | Budesonide | Inhalation | Anti‐inflammatory effects in the lungs, reducing expression of ACE‐2 and TMPRSS2 | 4 | Recruiting | |
| NCT04381364 | Ciclesonide | Inhalation | Inhibits the replication of SARS‐CoV‐2 genomic RNA by targeting the viral endonuclease NSP15 | 2 | Recruiting | |
| NCT04412252 | Tofacitinib | Orally | Inhibits JAK1 and JAK3 | 2 | Withdrawn | |
| NCT04415151 | Tofacitinib | Orally | 2 | Recruiting | ||
| NCT04280588 | Fingolimod | Orally | Used for immune therapy in patients with multiple sclerosis | 2 | Withdrawn | |
| NCT04532372 | Leflunomide | Orally | An immunosuppressive drug used for rheumatoid arthritis(RA) | 1 and 2 | Recruiting | |
| NCT04869358 | Ofatumumab | Subcutaneously | A recombinant human monoclonal antibody to CD20 of B lymphocytes | 4 | Recruiting | |
| NCT04878211 | Ofatumumab | Subcutaneously | 4 | Recruiting | ||
| NCT04346797 | Eculizumab | Intravenously | A monoclonal antibody against C5 which blocks the generation of pro‐inflammatory molecules | 2 | Recruiting | |
| NCT04802083 | Eculizumab | Intravenously | Unknown | Available | ||
| NCT04891172 | Intravenous immunoglobulin | Intravenously | Pooled polyclonal serum IgG from healthy donors | 2 and 3 | Recruiting | |
| NCT04548557 | Intravenous immunoglobulin | Intravenously | 3 | Not yet recruiting | ||
| Hypoxia‐related trials | NCT04359862 | Propofol and Sevoflurane | Sevoflurane: Inhalation Propofol: Intravenously | Anaesthetics with probable neuroprotective effects | 4 | Terminated |
| NCT04771000 | Ambrisentan | Orally | A selective endothelin type A receptor antagonist | 2 | Recruiting | |
| NCT04356937 | Tocilizumab | Intravenously | Blocks the IL‐6 signalling pathways | 3 | Completed | |
| Coagulation‐related trials | NCT04743011 | Heparin sodium | Inhalation | Anticoagulant | 1 and 2 | Not yet recruiting |
| NCT04723563 | Heparin | Inhalation | 4 | Completed | ||
| NCT04427098 | Enoxaparin | Subcutaneously injection | 2 | Recruiting | ||
| NCT04492254 | Enoxaparin | Subcutaneously injection | 3 | Recruiting | ||
| NCT04646655 | Enoxaparin | Subcutaneously injection | 3 | Recruiting | ||
| NCT04354155 | Enoxaparin | Subcutaneously injection | 2 | Completed | ||
| NCT04408235 | Enoxaparin | Subcutaneously injection | 3 | Not yet recruiting | ||
| NCT04360824 | Enoxaparin | Subcutaneously injection | 4 | Recruiting | ||
| NCT04530578 | Nebulized Heparin + Enoxaparin | Nebulized Heparin: inhalation, Enoxaparin: Subcutaneously | 4 | Recruiting | ||
| NCT04355026 | Hydroxychloroquine + Bromhexine | Orally | Hydroxychloroquine: reducing activated T cells and the production of cytokines by lymphocytes, Bromhexine: TMPRSS2 inhibitor | 4 | Recruiting | |
| NCT04332666 | Angiotensin 1–7 | Intravenously | Vasodilation effect and increases endothelial function and inhibits Angiotensin II‐induced signalling | 2 and 3 | Not yet recruiting | |
| NCT04328012 | Losartan | Orally | Blocks the AT1 receptor and may be protective in stroke | 2 and 3 | Recruiting | |
| NCT04312009 | Losartan | Orally | 2 | Completed | ||
| NCT04340557 | Losartan | Orally | 4 | Completed | ||
| NCT04643691 | Losartan and Spironolactone | Orally | Losartan: blocks the AT1 receptor and may be protective in stroke, Spironolactone: a competitive aldosterone antagonist that may provide protection from SARS‐CoV‐2 | 2 | Recruiting | |
| NCT04899232 | Antithrombin III | Intravenously | An anticoagulant with anti‐inflammatory properties | 2 | Recruiting | |
| NCT04466670 | Acetylsalicylic acid (aspirin) | Orally | Inhibits platelet aggregation triggered by the release of arachidonic acid (AA) from platelet cells | 2 | Recruiting | |
| NCT04363840 | Acetylsalicylic acid (aspirin) | Orally | 2 | Not yet recruiting | ||
| NCT04424901 | Dipyridamole | Orally | Antiplatelet drug | 2 | Recruiting | |
| NCT04391179 | Dipyridamole | Orally | 2 | Completed | ||
| NCT04410328 | Dipyridamole and Aspirin | Orally | Dipyridamole: antiplatelet drug, Aspirin: inhibits platelet aggregation triggered by the release of arachidonic acid (AA) from platelet cells | 3 | Recruiting | |
| NCT04570397 | Ravulizumab | Intravenously | Inhibits C5 complement | 3 | Recruiting | |
| NCT04369469 | Ravulizumab | Intravenously | 3 | Active, not recruiting | ||
| NCT04390464 | Ravulizumab and Baricitinib | Ravulizumab: Intravenously, Bariticinib: Orally | Ravulizumab: a complement C5 inhibitor, Baricitinib: Inhibitor of the Janus kinases JAK1 and JAK2 which leads to dampen inflammatory immune responses | 4 | Recruiting | |
| NCT04333420 | IFX‐1 | Unknown | A monoclonal antibody that blocks the effect of C5a | 2 and 3 | Recruiting | |
| NCT04371367 | Avdoralimab | Intravenously | An IgG1‐κ anti‐C5aR1 blocking antibody | 2 | Completed | |
| NCT05010876 | C1 inhibitor | Slow infusion | Inhibits lectin pathway of complement | 2 | Completed |
Abbreviations: ACE‐2, angiotensin converting enzyme 2; Covid‐19, coronavirus disease 2019; SARS‐CoV‐2, severe acute respiratory syndrome coronavirus 2; TMPRSS2, Transmembrane Serine Protease 2.
5. THE INDIRECT EFFECTS ON THE NERVOUS SYSTEM
5.1. Blood‐brain barrier disruption
Segregating the CNS from peripheral blood circulation, the BBB is a highly selective barrier formed by unique ECs and supporting cellular and non‐cellular elements, including astrocytes, pericytes, and extracellular matrix (ECM). As a part of the ‘neurovascular unit’ (NVU), the highly coordinated activity of BBB components results in tight control of molecules and ions passage, precise delivery of oxygen and nutrients according to tissue needs, and protection of the CNS from toxins and pathogens. 124 , 125 The balanced permeability of BBB is crucial for the maintenance of an environment that neurons could properly function in 126 ; however, emerging evidence suggests that SARS‐CoV‐2 infection has the potency to disturb the integrity of BBB and induce hyperpermeability in the barrier. A recent study in a BBB‐on‐a‐chip in vitro system demonstrated that the SARS‐CoV‐2 spike protein could cause dysfunction and loss of integrity of the BBB. 127 In line with this, a case study of 31 Covid‐19 patients with neurological manifestations reported that 58% of patients exhibited signs of BBB disruption and leakage. 128 Considering the neurotropism characteristic of previous CoVs 73 , 129 , 130 and emerging reports of neurological manifestations such as encephalopathy/encephalitis, acute disseminated encephalomyelitis, seizures, impaired consciousness, and delirium in Covid‐19 patients, 6 , 17 , 18 , 19 , 131 , 132 it is feasible to attribute these neurological complications, at least partly, to BBB impairment followed by the infection; however, it is not precisely clear whether the initial damage to the barrier is due to the direct invasion of SARS‐CoV‐2 to cellular structures of the BBB, or a response of barrier components to exacerbated inflammatory state associated with Covid‐19. 45 , 39
As the core anatomical elements of the BBB, ECs in the brain are uniquely specialised in structure and function. Continuous intercellular tight junctions (TJs), lack of common fenestrations, and suppressed transcytosis are distinguishing characteristics of these cells, compared to ECs in other tissues, making them capable of limiting both the paracellular and transcellular passage of molecules through the neurovascular endothelium. 89 , 124 However, barrier properties of brain ECs could be altered directly or/and indirectly by the virus infection. Because of the fact that expression of ACE2 receptor and NRP1 has been observed in human brain microvascular ECs, 77 , 133 and the presence of SARS‐CoV‐2 particles in capillary ECs of the brain is reported in an autopsy study, 80 the direct effect of SARS‐CoV‐2 on ECs could be proposed as a possible route of damage to the BBB (Figure 3a); however, the indirect effect of the hyperinflammatory state is the most likely culprit of disruption of the BBB associated with Covid‐19. 45 Elevation in levels of pro‐inflammatory factors is strongly related to alteration in TJ function and BBB disruption. For instance, in a rat study, an increased level of IL‐1 has been indicated as a causative factor for meningitis and compromised BBB integrity. 134 Another study suggested that IL‐1β induces discontinuous distribution of claudin‐5, one of the TJ proteins, along the plasma membrane of brain ECs. 135 Moreover, cytokines and chemokines such as TNF‐α, IL‐6, IL‐12, CCL2, and cxcl10 are demonstrated to cause distribution of TJ proteins (occludin, claudin‐5, ZO‐1, and ZO‐2), modulation in the function of BBB transporters like P‐glycoprotein, and alteration of adsorptive transcytosis properties, all resulting in compromised BBB permeability. 84 , 136
FIGURE 3.

