Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Apr 1.
Published in final edited form as: J Med Virol. 2020 Dec 23;93(4):1983–1998. doi: 10.1002/jmv.26720

Cellular mechanisms underlying neurological/neuropsychiatric manifestations of COVID-19

Brittany Bodnar 1,2,3, Kena Patel 1,2,5, Wenzhe Ho 1, Jin Jun Luo 4, Wenhui Hu 1,2,3,*
PMCID: PMC7897247  NIHMSID: NIHMS1655249  PMID: 33300152

Abstract

Patients with SARS-CoV-2 infection manifest mainly respiratory symptoms. However, clinical observations frequently identified neurological symptoms and neuropsychiatric disorders related to COVID-19 (Neuro-SARS2). Accumulated robust evidence indicates that Neuro-SARS2 may play an important role in aggravating the disease severity and mortality. Understanding the neuropathogenesis and cellular mechanisms underlying Neuro-SARS2 is crucial for both basic research and clinical practice to establish effective strategies for early detection/diagnosis, prevention, and treatment paradigms. In this review, we will comprehensively examine current evidence of SARS-CoV-2 infection in various neural cells including neurons, microglia/macrophages, astrocytes, pericytes/endothelial cells, ependymocytes/choroid epithelial cells, and neural stem/progenitor cells. Although significant progress has been made in studying Neuro-SARS2, much remains to be learned about the neuroinvasive routes (transneuronal and hematogenous) of the virus and the cellular/molecular mechanisms underlying the development/progression of this disease. Future and ongoing studies require the establishment of more clinically relevant and suitable neural cell models using human induced pluripotent stem cells, brain organoids and postmortem specimens.

Keywords: SARS-CoV-2, COVID-19, neural cells, neurology, neuropsychiatry, brain organoids, iPS cells, neuroinvasion, neurovirulence

Introduction

SARS-CoV-2 virus has caused a global pandemic affecting 218 countries and territories with 51,791,296 confirmed cases and 1,278,460 confirmed deaths, as of November 10, 2020 (www.worldometers.info/coronavirus), and large numbers of new cases still occur daily. The unprecedented scale of the pandemic continues to push the boundaries of scientific research and development as the community proceeds to further understand the virus and its complexity involving multiple organ systems with varying severities. The most common and predominant presentations of illness are respiratory symptoms. However, there are increasing reports of neurological and neuropsychiatric symptoms18. Such symptoms may be nonspecific, such as somatoform disorders including muscle sore, dizziness, headache, anxiety, sleep disturbances, depression, fatigue, anger/stress, loneliness, stigmatization; or specific, such as central nervous system (CNS) disorders including stroke, seizures, encephalitis, ataxia, and myelitis; peripheral nervous system (PNS) disorders including loss of smell and taste (anosmia/hyposmia and ageusia/dysgeusia), Guillain-Barré syndrome and Miller Fisher syndrome; or psychiatric and neuropsychiatric syndromes including major depressive disorder, bipolar disorder, obsessive-compulsive disorder and post-traumatic stress disorder. The exact mechanisms of SARS-CoV-2 underlying neurological/neuropsychiatric dysfunctions are not fully understood but seem to be due to a combination of potentially direct viral infection, or indirectly systemic disease complication plus para/post-infectious inflammation in the nervous system. Notably, knowledge of the long-term effects of SARS-CoV-2 on the nervous system in surviving COVID-19 patients is virtually absent.