Potential mechanisms of blood‐brain barrier (BBB) disruption by severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2) infection. (a) Dissemination of SARS‐CoV‐2 into the blood circulation leads to the interaction of viral particles with ACE2, basigin, or Neuropilin 1 receptors expressed by brain endothelial cells (ECs). SARS‐CoV‐2 could also infect leucocytes; (b) by facilitating entry receptors, SARS‐CoV‐2 infects the brain ECs and promotes activation of these cells. Moreover, due to systemic inflammation associated with the infection, ECs exposure to circulating cytokines also activates these cells. ECs activation induces upregulated expression of vascular and intercellular adhesion molecules (VCAM and ICAM) and matrix metalloproteinases (MMPs). Interaction of leucocyte surface β1 and β2 integrins with adhesion molecules results in the binding of circulating leucocytes to the ECs and facilitates extravasation of leucocytes through the tight junctions and basement membrane that are already degraded by the action of MMPs. In this manner, infiltration of infected leucocytes by the ‘Trojan horse’ mechanism facilitates viral entry to the brain parenchyma; (c) infection of the brain ECs and hyperinflammatory state associated with Covid‐19 induce apoptosis of ECs, leading to the disruption of the BBB. The compromised barrier allows extravasation of erythrocytes and leucocytes, leakage of plasma pro‐inflammatory agents such as cytokines, and free passage of circulating SARS‐CoV‐2 particles to the brain parenchyma. The presence of viral particles and pro‐inflammatory factors, as well as infiltrated leucocytes in cerebral tissue, triggers activation of astrocytes and microglia, which in turn causes further release of cytokines in the brain parenchyma, phagocytic hyperactivity of microglia, and disruption of astrocytes end feet, all results in more damage to the BBB and nervous tissue
Under the influence of systemic inflammation, activation of ECs by cytokines such as IL‐6, IFN‐γ, and TNF‐α triggers the overexpression of different proteases, including matrix metalloproteinases (MMPs). 136 , 45 MMPs are critical contributors to BBB disruption by digesting TJs and basement membrane proteins associated with ECs. 137 Numerous studies have investigated the deleterious effects of dysregulated MMPs, indicating their pivotal role in CNS pathologies, such as cerebral oedema, leucocyte infiltration, haemorrhage, and exacerbated inflammatory reactions. 134 On another note, pro‐inflammatory cytokines such as TNF‐α and IL‐1β have been shown to induce cyclooxygenase (COX) activity in several cell types, including brain ECs. 84 , 134 , 138 COX activation triggers the production of eicosanoids, including prostaglandins (PGs) and leukotrienes (LTs), which in turn affect various pathways and tissues. Studies have indicated that COX2‐mediated PGs induce the expression of MMP1, leading to further alteration of TJs and damage to BBB. 139 Furthermore, prostaglandin E (PGE) production in brain ECs by COX1 or COX2 plays a vital role in exhibiting symptoms such as fever and malaise/discomfort through the hypothalamic‐pituitary‐adrenal (HPA) axis activation. 140 Thus, it is tempting to speculate that PG release in response to systemic inflammation is an overlooked inducer of sickness behaviour related to Covid‐19 disease.
Promoted by systemic inflammation, activation of brain ECs triggers upregulated expression of the vascular and the intercellular adhesion molecules (VCAM and ICAM), which mediate immune cell infiltration into the brain parenchyma via interaction with β1 and β2 integrins expressed on the surface of leucocytes 45 , 134 (Figure 3b). In parallel with inflammatory mediated damage to BBB, increased extravasation of immune cells across the BBB leads to a higher presence of viral particles (by the ‘Trojan horse’ mechanism) and pro‐inflammatory cytokines and chemokines in the brain parenchyma, where they encounter the CNS defence system represented by astrocytes and microglia. Astrocytes exposure to viral particles and pro‐inflammatory mediators triggers activation and subsequent upregulated production of pro‐inflammatory factors by these cells, as well as vascular endothelial growth factor (VEGF). 140 , 141 This ultimately results in astroglial death and disruption of astrocytic end‐feet, the structural components of the BBB that form the outer layer of the mature capillaries 125 , 141 (Figure 3c). Moreover, secretion of VEGF‐A by activated astrocytes stimulates the endothelial nitric oxide synthase (eNOS) signalling in ECs and downregulates the expression of TJ proteins such as occludin and claudin‐5. 136 In parallel with this, sonic hedgehog production of astrocytes is shown to be suppressed by IL‐1β. This also results in further disruption of BBB integrity; as the sonic hedgehog signalling pathway plays a key role in upregulated expression of TJ proteins in brain ECs. 142 Similar to astrocytes, activation of microglia in an infectious/inflammatory condition is followed by an exacerbating response of overproduction of pro‐inflammatory mediators such as cytokines, chemokines, matrix proteases, PGs, nitric oxide, and ROS 143 (Figure 3c). Besides the impairment of the BBB permeability in a vicious circle led by pro‐inflammatory factors, exaggerated response of microglia is associated with a phagocytic hyperactivity characteristic, inducing neurodegeneration, synaptic loss, and demyelination in the CNS tissue. 141 These findings are in line with the evidence derived from post‐mortem case series, such as a study of 43 Covid‐19 patients, in which substantial astrogliosis, microglial activation, and cytotoxic T lymphocytes infiltration were prevalent in the brain specimens; and increased phagocytic activity of microglia was indicated by detection of overexpressed lysosomal marker CD68. 144 Additionally, another study of highly multiplexed spatial analysis of CNS tissue has also identified profound immune activation in Covid‐19 brains, accompanied by significant microglial alterations, parenchymal CD8 infiltration, and formation of microglial nodules, hotspots of microglia‐T‐cell interactions. Prominent perivascular leakage and considerable axonal damage were observed in this study to be tied with the broad neuroinflammation, indicating the association of immune activation with BBB disruption and neurodegeneration in the exhibition of neurological manifestations of Covid‐19. 145
5.2. Hypoxic associated CNS dysfunction
Hypoxia is a major stress factor that induces BBB disruption, leading to infiltration of peripheral immune cells and leakage of blood proteins, including cytokines, to the brain. 96 Due to the infection of SARS‐CoV‐2 in different organs, respiratory and circulatory failure can cause moderate to severe levels of hypoxia. Hypoxaemia (low level of oxygen with no sensation of dyspnoea) caused by alveolar damage and inflammatory exudate can lead to intrapulmonary shunting, loss of lung perfusion regulation, intravascular microthrombi, and impaired diffusion capacity. 146 , 147 Affected cells/tissues must respond to the hypoxic condition to sustain their function and prevent cell death. Hypoxia‐inducible factors (HIFs) are the most critical responses among different pathways and reactions of affected cells/tissues. 148 HIFs are heterodimeric transcription factors that possess two subunits: an oxygen‐regulated alpha subunit and an oxygen‐independent beta subunit, 149 and act as central regulators of tissue O2 metabolism and are known as master regulators of oxygen homoeostasis. 150 , 151 SARS‐CoV‐2 infection induces upregulated expression of HIF‐1α in immune cells, which results in further release of cytokines and causes ARDS. Pro‐inflammatory cytokines such as IL‐6 and TNF‐α can reduce Zonula occludens‐1 (ZO‐1) mRNA levels and increase the phosphorylation of ZO‐1 protein, which results in impairing BBB integrity. On the other hand, HIF‐1α stabilisation in microvascular ECs increases the transcription of VEGF and integrins, resulting in increased vascular permeability. 152 , 153
Based on the results of a study, VEGF enhances gap formation between ECs and induces fenestration in unfenestrated human and porcine endothelial monolayers in vitro. 154 Interestingly, a later study indicated that hypoxia can downregulate the expression of ZO‐1, increase the expression of HIF‐1α and VEGF and upregulate the phosphorylation of ZO‐1, which all together can disrupt the BBB integrity and facilitate the invasion of SARS‐CoV‐2 into the CNS tissue. Since HIF‐1α stabilisation is directly linked to barrier disruption, It is claimed that inhibition of HIF‐1α improves barrier stability and decreases BBB damages, and prevents or reduces further CNS dysfunctions caused by SARS‐CoV‐2. 148 Nevertheless, the function of the HIF‐1α inhibitor and VEGF antibody has been investigated, and according to evidence and documents, the expression of ZO‐1 has been increased by inhibiting HIF‐1α and VEGF. 96 It is noteworthy that HIF‐1α also activates miR‐let‐7b, which inhibits protein expression of ACE2 and subsequently by stimulation of ADAM17 and inhibition of TMPRSS2 takes part in decreasing SARS‐CoV‐2 entry to the cells. 152 Remarkably, in different intensities of Covid‐19 patients, there is a high probability of survival in patients with spO2 values greater than 90% with oxygen supplementation. 155 Given that hypoxaemia/hypoxia is the marker of severity, 156 and patients may be at high mortality risk, it has been speculated that in patients with spO2 values less than 90%, despite oxygen supplementation, maximum supportive care with more drug and other therapies is needed. 155 Hence, seldom drug investigations associated with hypoxic conditions are being trialed to improve the status of Covid‐19 patients with hypoxia (Table 1).
5.3. Hypercoagulable state
Several studies have reported coagulation abnormalities and thrombotic complications as common manifestations in patients with Covid‐19. 157 , 158 , 159 Presented with elevated prothrombin time (PT) and D‐dimer (coagulation function–related indicators), 160 , 161 the hypercoagulable state of Covid‐19 predisposes patients to thrombotic vascular events, including disseminated pulmonary microthrombi, venous thromboembolism, and brain strokes. 34 , 162 In a case series of Covid‐19 patients in China, elevated D‐dimer levels have been observed in 46.4% of 560 patients, while levels were even higher in severe cases of the disease (59.6%). 163 In another study of 288 patients, thromboembolic events and acute ischaemic strokes have been reported in 7.7% and 2.5% of patients, respectively. 164 Other case series have reported the rate of stroke incidence ranging from 1% to 3% in admitted patients and up to 6% in critically ill patients. 76 Moreover, a twofold higher risk of cryptogenic stroke has been reported in Covid‐19 patients, as the incidence observed in more than 65.6% of 3556 hospitalised cases, compared to 30.4% in contemporary controls. 165 According to these clinical reports and data from previous coronavirus outbreaks, 30 hypercoagulopathy is considered a life‐threatening aspect of Covid‐19 pathogenesis, especially among patients with hypertension, diabetes, and other cardiovascular comorbidities. 166
Given the complexity and multifactor dependence of the mechanism, the aetiology of hypercoagulopathy in Covid‐19 is not precisely explained. Downregulation of ACE2 by SARS‐CoV‐2 and subsequent Ang II accumulation, 167 pneumonia‐induced hypoxia, 168 and release of neutrophil extracellular traps (NETs) 169 are among proposed mechanisms for the condition. However, endotheliopathy and massive inflammatory response have been indicated as two main features of prothrombotic presentations associated with Covid‐19. Resting endothelium maintains vascular homoeostasis and prevents thrombosis through the production of several anti‐inflammatory and antithrombotic factors. Hence, the probable direct viral infection of ECs by SARS‐CoV2 and the independent response of ECs to the systemic inflammation phase of the disease are the major contributors to the endothelial dysfunction and subsequent coagulopathy associated with Covid‐19. 166 Due to the susceptibility of lung and brain ECs to SARS‐CoV‐2 infection, 77 , 80 it is plausible to suggest that the Covid‐19‐associated thrombosis is likely to be started in respiratory vascular tissue and then spreads into other organs, including the nervous system, through the circulation of viral particles and inflammatory agents. 170
Anticoagulant and anti‐inflammatory properties of intact vascular endothelium are massively inhibited by a viral infection and following vigorous inflammatory response. Studies have reported a down‐regulated expression of ‘tissue factor pathway inhibitor’ (tissue factor pathway inhibitor (TFPI)) and ‘thrombomodulin’ (THBD), two anticoagulatory factors, in virus‐infected ECs. 171 , 172 Other studies have suggested the impairment of thrombin generation control mechanisms such as antithrombin III, TFPI, and protein C system during inflammation by pro‐inflammatory cytokines. 173 On the other hand, overproduction of cytokines and chemokines such as IL‐6, IL‐1β and, TNF, in synergy with a direct viral infection, induces activation of ECs and promotes further secretion of pro‐inflammatory cytokines and pro‐thrombotic factors, amplifying the vicious cycle of endothelial damage and vessel thrombosis. 174
Complement activation is another aspect of the Covid‐19‐associated hypercoagulable inflammatory state. Studies have reported evident signs of complement hyperactivity in infected patients, indicated by the evaluation of soluble markers and histopathological observations. 114 It is plausible that SARS‐CoV‐2 infection induces three different pathways of complement activation, all converging in a common cascade and leading to the production of various molecules such as anaphylatoxins. These complement components are potent activators of inflammation and coagulation mechanisms, playing an essential role in the innate immune response against viral infections. However, dysregulated function of the complement system could end in thrombotic complications. For instance, anaphylatoxin ‘C5a’ promotes the release of ‘tissue factor’ from multiple sources, including ECs and neutrophils, which in turn activates another molecular cascade ending in thrombin production and clot formation. Furthermore, C5a impairs fibrinolysis by inhibiting the plasminogen/plasmin system and stimulates neutrophils to release excessive NETs, all resulting in a higher coagulable condition. 175 Taken together, infection‐triggered complement hyperactivation induces a maladaptive inflammatory and coagulatory response, which in turn, feeds back and amplifies complement activation and clot formation. 176
Considering all the above, patients with Covid‐19 are more likely to exhibit thrombotic events in multiple organs, including brain and cerebral circulation. 177 , 178 The covid‐19 infection has been described as a risk factor for stroke. 179 Therefore, besides ongoing anticoagulant and antiplatelet trials, administration of other possible therapeutic agents such as complement inhibitors and anti‐inflammatory agents are under investigation, as they could be beneficial in targeting multiple steps of coagulation‐related pathways and developing a combination therapy strategy with much more efficiency 166 (Table 1).
5.4. Autoimmune neuropathies
Based on various case reports of various autoimmune neuropathies associated with Covid‐19, it is considered that SARS‐CoV‐2 can also possess auto‐immunogenic effects mainly via molecular mimicry or other mechanisms. 180 , 181 , 182 , 183 Losing Immunologic tolerance to key antigenic sites on the different parts of neurons can lead to autoimmune peripheral neuropathies. 184 In other words, a potential trigger of multi‐organ autoimmunity in Covid‐19 could be the molecular mimicry between SARS‐CoV‐2 proteins and various human cell/tissue autoantigens, including the nervous system, which is involved in inflammatory polyneuropathies by analysing the peptide sharing between the virus and such protein antigens with BLAST (basic local alignment search tool). 185 , 186 Different parts of PNS, including the dorsal motor nucleus, nucleus ambiguous, nodose ganglion, and jugular ganglion, are potentially able to generate an autoimmune response due to having neurons presenting proteins with similar epitopes with SARS‐CoV‐2 proteins. 187 Moreover, the occurrence of autoimmunity caused by SARS‐CoV‐2 has been demonstrated in CNS. Multiple pathways of Covid‐19 initiated autoimmune cascade are shown in Figure 4. 188
FIGURE 4.

Multiple pathways of Covid‐19 initiated autoimmune cascade, which may result in neurodegenerative disease severity in post‐Covid‐19 patients in coming decades. The cross‐reactive response caused by the molecular mimicry of pathogen antigens to self‐antigens, activated lymphocytes, and memory of B lymphocytes against self‐antigens may lead to autoimmune response due to the interaction of antibodies with self‐epitopes. Besides, initiation of central nervous system (CNS) self‐tissue damage by the production of self‐antigens similar to viral antigens in the structure and function of antigen presenting cells (APCs) and stimulation of T‐cells by additional self‐epitopes may be due to the cytokine storm and leads to an autoimmune response and further neurodegenerative complications. On the other hand, neurotoxic pro‐inflammatory cytokines may have harmful effects on CNS cellular organelles such as mitochondria and lysosomes, which could be an initial point of demyelination, blood‐brain barrier disintegration, and other neurodegenerative processes. SARS‐CoV‐2, severe acute respiratory syndrome coronavirus 2
Several case studies have reported Guillain‐Barre syndrome (GBS) and its variants 189 in Covid‐19 patients. 183 , 190 , 191 , 192 , 193 GBS is not usually known as a form of Covid‐19 presentation, but as stated by a study by fragile, et al., the frequency of GBS is higher among patients with Covid‐19. 194 In addition, an increased incidence of GBS has been seen during the pandemic. Given that the majority of GBS patients are Covid‐19‐positive, there could be a pathogenic link between Covid‐19 and GBS. 195 GBS is an immune‐mediated disorder in which gangliosides, molecular markers expressed on peripheral nerves, 196 are attacked by the immune response generated by SARS‐CoV‐2, due to molecular mimicry. 197 Various gangliosides such as GD1a, GD1b, GQ1b, GT1b, GM1, and GM2 participate in patients with GBS neuropathies and play a key role in the pathophysiology of GBS. 196 In Covid‐19‐positive GBS patients, expression of antibodies against these gangliosides has been reported. 198 IgG, IgM, and membranolytic attack complex could imply complement‐fixing antibodies against myelinated fibres. Likewise, complement‐fixing IgM antibodies against a peripheral nerve glycolipid that contains carbohydrate epitopes and various sulfated or acidic glycosphingolipids have been detected in the serum of GBS patients. 184 Additionally, animal studies demonstrate that some anti‐ganglioside antibodies can cause blockade of nerve transmission and destruction of nerve terminal or may affect different membrane channels of neurons due to complement activation and formation of the membrane attack complex. 199
Different trials concerning inhibiting the neurotoxic effects of antibodies have indicated that there is effective immunotherapy with Intravenous immunoglobulin (IVIg) in Covid‐19 patients for treating autoimmune and inflammatory diseases as well as GBS 198 , 199 , 200 , 201 (Table 1). IVIg consists of accumulated human IgG purified from healthy donors and could improve GBS patients' status by complement scavenging, neutralisation, or enhancement of degradation of auto‐antibodies, inhibition of activation of various innate immune cells, increasing the number of plasmablasts, and other plausible mechanisms. 200 , 202 , 203 Moreover, transient axonal glycoprotein‐1 (TAG‐1) and the expression of inhibitory FcᵧRIIB receptors on immune B cells, participate in responsiveness to IVIg treatment. Since TAG‐1 polymorphism is associated with IVIg responsiveness, response to IVIg can be genetically determined. 184 Besides the above, other GBS treatments are needed to be trialed and approved to improve Covid‐19 patients suffering GBS, by inhibiting the autoantibodies caused by viral infection.
Covid‐19 infection in Patients with pre‐existing impaired regulation of immune responses such as Multiple Sclerosis (MS) may potentially trigger a further amplification of immune responses. 204 Thus, MS patients may exhibit more acute neurologic symptoms during Covid‐19 infection. 205 The relationship between Covid‐19 and MS is complicated, and there is not enough immunological and physiological evidence regarding Covid‐19 implications in MS‐related neurodegeneration. 204 Nevertheless, it is frequently claimed that immunocompromised patients or patients receiving immunosuppressive treatments may be at increased risk of SARS‐CoV‐2 infection due to the impairment in the immune system caused by high‐efficacy disease‐modifying therapies (DMTs) 204 , 206 , 207 , 208 , 209 ; or may experience a more severe course of Covid‐19 compared with general population due to the limited immune response and subsequently allowing more significant viral replication. 204 , 210 So, it is suggested that cell‐depleting DMTs would be associated with higher Covid‐19 risk. 211 Other factors of MS patients such as age, sex, worse physical disability, and comorbidities can also increase the risk of infection and hospitalisation in MS patients with Covid‐19. 204 , 207
Ocrelizumab is one of most widely used therapeutics for MS patients, 212 to treat relapsing and primary progressive phase of the disease. 213 In patients treated by Ocrelizumab, severe infections was found to be very low compared to patients who formerly used rituximab, which is commonly used in the population of MS patients and this group of patients is at the risk of higher rates of infections. 214 As claimed by a study on a case report, rituximab enhances the rate of Covid‐19 infection in MS patients 207 and decreases immunoglobulins, especially IgM. 214 While in other studies, it has been observed that there is no interdependence between specific DMTs and higher risk of Covid‐19 in MS patients, which needs more supporting investigations. Moreover, IFN‐β and glatiramer acetate are probably not related to severe infection in MS patients due to not exhibiting immunosuppressive effects. 207 It is also speculated that dimethyl fumarate (DMF) may increase the risk of Covid‐19 by reducing the lymphocyte count in patients. 215 In brief, clinical trials are essential for attaining more data regarding the protective or harmful effects of immunosuppressive agents, risk factors associated with severe Covid‐19, and antibody formation in MS or other autoimmune patients infected by SARS‐CoV‐2 216 to prevent disease activation or progression and limit the need for hospitalisation in the patients suffering autoimmune diseases 213 (Table 1).