In this review, we will focus on the current understanding of cellular mechanisms underlying COVID-19 neurological/neuropsychiatric pathogenesis and symptoms. Accumulated evidence has shown that SARS-CoV-2 may directly infect various neural cells. Additionally, systemic immune activation and chronic neuroinflammation may also contribute to neurological dysfunctions. Although severe neurological complications seem to be rare, the global scale of the pandemic can amount to a large proportion of cases in the long-term as symptoms and latent effects of the virus continue to manifest, potentially leaving patients with prolonged existing of neurological and/or neuropsychiatric disturbances7, 8. Presumably, neurological and neuropsychiatric manifestations in surviving COVID-19 patients may have equal, if not more significant, adverse consequences when compared to pulmonary manifestations. Understanding the cellular mechanism will pave the way for basic research directions and clinical therapeutic strategies. For better clarity and understanding, we tentatively assign SARS1 for the original 2003 SARS caused by SARS-CoV-1 and SARS2 for the 2019 COVID caused by SARS-CoV-2, while SARS defines both SARS1 and SARS2, and SARS-CoV represents SARS-CoV-1 and SARS-CoV-2.

1. Clinical evidence of SARS-CoV-2 infection in the nervous system

Neurologic and neuropsychiatric dysfunctions in SARS2 patients have recently been well discussed16, 9. However, many questions remain to be clarified, particularly if these neurologic/psychiatric disturbances are caused by a direct infection of SARS-CoV-2 to the brain or the downstream consequences via indirect mechanisms caused by SARS-CoV-2 systematically. The previous clinical findings of the brains from patients infected with SARS-CoV-1 have suggested that these SARS-CoVs are able to invade the brain. Subsequent clinical reports have documented both CNS and PNS involvement including stroke, seizures, smell loss, Guillain-Barré syndrome, and myopathies (manifested as rhabdomyolysis)10. Autopsies from several SARS1 patients revealed the presence of SARS-CoV-1 in the brain, primarily in the cytoplasm of neurons in the cortex and hypothalamus11. Additionally, SARS-CoV-1 could be detected in the cerebrospinal fluid (CSF) of SARS1 patients who were experiencing seizures10. Similarly, neurological complications were reported in some patients with MERS, including peripheral neuropathies, encephalopathies, intracerebral hemorrhage, stroke, seizures, and confusion12. However, there is no confirmatory evidence from post-mortem or CSF viral detection studies to support MERS-CoV directly infecting the brain.

In SARS2 cases, SARS-CoV-2 has been detected by RT-PCR assay in the CSF of numerous patients with various neurological symptoms1317 although many anecdotal clinical reports showed a negative test for SARS-CoV-2 by RT-PCR assays of CSF samples18. The reason why no viral load was detectable in the CSF of the majority of SARS2 patients with an active neurological condition remains unclear. This discrepancy could be due to the differences in sensitivity of RT-PCR assays, the stages of infection and the timing when the CSF sample is manipulated and the assay is performed, or the clearance of SARS-CoV-2 from the CSF. Nonetheless, detection of SARS-CoV-2 by RT-PCR in CSF from patients with active neurological symptoms has provided undeniable evidence that SARS-CoV-2 could invade the nervous system. Undoubtedly, more studies are needed to secure convincible evidence.

A case series study of 18 post-mortem SARS2 patients with various neurological symptoms has shown evidence of acute hypoxic ischemic damages in the brains of all patients, including neuronal loss19. Of these patients, the virus was detected by RT-PCR in 5 brains. SARS-CoV-2 spike (S) protein was detected in cortical neurons and endothelial cells in SARS2 brain samples20 but not the viral nucleocapsid (N) protein by immunohistochemical analyses19.

Other neuropathological case studies have found histological evidence of vascular etiologies, encephalitis (both global and brain stem-specific) and/or meningitis, demyelination, and other encephalopathies21, 22. Postmortem analysis using transmission electron microscopy and RT-PCR assay in a SARS2 patient with Parkinson’s disease revealed the presence of SARS-CoV-2 in neurons and capillary endothelial cells in the frontal lobe23. However, the question remains whether the outcome neuropathology resulted from direct infection or a downstream event following a systemic infection; or whether the lack of evidence of vital direct infection was an outcome of low sensitivity of detection threshold in methodologies. New technologies such as RNAscope have been utilized to detect SARS-CoV-2 RNA in cultured cells24 and lung specimen of SARS2 patients, and are expected to validate the existence of SARS-CoV-2 in brain tissues. Digital droplet PCR, viral outgrowth assay, and single cell RNA sequencing of brain samples from SARS2 patients may also help to validate the neurotropism of SARS-CoV-2. Further multilabeled immunohistochemical analyses are needed to better understand the cellular distribution and mechanisms of SARS-CoV-2 in the nervous system.