6. CONCLLUSION
SARS‐CoV‐2 could affect the nervous system in various ways. The direct invasion of the CNS by the virus could possibly occur through the infection of peripheral nerves such as OSNs, pulmonary network, or ENS. Additionally, dissemination of viral particles and infected leucocytes from heavily involved pulmonary tissue into the systemic circulation could serve as another gateway for SARS‐CoV‐2 to invade the CNS. However, few studies have reported the presence of SARS‐CoV‐2 in CSF and brain parenchyma, which could not be indicated as consistent evidence for the direct invasion of the virus to the CNS. On the contrary, mounting evidence implicates the indirect effects of SARS‐CoV‐2 on the nervous system via exacerbating inflammation and pneumonia‐induced hypoxia as key drivers of neurological manifestations in Covid‐19. Destructive effects of infection‐associated cytokine storm and hypoxia on the BBB have been well investigated, and mechanisms by which infection could cause coagulation abnormalities and autoimmune neuropathies are partly elucidated by previous studies on other types of infection. These findings are also applicable for Covid‐19 infection as so many neurological symptoms in critically ill patients are linked to BBB disruption, thrombovascular events, and molecular mimicry‐related neuropathies.
Here we reviewed some of the molecular mechanisms by which SARS‐CoV‐2 could directly or indirectly alter the structural and functional properties of the nervous system. Currently, there is no particular treatment for neurological complications associated with Covid‐19, and most of the therapeutic efforts so far have gone into the development of effective vaccines. Despite the significant achievement, none of the developed vaccines are 100% protective against the infection. The pandemic is still a major public health issue, even in countries with a high vaccinated proportion of the population. Moreover, therapeutic approaches dependent on anti‐viral agents have not been as effective as expected in the case of Covid‐19. In this regard, further investigations are still needed to elucidate the molecular basis of the infection, which is an essential aspect of developing more effective therapeutic strategies. Several clinical trials are currently underway to evaluate the effects of anti‐inflammatory agents, anticoagulants, and immunomodulatory therapies. The results of these trials could assist in the development of a combination therapy strategy that targets multiple aspects of SARS‐CoV‐2 pathogenesis, such as respiratory insufficiency, immune dysregulation, hypercoagulopathy, and multiple organ failure. In parallel with anti‐viral therapies, targeting the deleterious side issues of Covid‐19 by this strategy could aid not only in ameliorating neurological complications but also in improving disease severity and achieving a more favourable outcome.
CONFLICT OF INTERESTS
The authors declare that there is no conflict of interest.
AUTHOR CONTRIBUTIONS
Mohammad Mahboubi Mehrabani and Mohammad Sobhan Karvandi: Writing – original draft, Reviewing and Editing. Pedram Maafi: Reviewing, and Editing the Final manuscript. Mohammad Doroudian: Conceptualization; Preparation; Writing, Reviewing, and Editing the Final manuscript.
ACKNOWLEDGEMENTS
The authors would like to express very great appreciation to Mr. Mohammad H Azhdari and Mr. Nima Goodarzi for generation of graphical abstracts and schematic figures of this study.
Mahboubi Mehrabani M, Karvandi MS, Maafi P, Doroudian M. Neurological complications associated with Covid‐19; molecular mechanisms and therapeutic approaches. Rev Med Virol. 2022;e2334. 10.1002/rmv.2334
Mohammad Mahboubi Mehrabani and Mohammad Sobhan Karvandi contributed equally.
DATA AVAILABILITY STATEMENT
Data sharing is not applicable as there is no new data were analysed in this study.
REFERENCES
- 1. Khan M, Adil SF, Alkhathlan HZ, et al. COVID‐19: a global challenge with old history, epidemiology and progress so far. Molecules. 2020;26(1). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Tsang HF, Chan LWC, Cho WCS, et al. An update on COVID‐19 pandemic: the epidemiology, pathogenesis, prevention and treatment strategies. Expert Rev Anti Infect Ther. 2021;19(7):877‐888. [DOI] [PubMed] [Google Scholar]
- 3. Harapan BN, Yoo HJ. Neurological symptoms, manifestations, and complications associated with severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2) and coronavirus disease 19 (COVID‐19). J Neurol. 2021;268(9):3059‐3071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Cucinotta D, Vanelli M. WHO declares COVID‐19 a pandemic. Acta Biomed. 2020;91(1):157‐160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Bobker SM, Robbins MS. COVID‐19 and headache: a primer for trainees. Headache. 2020;60(8):1806‐1811. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Mao L, Jin H, Wang M, et al. Neurologic manifestations of hospitalized patients with coronavirus disease 2019 in Wuhan, China. JAMA Neurol. 2020;77(6):683‐690. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Bolay H, Gül A, Baykan B. COVID‐19 is a real headache! Headache. 2020;60(7):1415‐1421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Collantes MEV, Espiritu AI, Sy MCC, Anlacan VMM, Jamora RDG. Neurological manifestations in COVID‐19 infection: a systematic review and meta‐analysis. Can J Neurol Sci. 2021;48(1):66‐76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Brouwer MC, Ascione T, Pagliano P. Neurologic aspects of covid‐19: a concise review. Infez Med. 2020;28(suppl 1):42‐45. [PubMed] [Google Scholar]
- 10. Pennisi M, Lanza G, Falzone L, Fisicaro F, Ferri R, Bella R. SARS‐CoV‐2 and the nervous system: from clinical features to molecular mechanisms. Int J Mol Sci. 2020;21(15). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Vohora D, Jain S, Tripathi M, Potschka H. COVID‐19 and seizures: is there a link? Epilepsia. 2020;61(9):1840‐1853. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Swanson PA, 2nd , McGavern DB. Viral diseases of the central nervous system. Curr Opin Virol. 2015;11:44‐54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Kremer S, Lersy F, de Sèze J, et al. Brain MRI findings in severe COVID‐19: a retrospective observational study. Radiology. 2020;297(2):E242‐E251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Moriguchi T, Harii N, Goto J, et al. A first case of meningitis/encephalitis associated with SARS‐Coronavirus‐2. Int J Infect Dis. 2020;94:55‐58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Dong M, Zhang J, Ma X, et al. ACE2, TMPRSS2 distribution and extrapulmonary organ injury in patients with COVID‐19. Biomed Pharmacother. 2020;131:110678. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Espíndola OM, Brandão CO, Gomes YCP, et al. Cerebrospinal fluid findings in neurological diseases associated with COVID‐19 and insights into mechanisms of disease development. Int J Infect Dis. 2021;102:155‐162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Garg RK, Paliwal VK, Gupta A. Encephalopathy in patients with COVID‐19: a review. J Med Virol. 2021;93(1):206‐222. [DOI] [PubMed] [Google Scholar]
- 18. Ye M, Ren Y, Lv T. Encephalitis as a clinical manifestation of COVID‐19. Brain Behav Immun. 2020;88:945‐946. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Emami A, Fadakar N, Akbari A, et al. Seizure in patients with COVID‐19. Neurol Sci. 2020;41(11):3057‐3061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Mehraeen E, Behnezhad F, Salehi MA, Noori T, Harandi H, SeyedAlinaghi S. Olfactory and gustatory dysfunctions due to the coronavirus disease (COVID‐19): a review of current evidence. Eur Arch Otorhinolaryngol. 2021;278(2):307‐312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Alberti P, Beretta S, Piatti M, et al. Guillain‐Barré syndrome related to COVID‐19 infection. Neurol Neuroimmunol Neuroinflamm. 2020;7(4). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Shabani Z. Demyelination as a result of an immune response in patients with COVID‐19. Acta Neurol Belg. 2021;121(4):859‐866. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Nalleballe K, Reddy Onteddu S, Sharma R, et al. Spectrum of neuropsychiatric manifestations in COVID‐19. Brain Behav Immun. 2020;88:71‐74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Huang C, Huang L, Wang Y, et al. 6‐month consequences of COVID‐19 in patients discharged from hospital: a cohort study. Lancet. 2021;397(10270):220‐232. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Wang Q, Zhang Y, Wu L, et al. Structural and functional basis of SARS‐CoV‐2 entry by using human ACE2. Cell. 2020;181(4):894‐904.e9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Gadanec LK, McSweeney KR, Qaradakhi T, Ali B, Zulli A, Apostolopoulos V. Can SARS‐CoV‐2 virus use multiple receptors to enter host cells? Int J Mol Sci. 2021;22(3). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Hamming I, Cooper M, Haagmans B, et al. The emerging role of ACE2 in physiology and disease. J Pathol. 2007;212(1):1‐11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Campos DMO, Fulco UL, Oliveira CBS, Oliveira JIN. SARS‐CoV‐2 virus infection: targets and antiviral pharmacological strategies. J Evid Based Med. 2020;13(4):255‐260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Mahmoud IS, Jarrar YB, Alshaer W, Ismail S. SARS‐CoV‐2 entry in host cells‐multiple targets for treatment and prevention. Biochimie. 2020;175:93‐98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Giannis D, Ziogas IA, Gianni P. Coagulation disorders in coronavirus infected patients: COVID‐19, SARS‐CoV‐1, MERS‐CoV and lessons from the past. J Clin Virol. 2020;127:104362. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Gheblawi M, Wang K, Viveiros A, et al. Angiotensin‐converting enzyme 2: SARS‐CoV‐2 receptor and regulator of the renin‐angiotensin system: celebrating the 20th anniversary of the discovery of ACE2. Circ Res. 2020;126(10):1456‐1474. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Li MY, Li L, Zhang Y, Wang X‐S. Expression of the SARS‐CoV‐2 cell receptor gene ACE2 in a wide variety of human tissues. Infect Dis Poverty. 2020;9(1):45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Zamorano Cuervo N, Grandvaux N. ACE2: evidence of role as entry receptor for SARS‐CoV‐2 and implications in comorbidities. Elife. 2020;9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Cui S, Chen S, Li X, Liu S, Wang F. Prevalence of venous thromboembolism in patients with severe novel coronavirus pneumonia. J Thromb Haemost. 2020;18(6):1421‐1424. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Dehbidi MY, Goodarzi N, Azhdari MH, Doroudian M. Mesenchymal stem cells and their derived exosomes to combat Covid–19. Rev Med Virol. n/a(n/a):e2281. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Schmidt AL, Tucker MD, Bakouny Z, et al. Association between androgen deprivation therapy and mortality among patients with prostate cancer and COVID‐19. JAMA Netw Open. 2021;4(11):e2134330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Peng R, Wu L‐A, Wang Q, Qi J, Gao GF. Cell entry by SARS‐CoV‐2. Trends Biochem Sci. 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Ibrahim IM, Abdelmalek DH, Elshahat ME, Elfiky AA. COVID‐19 spike‐host cell receptor GRP78 binding site prediction. J Infect. 2020;80(5):554‐562. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Burks SM, Rosas‐Hernandez H, Alejandro Ramirez‐Lee M, Cuevas E, Talpos JC. Can SARS‐CoV‐2 infect the central nervous system via the olfactory bulb or the blood‐brain barrier? Brain Behav Immun. 2021;95:7‐14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Cantuti‐Castelvetri L, Ojha R, Pedro LD, et al. Neuropilin‐1 facilitates SARS‐CoV‐2 cell entry and infectivity. Science. 2020;370(6518):856‐860. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Shang J, Wan Y, Luo C, et al. Cell entry mechanisms of SARS‐CoV‐2. Proc Natl Acad Sci U. S. A. 2020;117(21):11727‐11734. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Glebov OO. Understanding SARS‐CoV‐2 endocytosis for COVID‐19 drug repurposing. FEBS J. 2020;287(17):3664‐3671. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Zanganeh S, Goodarzi N, Doroudian M, Movahed E. Potential COVID‐19 therapeutic approaches targeting angiotensin‐converting enzyme 2; an updated review. Reviews in Medical Virology. n/a(n/a):e2321. [DOI] [PubMed] [Google Scholar]
- 44. Wang K, Chen W, Zhang Z, et al. CD147‐spike protein is a novel route for SARS‐CoV‐2 infection to host cells. Signal Transduct Target Ther. 2020;5(1):283. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Alquisiras‐Burgos I, Peralta‐Arrieta I, Alonso‐Palomares LA, Zacapala‐Gómez AE, Salmerón‐Bárcenas EG, Aguilera P. Neurological complications associated with the blood‐brain barrier damage induced by the inflammatory response during SARS‐CoV‐2 infection. Mol Neurobiol. 2021;58(2):520‐535. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Baig AM, Khaleeq A, Ali U, Syeda H. Evidence of the COVID‐19 virus targeting the CNS: tissue distribution, host‐virus interaction, and proposed neurotropic mechanisms. ACS Chem Neurosci. 2020;11(7):995‐998. [DOI] [PubMed] [Google Scholar]
- 47. Qiao J, Li W, Bao J, et al. The expression of SARS‐CoV‐2 receptor ACE2 and CD147, and protease TMPRSS2 in human and mouse brain cells and mouse brain tissues. Biochem Biophys Res Commun. 2020;533(4):867‐871. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Lima M, Siokas V, Aloizou A‐M, et al. Unraveling the possible routes of SARS‐COV‐2 invasion into the central nervous system. Curr Treat Options Neurol. 2020;22(11):37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. The Lancet Respiratory, M . COVID‐19 transmission‐up in the air. Lancet Respir Med. 2020;8(12):1159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Li Z, Liu T, Yang N, et al. Neurological manifestations of patients with COVID‐19: potential routes of SARS‐CoV‐2 neuroinvasion from the periphery to the brain. Front Med. 2020;14(5):533‐541. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Barrantes FJ. Central nervous system targets and routes for SARS‐CoV‐2: current views and new hypotheses. ACS Chem Neurosci. 2020;11(18):2793‐2803. [DOI] [PubMed] [Google Scholar]
- 52. Aghagoli G, Gallo Marin B, Katchur NJ, Chaves‐Sell F, Asaad WF, Murphy SA. Neurological involvement in COVID‐19 and potential mechanisms: a review. Neurocrit Care. 2021;34(3):1062‐1071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Awogbindin IO, Ben‐Azu B, Olusola BA, et al. Microglial implications in SARS‐CoV‐2 infection and COVID‐19: lessons from viral RNA neurotropism and possible relevance to Parkinson’s disease. Front Cell Neurosci. 2021;15:670298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Reza‐Zaldívar EE, Hernández‐Sapiéns MA, Minjarez B. Infection mechanism of SARS‐COV‐2 and its implication on the nervous system. Front Immunol. 2020;11:621735. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Netland J, Meyerholz DK, Moore S, Cassell M, Perlman S. Severe acute respiratory syndrome coronavirus infection causes neuronal death in the absence of encephalitis in mice transgenic for human ACE2. J Virol. 2008;82(15):7264‐7275. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Zhang AJ, Lee AC‐Y, Chu H, et al. Severe acute respiratory syndrome coronavirus 2 infects and damages the mature and immature olfactory sensory neurons of Hamsters. Clin Infect Dis. 2021;73(2):e503‐e512. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Li K, Wohlford‐Lenane C, Perlman S, et al. Middle East respiratory syndrome coronavirus causes multiple organ damage and lethal disease in mice transgenic for human dipeptidyl peptidase 4. J Infect Dis. 2016;213(5):712‐722. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Giacomelli A, Pezzati L, Conti F, et al. Self‐reported olfactory and taste disorders in patients with severe acute respiratory coronavirus 2 infection: a cross‐sectional study. Clin Infect Dis. 2020;71(15):889‐890. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Lechien JR, Chiesa‐Estomba CM, De Siati DR, 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 Otorhinolaryngol. 2020;277(8):2251‐2261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Villerabel C, Makinson A, Jaussent A, et al. Diagnostic value of patient‐reported and clinically tested olfactory dysfunction in a population screened for COVID‐19. JAMA Otolaryngol Head Neck Surg. 2021;147(3):271‐279. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Sbrana MF, Fornazieri MA, Bruni‐Cardoso A, et al. Olfactory dysfunction in frontline health care professionals during COVID‐19 pandemic in Brazil. Front Physiol. 2021;12:622987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Berth SH, Leopold PL, Morfini GN. Virus‐induced neuronal dysfunction and degeneration. Front Biosci (Landmark Ed). 2009;14:5239‐5259. [DOI] [PubMed] [Google Scholar]
- 63. Dubé M, Le Coupanec A, Wong AHM, Rini JM, Desforges M, Talbot PJ. Axonal transport enables neuron‐to‐neuron propagation of human coronavirus OC43. J Virol. 2018;92(17). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Bodnar B, Patel K, Ho W, Luo JJ, Hu W. Cellular mechanisms underlying neurological/neuropsychiatric manifestations of COVID‐19. J Med Virol. 2021;93(4):1983‐1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Vallee RB, Bloom GS. Mechanisms of fast and slow axonal transport. Annu Rev Neurosci. 1991;14(1):59‐92. [DOI] [PubMed] [Google Scholar]
- 66. Briguglio M, Bona A, Porta M, Dell'Osso B, Pregliasco FE, Banfi G. Disentangling the hypothesis of host dysosmia and SARS‐CoV‐2: the bait symptom that hides neglected neurophysiological routes. Front Physiol. 2020;11:671. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Esposito G, Pesce M, Seguella L, Sanseverino W, Lu J, Sarnelli G. Can the enteric nervous system be an alternative entrance door in SARS‐CoV2 neuroinvasion? Brain Behav Immun. 2020;87:93‐94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Xiao F, Tang M, Zheng X, Liu Y, Li X, Shan H. Evidence for gastrointestinal infection of SARS‐CoV‐2. Gastroenterology. 2020;158(6):1831‐1833. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Han C, Duan C, Zhang S, et al. Digestive symptoms in COVID‐19 patients with mild disease severity: clinical presentation, stool viral RNA testing, and outcomes. Am J Gastroenterol. 2020;115(6):916‐923. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Chen Y, Chen L, Deng Q, et al. The presence of SARS‐CoV‐2 RNA in the feces of COVID‐19 patients. J Med Virol. 2020;92(7):833‐840. [DOI] [PubMed] [Google Scholar]
- 71. Cheung KS, Hung IFN, Chan PPY, et al. Gastrointestinal manifestations of SARS‐CoV‐2 infection and virus load in fecal samples from a Hong Kong cohort: systematic review and meta‐analysis. Gastroenterology. 2020;159(1):81‐95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Li LY, Wu W, Chen S, et al. Digestive system involvement of novel coronavirus infection: prevention and control infection from a gastroenterology perspective. J Dig Dis. 2020;21(4):199‐204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Desforges M, Le Coupanec A, Dubeau P, et al. Human coronaviruses and other respiratory viruses: underestimated opportunistic pathogens of the central nervous system? Viruses. 2019;12(1). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Haidar MA, Jourdi H, Hassan ZH, et al. Neurological and neuropsychological changes associated with SARS‐CoV‐2 infection: new observations, new mechanisms. Neuroscientist. 2021.1073858420984106. [DOI] [PubMed] [Google Scholar]