In summary, accumulated evidence from clinical studies has shown a variety of neurological and neuropsychiatric manifestations in SARS2 patients (Table 1). However, more convincible evidence for direct viral infection to the nervous system remains to be found in addition to the indirect contribution of systemic immune/inflammatory responses after SARS-CoV-2 infection25. The availability of the advanced new technologies for neuropathogenic studies on human brain specimen are expected to continuously provide in-depth findings of SARS-CoV-2 infection in the nervous system.

Table 1:

Clinical Features of Neurological and Neuropsychiatric Symptoms in SARS2 Patients (Percentages derived from the clinical studies with >100 patients; The others noted the case numbers)

Manifestation Type Symptom/Feature Percentage Reference
General Neurological Symptoms Any nervous symptoms 36.4%–82.3% 9799
Headache 6.5%–70.3% 79, 97105
Muscle pain (myalgia) 22%–62.5% 79, 97105
Dizziness 8%–29.7% 79, 98, 99, 101, 105
Fatigue 50%–69.6% 79, 101, 103, 105
Seizures <1% 98, 99, 106108
Impaired/disordered consciousness 4.3%–9% 98, 99, 101, 109
Rhabdomyolysis 3.5% 98
Sensory Impairments Anosmia/hyposmia 5.1%–70.2% 76, 79, 9799, 103, 104
Ageusia/dysgeusia 5.6%–54.2% 76, 79, 9799, 103, 104
Vision impairment 1.4%–7.4% 97, 99, 103
Neuropathies Peripheral neuropathy 0.4%–8.9% 98, 99, 107, 108
Gullian-Barre Syndrome and variants 23 cases 107, 108, 110
Nerve pain 2.3% 99
Focal weakness and/or motor deficit 0.6%–2.5% 97, 98
Encephalopathies Encephalopathy (undefined) 7.2%–31.8% 98, 107, 108
Confusion/disorientation/altered mental state 2.0%–27.9 97, 100, 106, 108, 109
Acute necrotizing encephalopathy (ANE) 3 cases 17, 111
Leukoencephalopathy 2 cases 21, 107
Hypoxic encephalopathy 9% 101
Inflammations Acute demyelinating encephalomyelitis
(ADEM)
10 cases 22, 107
Myelitis (CNS) 3 cases 107
Encephalitis 15 cases 98, 107, 108, 112
Lymphocytic meningitis 6 cases 112
Acute cerebrovascular disease (cumulative cases and/or unspecified) Ischemic stroke 1.4%–5.0% 21, 99, 106108, 112, 113
Intracerebral hemorrhage 25 cases 21, 98, 106, 108, 112, 113
Depression 32.6% 109
Anxiety 35.7% 109
Neuropsychiatric Presentations Sleep disorders/insomnia 37.2%–41.9% 97, 109
Anger/stress/irritability 3.9% 109
Psychosis 0.8%–4.4% 108, 109
Behavioral disorder (not specified) 2.0% 97
Frequent recall of traumatic memories 109
Cognitive disorder 0.5% 108
Acute hypoxic ischemic damage 19, 101
Astrogliosis 22, 114, 115
Neuropathologic Features Microgliosis 115
Demyelination 22
Neuronal Cell Loss/Injury 22, 112
Cytotoxic T-cell infiltration 115
Macrophage infiltration 22

2. The neuropathogenic modeling for SARS2

Due to the limited sources of human brain specimen from SARS2 patients, various models have been developed to explore the neurovirulent infection of SARS-CoV-2 in various types of neural cells and recapitulate the neuropathogenesis and neurological manifestation as well as their correlations with viral neuroinvasion (Table 2).