- 75. Zhou Z, Kang H, Li S, Zhao X. Understanding the neurotropic characteristics of SARS‐CoV‐2: from neurological manifestations of COVID‐19 to potential neurotropic mechanisms. J Neurol. 2020;267(8):2179‐2184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Iadecola C, Anrather J, Kamel H. Effects of COVID‐19 on the nervous system. Cell. 2020;183(1):16‐27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Hamming I, Timens W, Bulthuis M, Lely A, Navis G, van Goor H. Tissue distribution of ACE2 protein, the functional receptor for SARS coronavirus. A first step in understanding SARS pathogenesis. J Pathol. 2004;203(2):631‐637. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. McQuaid C, Brady M, Deane R. SARS‐CoV‐2: is there neuroinvasion? Fluids Barriers CNS. 2021;18(1):32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Monteil V, Kwon H, Prado P, et al. Inhibition of SARS‐CoV‐2 infections in engineered human tissues using clinical‐grade soluble human ACE2. Cell. 2020;181(4):905‐913.e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Paniz‐Mondolfi A, Bryce C, Grimes Z, et al. Central nervous system involvement by severe acute respiratory syndrome coronavirus‐2 (SARS‐CoV‐2). J Med Virol. 2020;92(7):699‐702. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Varga Z, Flammer AJ, Steiger P, et al. Endothelial cell infection and endotheliitis in COVID‐19. Lancet. 2020;395(10234):1417‐1418. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Torices S, Cabrera R, Stangis M, et al. Expression of SARS‐CoV‐2‐related receptors in cells of the neurovascular unit: implications for HIV‐1 infection. J Neuroinflammation. 2021;18(1):167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Alam SB, Willows S, Kulka M, Sandhu JK. Severe acute respiratory syndrome coronavirus 2 may be an underappreciated pathogen of the central nervous system. Eur J Neurol. 2020;27(11):2348‐2360. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Erickson MA, Rhea EM, Knopp RC, Banks WA. Interactions of SARS‐CoV‐2 with the blood–brain barrier. Int J Mol Sci. 2021;22(5):2681. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Deffner F, Scharr M, Klingenstein S, Klingenstein M. Histological evidence for the enteric nervous system and the choroid plexus as alternative routes of neuroinvasion by SARS‐CoV2. Front Neuroanat. 2020;14(74). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Pellegrini L, Albecka A, Mallery DL, et al. SARS‐CoV‐2 infects the brain choroid plexus and disrupts the blood‐CSF barrier in human brain organoids. Cell Stem Cell. 2020;27(6):951‐961.e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Huang YH, Jiang D, Huang JT. SARS‐CoV‐2 detected in cerebrospinal fluid by PCR in a case of COVID‐19 encephalitis. Brain Behav Immun. 2020;87:149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Zhou L, Zhang M, Wang J, Gao J. Sars‐Cov‐2: underestimated damage to nervous system. Trav Med Infect Dis. 2020;36:101642. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Daneman R, Prat A. The blood‐brain barrier. Cold Spring Harb Perspect Biol. 2015;7(1):a020412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Nampoothiri S, Sauve F, Ternier G, et al. The hypothalamus as a hub for SARS‐CoV‐2 brain infection and pathogenesis. bioRxiv; 2020. p. 2020.06.08.139329. [Google Scholar]
- 91. Burdo TH, Lackner A, Williams KC. Monocyte/macrophages and their role in HIV neuropathogenesis. Immunol Rev. 2013;254(1):102‐113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Paul AM, Acharya D, Duty L, et al. Osteopontin facilitates West Nile virus neuroinvasion via neutrophil “Trojan horse” transport. Sci Rep. 2017;7(1):4722. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Zubair AS, McAlpine LS, Gardin T, Farhadian S, Kuruvilla DE, Spudich S. Neuropathogenesis and neurologic manifestations of the coronaviruses in the age of coronavirus disease 2019: a review. JAMA Neurol. 2020;77(8):1018‐1027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Achar A, Ghosh C. COVID‐19‐Associated neurological disorders: the potential route of CNS invasion and blood‐brain relevance. Cells. 2020;9(11). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Yang D, Chu H, Hou Y, et al. Attenuated interferon and proinflammatory response in SARS‐CoV‐2–infected human dendritic cells is associated with viral antagonism of STAT1 phosphorylation. J Infect Dis. 2020;222(5):734‐745. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Chen W, Ju XZ, Lu Y, Ding XW, Miao CH, Chen JW. Propofol improved hypoxia‐impaired integrity of blood‐brain barrier via modulating the expression and phosphorylation of zonula occludens‐1. CNS Neurosci Ther. 2019;25(6):704‐713. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Pacheco PAF, Faria RX. The potential involvement of P2X7 receptor in COVID‐19 pathogenesis: a new therapeutic target? Scand J Immunol. 2021;93(2):e12960. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Neurath MF. COVID‐19 and immunomodulation in IBD. Gut. 2020;69(7):1335‐1342. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Guerrero JI, Barragán LA, Martínez JD, et al. 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):515. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Liu B, Li M, Zhou Z, Guan X, Xiang Y. Can we use interleukin‐6 (IL‐6) blockade for coronavirus disease 2019 (COVID‐19)‐induced cytokine release syndrome (CRS)? J Autoimmun. 2020;111:102452. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Goker Bagca B, Biray Avci C. The potential of JAK/STAT pathway inhibition by ruxolitinib in the treatment of COVID‐19. Cytokine Growth Factor Rev. 2020;54:51‐61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Amruta N, Chastain WH, Paz M, et al. SARS‐CoV‐2 mediated neuroinflammation and the impact of COVID‐19 in neurological disorders. Cytokine Growth Factor Rev. 2021;58:1‐15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. O'Reilly C, Doroudian M, Mawhinney L, Donnelly SC. Targeting MIF in cancer: therapeutic strategies, current developments, and future opportunities. Med Res Rev. 2016;36(3):440‐460. [DOI] [PubMed] [Google Scholar]
- 104. Satarker S, Tom AA, Shaji RA, Alosious A, Luvis M, Nampoothiri M. JAK‐STAT pathway inhibition and their implications in COVID‐19 therapy. Postgrad Med. 2021;133(5):489‐507. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Leonardi M, Padovani A, McArthur JC. Neurological manifestations associated with COVID‐19: a review and a call for action. J Neurol. 2020;267(6):1573‐1576. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Soy M, Keser G, Atagündüz P, Tabak F, Atagündüz I, Kayhan S. Cytokine storm in COVID‐19: pathogenesis and overview of anti‐inflammatory agents used in treatment. Clin Rheumatol. 2020;39(7):2085‐2094. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Rezaei M, Mostafaei S, Aghaei A, et al. The association between HPV gene expression, inflammatory agents and cellular genes involved in EMT in lung cancer tissue. BMC Cancer. 2020;20(1):916. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Chen G, Wu D, Guo W, et al. Clinical and immunological features of severe and moderate coronavirus disease 2019. J Clin Invest. 2020;130(5):2620‐2629. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Feldmann M, Maini RN, Woody JN, et al. Trials of anti‐tumour necrosis factor therapy for COVID‐19 are urgently needed. Lancet. 2020;395(10234):1407‐1409. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Zhou F, Yu T, Du R, et al. Clinical course and risk factors for mortality of adult inpatients with COVID‐19 in Wuhan, China: a retrospective cohort study. Lancet. 2020;395(10229):1054‐1062. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Cao Y, Wei J, Zou L, et al. Ruxolitinib in treatment of severe coronavirus disease 2019 (COVID‐19): a multicenter, single‐blind, randomized controlled trial. J Allergy Clin Immunol. 2020;146(1):137‐146.e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Ye Q, Wang B, Mao J. The pathogenesis and treatment of the `Cytokine Storm' in COVID‐19. J Infect. 2020;80(6):607‐613. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Freeman TL, Swartz TH. Targeting the NLRP3 inflammasome in severe COVID‐19. Front Immunol. 2020;11:1518. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Java A, Apicelli AJ, Liszewski MK, et al. The complement system in COVID‐19: friend and foe? JCI Insight. 2020;5(15). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Risitano AM, Mastellos DC, Huber‐Lang M, et al. Complement as a target in COVID‐19? Nat Rev Immunol. 2020;20(6):343‐344. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Haidar MA, Jourdi H, Hassan ZH, et al. Neurological and Neuropsychological Changes Associated with SARS‐CoV‐2 Infection: New Observations, New Mechanisms. Neuroscientist; 2021.1073858420984106. [DOI] [PubMed] [Google Scholar]
- 117. Talukdar J, Bhadra B, Dattaroy T, Nagle V, Dasgupta S. Potential of natural astaxanthin in alleviating the risk of cytokine storm in COVID‐19. Biomed Pharmacother. 2020;132:110886. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Di Virgilio F, Tang Y, Sarti AC, Rossato M. A rationale for targeting the P2X7 receptor in Coronavirus disease 19. Br J Pharmacol. 2020;177(21):4990‐4994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Ribeiro DE, Oliveira‐Giacomelli Á, Glaser T, et al. Hyperactivation of P2X7 receptors as a culprit of COVID‐19 neuropathology. Mol Psychiatry. 2021;26(4):1044‐1059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Jiang M, Li R, Lyu J, et al. MCC950, a selective NLPR3 inflammasome inhibitor, improves neurologic function and survival after cardiac arrest and resuscitation. J Neuroinflammation. 2020;17(1):256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Luo J, Lu S, Yu M, et al. The potential involvement of JAK‐STAT signaling pathway in the COVID‐19 infection assisted by ACE2. Gene. 2021;768:145325. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. Calabrese LH, Lenfant T, Calabrese C. Cytokine storm release syndrome and the prospects for immunotherapy with COVID‐19, part 4: the role of JAK inhibition. Cleve Clin J Med. 2021. [DOI] [PubMed] [Google Scholar]
- 123. Luo W, Li Y‐X, Jiang L‐J, Chen Q, Wang T, Ye D‐W. Targeting JAK‐STAT signaling to control cytokine release syndrome in COVID‐19. Trends Pharmacol Sci. 2020;41(8):531‐543. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124. Obermeier B, Daneman R, Ransohoff RM. Development, maintenance and disruption of the blood‐brain barrier. Nat Med. 2013;19(12):1584‐1596. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Zhao Z, Nelson AR, Betsholtz C, Zlokovic BV. Establishment and dysfunction of the blood‐brain barrier. Cell. 2015;163(5):1064‐1078. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Liebner S, Dijkhuizen RM, Reiss Y, Plate KH, Agalliu D, Constantin G. Functional morphology of the blood‐brain barrier in health and disease. Acta Neuropathol. 2018;135(3):311‐336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Buzhdygan TP, DeOre BJ, Baldwin‐Leclair A, 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. Bellon M, Schweblin C, Lambeng N, et al. Cerebrospinal fluid features in SARS‐CoV‐2 RT‐PCR positive patients. Clin Infect Dis. 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. Gu J, Gong E, Zhang B, et al. Multiple organ infection and the pathogenesis of SARS. J Exp Med. 2005;202(3):415‐424. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Steardo L, Zorec R, Verkhratsky A. Neuroinfection may contribute to pathophysiology and clinical manifestations of COVID‐19. Acta Physiol (Oxf). 2020;229(3):e13473. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Reichard RR, Kashani KB, Boire NA, Constantopoulos E, Guo Y, Lucchinetti CF. Neuropathology of COVID‐19: a spectrum of vascular and acute disseminated encephalomyelitis (ADEM)‐like pathology. Acta Neuropathol. 2020;140(1):1‐6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Zambrelli E, Canevini M, Gambini O, D'Agostino A. Delirium and sleep disturbances in COVID‐19: a possible role for melatonin in hospitalized patients? Sleep Med. 2020;70:111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. Wang Y, Cao Y, Mangalam AK, et al. Neuropilin‐1 modulates interferon‐γ‐stimulated signaling in brain microvascular endothelial cells. J Cell Sci. 2016;129(20):3911‐3921. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134. Almutairi MM, Gong C, Xu YG, Chang Y, Shi H. Factors controlling permeability of the blood‐brain barrier. Cell Mol Life Sci. 2016;73(1):57‐77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135. Labus J, Wöltje K, Stolte KN. IL‐1β promotes transendothelial migration of PBMCs by upregulation of the FN/α5β1 signalling pathway in immortalised human brain microvascular endothelial cells. Exp Cell Res. 2018;373(1):99‐111. [DOI] [PubMed] [Google Scholar]
- 136. Huang X, Hussain B, Chang J. Peripheral inflammation and blood–brain barrier disruption: effects and mechanisms. CNS Neurosci Ther. 2021;27(1):36‐47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137. Rempe RG, Hartz AMS, Bauer B. Matrix metalloproteinases in the brain and blood–brain barrier: versatile breakers and makers. J Cereb Blood Flow Metab. 2016;36(9):1481‐1507. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138. Yao C, Narumiya S. Prostaglandin‐cytokine crosstalk in chronic inflammation. Br J Pharmacol. 2019;176(3):337‐354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139. Wu K, Fukuda K, Xing F, et al. Roles of the cyclooxygenase 2 matrix metalloproteinase 1 pathway in brain metastasis of breast cancer*. J Biol Chem. 2015;290(15):9842‐9854. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140. Erickson MA, Banks WA. Neuroimmune axes of the blood–brain barriers and blood–brain interfaces: bases for physiological regulation, disease states, and pharmacological interventions. Pharmacol Rev. 2018;70(2):278‐314. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141. Tremblay M‐E, Madore C, Bordeleau M, Tian L, Verkhratsky A. Neuropathobiology of COVID‐19: the role for glia. Front Cell Neurosci. 2020;14(363). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142. Wang Y, Jin S, Sonobe Y, et al. Interleukin‐1β induces blood‐brain barrier disruption by downregulating Sonic hedgehog in astrocytes. PLoS ONE. 2014;9(10):e110024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143. da Fonseca ACC, Matias D, Garcia C, et al. The impact of microglial activation on blood‐brain barrier in brain diseases. Front Cell Neurosci. 2014;8(362). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144. Matschke J, Lütgehetmann M, Hagel C, et al. Neuropathology of patients with COVID‐19 in Germany: a post‐mortem case series. Lancet Neurol. 2020;19(11):919‐929. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145. Schwabenland M, Salié H, Tanevski J, et al. Deep spatial profiling of human COVID‐19 brains reveals neuroinflammation with distinct microanatomical microglia‐T‐cell interactions. Immunity. 2021;54(7):1594‐1610.e11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146. Dhont S, Derom E, Van Braeckel E, Depuydt P, Lambrecht BN. The pathophysiology of ‘happy’ hypoxemia in COVID‐19. Respir Res. 2020;21(1):198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147. González‐Duarte A, Norcliffe‐Kaufmann L. Is ‘happy hypoxia’ in COVID‐19 a disorder of autonomic interoception? A hypothesis. Clin Aut Res. 2020;30(4):331‐333. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148. Engelhardt S, Patkar S, Ogunshola OO. Cell‐specific blood‐brain barrier regulation in health and disease: a focus on hypoxia. Br J Pharmacol. 2014;171(5):1210‐1230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149. Jaakkola P, Mole DR, Tian Y‐M, et al. Targeting of HIF‐alpha to the von Hippel‐Lindau ubiquitylation complex by O2‐regulated prolyl hydroxylation. Science. 2001;292(5516):468‐472. [DOI] [PubMed] [Google Scholar]
- 150. Colgan SP, Furuta GT, Taylor CT. Hypoxia and innate immunity: keeping up with the HIFsters. Annu Rev Immunol. 2020;38:341‐363. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151. Taniguchi‐Ponciano K, Vadillo E, Mayani H, et al. Increased expression of hypoxia‐induced factor 1α mRNA and its related genes in myeloid blood cells from critically ill COVID‐19 patients. Ann Med. 2021;53(1):197‐207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152. Serebrovska ZO, Chong EY, Serebrovska TV, Tumanovska LV, Xi L. Hypoxia, HIF‐1α, and COVID‐19: from pathogenic factors to potential therapeutic targets. Acta Pharmacol Sin. 2020;41(12):1539‐1546. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153. Rochfort KD, Cummins PM. Cytokine‐mediated dysregulation of zonula occludens‐1 properties in human brain microvascular endothelium. Microvasc Res. 2015;100:48‐53. [DOI] [PubMed] [Google Scholar]
- 154. Ballabh P, Braun A, Nedergaard M. The blood‐brain barrier: an overview: structure, regulation, and clinical implications. Neurobiol Dis. 2004;16(1):1‐13. [DOI] [PubMed] [Google Scholar]
- 155. Xie J, Covassin N, Fan Z, et al. Association between hypoxemia and mortality in patients with COVID‐19. Mayo Clin Proc. 2020;95(6):1138‐1147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156. Rello J, Storti E, Belliato M, Serrano R. Clinical phenotypes of SARS‐CoV‐2: implications for clinicians and researchers. Eur Respir J. 2020;55(5):2001028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157. Li Y, Li M, Wang M, et al. Acute cerebrovascular disease following COVID‐19: a single center, retrospective, observational study. Stroke Vasc Neurol. 2020;5(3):279‐284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158. Klok FA, Kruip MJHA, van der Meer NJM, et al. Incidence of thrombotic complications in critically ill ICU patients with COVID‐19. Thromb Res. 2020;191:145‐147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159. Wu T, Zuo Z, Yang D, et al. Venous thromboembolic events in patients with COVID‐19: a systematic review and meta‐analysis. Age Ageing. 2021;50(2):284‐293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160. Wu C, Chen X, Cai Y, et al. Risk factors associated with acute respiratory distress syndrome and death in patients with coronavirus disease 2019 pneumonia in Wuhan, China. JAMA Intern Med. 2020;180(7):934‐943. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161. Connors JM, Levy JH. COVID‐19 and its implications for thrombosis and anticoagulation. Blood. 2020;135(23):2033‐2040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162. Divani AA, Andalib S, Di Napoli M, et al. Coronavirus disease 2019 and stroke: clinical manifestations and pathophysiological insights. J Stroke Cerebrovasc Dis. 2020;29(8):104941. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163. Guan W‐j, Ni Z‐y, Hu Y, et al. Clinical characteristics of coronavirus disease 2019 in China. N. Engl J Med. 2020;382(18):1708‐1720. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164. Lodigiani C, Iapichino G, Carenzo L, et al. Venous and arterial thromboembolic complications in COVID‐19 patients admitted to an academic hospital in Milan, Italy. Thromb Res. 2020;191:9‐14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165. Yaghi S, Ishida K, Torres J, et al. SARS‐CoV‐2 and stroke in a New York healthcare system. Stroke. 2020;51(7):2002‐2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166. Gu SX, Tyagi T, Jain K, et al. Thrombocytopathy and endotheliopathy: crucial contributors to COVID‐19 thromboinflammation. Nat Rev Cardiol. 2021;18(3):194‐209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167. Mehrabadi ME, Hemmati R, Tashakor A, et al. Induced dysregulation of ACE2 by SARS‐CoV‐2 plays a key role in COVID‐19 severity. Biomed Pharmacother. 2021;137:111363. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168. Goudarzi E, Yousefimoghaddam F, Ramandi A, Khaheshi I. COVID‐19 and peripheral artery thrombosis: a mini review. Curr Probl Cardiol. 2021:100992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169. Middleton EA, He X‐Y, Denorme F, et al. Neutrophil extracellular traps contribute to immunothrombosis in COVID‐19 acute respiratory distress syndrome. Blood. 2020;136(10):1169‐1179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170. Puelles VG, Lütgehetmann M, Lindenmeyer MT, et al. Multiorgan and renal tropism of SARS‐CoV‐2. N. Engl J Med. 2020;383(6):590‐592. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171. Thacker VV, Sharma K, Dhar N, Mancini G‐F, Sordet‐Dessimoz J, McKinney JD. Rapid endotheliitis and vascular damage characterize SARS‐CoV‐2 infection in a human lung‐on‐chip model. EMBO Rep. 2021;22(6):e52744. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172. Jose RJ, Manuel A. COVID‐19 cytokine storm: the interplay between inflammation and coagulation. Lancet Respir Med. 2020;8(6):e46‐e47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173. Levi M, van der Poll T. Coagulation and sepsis. Thromb Res. 2017;149:38‐44. [DOI] [PubMed] [Google Scholar]
- 174. Perico L, Benigni A, Casiraghi F, Ng LFP, Renia L, Remuzzi G. Immunity, endothelial injury and complement‐induced coagulopathy in COVID‐19. Nat Rev Nephrol. 2021;17(1):46‐64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175. Chauhan AJ, Wiffen LJ, Brown TP. COVID‐19: a collision of complement, coagulation and inflammatory pathways. J Thromb Haemost. 2020;18(9):2110‐2117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176. Conway EM, Pryzdial ELG. Is the COVID‐19 thrombotic catastrophe complement‐connected? J Thromb Haemost. 2020;18(11):2812‐2822. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177. Rapkiewicz AV, Mai X, Carsons SE, et al. Megakaryocytes and platelet‐fibrin thrombi characterize multi‐organ thrombosis at autopsy in COVID‐19: a case series. EClinicalMedicine. 2020;24:100434. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178. Libby P, Lüscher T. COVID‐19 is, in the end, an endothelial disease. Eur Heart J. 2020;41(32):3038‐3044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179. Markus HS, Brainin M. COVID‐19 and stroke—a global World Stroke Organization perspective. Int J Stroke. 2020;15(4):361‐364. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180. Finsterer J, Scorza FA, Fiorini AC. SARS‐CoV‐2‐associated Guillain‐Barre syndrome in 62 patients. Eur J Neurol. 2021;28(1):e10‐e12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181. Galeotti C, Bayry J. Autoimmune and inflammatory diseases following COVID‐19. Nat Rev Rheumatol. 2020;16(8):413‐414. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182. Dalakas MC. Guillain‐Barré syndrome: the first documented COVID‐19‐triggered autoimmune neurologic disease: more to come with myositis in the offing. Neurol Neuroimmunol Neuroinflamm. 2020;7(5). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183. Toscano G, Palmerini F, Ravaglia S, et al. Guillain‐Barré syndrome associated with SARS‐CoV‐2. N Engl J Med. 2020;382(26):2574‐2576. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184. Dalakas MC. Pathogenesis of immune‐mediated neuropathies. Biochim Biophys Acta. 2015;1852(4):658‐666. [DOI] [PubMed] [Google Scholar]
- 185. Lucchese G, Flöel A. SARS‐CoV‐2 and Guillain‐Barré syndrome: molecular mimicry with human heat shock proteins as potential pathogenic mechanism. Cell Stress Chaperones. 2020;25(5):731‐735. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186. Lucchese G, Flöel A. Guillain‐Barré syndrome, SARS‐CoV‐2 and molecular mimicry. Brain. 2021;144(5):e43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187. Marino Gammazza A, Légaré S, Lo Bosco G, et al. Molecular mimicry in the post‐COVID‐19 signs and symptoms of neurovegetative disorders? Lancet Microbe. 2021;2(3):e94. [DOI] [PubMed] [Google Scholar]
- 188. Gupta M, Weaver DF. COVID‐19 as a trigger of brain autoimmunity. ACS Chem Neurosci. 2021;12(14):2558‐2561. [DOI] [PubMed] [Google Scholar]
- 189. Sriwastava S, Kataria S, Tandon M, et al. Guillain Barré Syndrome and its variants as a manifestation of COVID‐19: a systematic review of case reports and case series. J Neurol Sci. 2021;420:117263. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190. Zhao H, Shen D, Zhou H, Liu J, Chen S. Guillain‐Barré syndrome associated with SARS‐CoV‐2 infection: causality or coincidence? Lancet Neurol. 2020;19(5):383‐384. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191. Ellul MA, Benjamin L, Singh B, et al. Neurological associations of COVID‐19. Lancet Neurol. 2020;19(9):767‐783. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192. Lunn MP, Cornblath DR, Jacobs BC, et al. COVID‐19 vaccine and Guillain‐Barré syndrome: let’s not leap to associations. Brain. 2021;144(2):357‐360. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193. Abu‐Rumeileh S, Abdelhak A, Foschi M, Tumani H, Otto M. Guillain‐Barré syndrome spectrum associated with COVID‐19: an up‐to‐date systematic review of 73 cases. J Neurol. 2021;268(4):1133‐1170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194. Fragiel M, Miró Ò, Llorens P, et al. Incidence, clinical, risk factors and outcomes of Guillain‐Barré in Covid‐19. Ann Neurol. 2021;89(3):598‐603. [DOI] [PubMed] [Google Scholar]
- 195. Filosto M, Cotti Piccinelli S, Gazzina S, et al. Guillain‐Barré syndrome and COVID‐19: an observational multicentre study from two Italian hotspot regions. J Neurol Neurosurg Psychiatry. 2021;92(7):751‐756. [DOI] [PubMed] [Google Scholar]
- 196. Dufour C, Co TK, Liu A. GM1 ganglioside antibody and COVID‐19 related Guillain Barre Syndrome ‐ a case report, systemic review and implication for vaccine development. Brain Behav Immun Health. 2021;12:100203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197. Ehrenfeld M, Tincani A, Andreoli L, et al. Covid‐19 and autoimmunity. Autoimmun Rev. 2020;19(8):102597. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198. Koike H, Chiba A, Katsuno M. Emerging infection, vaccination, and Guillain‐Barré syndrome: a review. Neurol Ther; 2021:1‐15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199. van Doorn PA, Kuitwaard K, Walgaard C, van Koningsveld R, Ruts L, Jacobs BC. IVIG treatment and prognosis in Guillain‐Barré syndrome. J Clin Immunol. 2010;30(suppl 1):S74‐S78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200. Galeotti C, Kaveri SV, Bayry J. Intravenous immunoglobulin immunotherapy for coronavirus disease‐19 (COVID‐19). Clin Transl Immunol. 2020;9(10):e1198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201. Çolak M, Kalemci S, Sarıhan A. Treatment of a case of COVID‐19 by intravenous immunoglobulin. J Glob Antimicrob Resist. 2021;24:106‐107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202. Brem MD, Jacobs BC, van Rijs W, et al. IVIg‐induced plasmablasts in patients with Guillain‐Barré syndrome. Ann Clin Transl Neurol. 2019;6(1):129‐143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203. Archelos JJ, Fazekas F. IVIG therapy in neurological disorders of childhood. J Neurol. 2006;253(5):v80‐v86. [DOI] [PubMed] [Google Scholar]
- 204. Ferini‐Strambi L, Salsone M. COVID‐19 and neurological disorders: are neurodegenerative or neuroimmunological diseases more vulnerable? J Neurol. 2021;268(2):409‐419. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205. Barzegar M, Mirmosayyeb O, Nehzat N, et al. COVID‐19 infection in a patient with multiple sclerosis treated with fingolimod. Neurol Neuroimmunol Neuroinflamm. 2020;7(4). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206. Mateen FJ, Rezaei S, Alakel N, Gazdag B, Kumar AR, Vogel A. Impact of COVID‐19 on U.S. and Canadian neurologists' therapeutic approach to multiple sclerosis: a survey of knowledge, attitudes, and practices. J Neurol. 2020;267(12):3467‐3475. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207. Rostami Mansoor S, Ghasemi‐Kasman M. Impact of disease‐modifying drugs on the severity of COVID‐19 infection in multiple sclerosis patients. J Med Virol. 2021;93(3):1314‐1319. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208. Willis MD, Robertson NP. Multiple sclerosis and the risk of infection: considerations in the threat of the novel coronavirus, COVID‐19/SARS‐CoV‐2. J Neurol. 2020;267(5):1567‐1569. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209. Persson R, Lee S, Ulcickas Yood M, et al. Infections in patients diagnosed with multiple sclerosis: a multi‐database study. Mult Scler Relat Disord. 2020;41:101982. [DOI] [PubMed] [Google Scholar]
- 210. Hughes R, Whitley L, Fitovski K, et al. COVID‐19 in ocrelizumab‐treated people with multiple sclerosis. Mult Scler Relat Disord. 2021;49:102725. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211. Chaudhry F, Bulka H, Rathnam AS, et al. COVID‐19 in multiple sclerosis patients and risk factors for severe infection. J Neurol Sci. 2020;418:117147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212. Hughes R, Pedotti R, Koendgen H. COVID‐19 in persons with multiple sclerosis treated with ocrelizumab ‐ a pharmacovigilance case series. Mult Scler Relat Disord. 2020;42:102192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213. Baker D, Amor S, Kang AS, Schmierer K, Giovannoni G. The underpinning biology relating to multiple sclerosis disease modifying treatments during the COVID‐19 pandemic. Mult Scler Relat Disord. 2020;43:102174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214. Meca‐Lallana V, Aguirre C, Beatrizdel Río fnm, Cardeñoso L, Alarcon T, Vivancos J. COVID‐19 in 7 multiple sclerosis patients in treatment with ANTI‐CD20 therapies. Mult Scler Relat Disord. 2020;44:102306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215. Mantero V, Abate L, Basilico P, et al. COVID‐19 in dimethyl fumarate‐treated patients with multiple sclerosis. J Neurol. 2021;268(6):2023‐2025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216. Loonstra FC, Hoitsma E, van Kempen ZL, Killestein J, Mostert JP. COVID‐19 in multiple sclerosis: the Dutch experience. Mult Scler. 2020;26(10):1256‐1260. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
Data sharing is not applicable as there is no new data were analysed in this study.