Table 2:

Experimental Models for SARS2 Neuropathogenesis

Model Description Model Characteristics References
In vivo Mouse Models Transgenic K18-hACE2 mice Human ACE2 expression driven by the epithelial cell-specific K18 promoter High susceptibility to SARS-CoV-2 infection (2×103~1×105 plague formation unit) and best recapitulation of several SARS2 clinical features; Several studies also report presence of viral RNA and/or histopathological findings in the brains of some mice20, 32, 35, 36, 80, 116 20, 32, 33, 35, 36, 80, 116, 117
Transgenic HFH4-hACE2 mice Human ACE2 expression driven by the lung ciliated epithelial cell-specific HFH4/FOXJ1 promoter Susceptibility to SARS-CoV-2 infection and presentation of interstitial pneumonia; High viral RNA levels were detected in the brain of a portion of mice and was highly correlated with increased mortality 34
Transgenic hACE2 mice CRISPR/Cas9 edited knock-in mice that express human ACE2 under the mouse genomic Ace2 promoter Human ACE2 replaces mouse Ace2 expression; High susceptibility to SARS-CoV-2 infection, with particularly high viral RNA levels found in the brain 31
Adenovirus delivery of hACE2 Human ACE2 packaged in adenoviral particles under the CMV promoter and delivered to wildtype mice for exogenous expression of hACE2 Human ACE2 expressing cells show susceptibility to SARS-CoV-2 infection, with high viral titers in lung tissues; Mice develop some pathologies but do not display severe SARS2 features 32, 118, 119
In vitro Human iPSC-Derived Culture Models Monolayer neural cell cultures Human iPSC-derived neural cell cultures patterned into specific cell types. Monolayer cultures of mature neurons41, 42, 47 of various subtypes (e.g.- dopaminergic, cortical, etc.), NSCs/NPCs20, 40, 73, and glial cell cultures41, 42, including microglia and astrocytes; Susceptibility to SARS-CoV-2 infection varies by cell type 20, 4042, 47, 73
Cerebral organoids 3D cerebral organoids generated from human iPSCs Mature organoids present as a heterogenous 3D structure consisting of multiple neural cell types, including neurons, NSCs/NPCs, and glial cells; Susceptible to SARS-CoV-2 infection to various extent 20, 40, 44, 45, 47, 120
Region-specific cerebral organoids 3D cerebral organoids generated from human iPSCs that are patterned to specific regional lineages Organoids of hippocampal, midbrain, hypothalamic, and forebrain subtypes with heterogenous mixture of neural cells; Varied susceptibility to SARS-CoV-2 infection 41
Choroid plexus organoids 3D cerebral organoids generated from human iPSCs primarily composed of choroid plexus epithelial cells High susceptibility to SARS-CoV-2 infection; SARS-CoV-2 infection leads to transcriptional dysregulation and impaired barrier function 41, 46
Blood vessel organoids 3D organoid structures derived from human iPSCs composed of vascular networks of endothelial cells High susceptibility to SARS-CoV-2 infection that can be inhibited in an ACE2-dependent manner 65

a. Preclinical animal models

Angiotensin-converting enzyme-2 (ACE2) has been well recognized to serve as the major receptor for SARS-CoV-2 to gain entry into host cells26. The homology conservation and the expression patterns of ACE2 in various species have been identified27. Therefore, numerous non-rodent species have been tested as natural animal models for SARS2, including monkey, ferret, hamster, cat, bat, and pig28. Cynomolgus macaques infected with SARS-CoV-2 exhibit virus shedding and replication in lung tissues and certain pathological changes, but no clinical signs29, while another study showed moderate clinical symptoms in rhesus macaques infected with SARS-CoV-230. Thus, non-human primate models may be useful for further preclinical tests; however, monkey models in general fail to manifest clinical illness and are very expensive and inconvenient. Mouse models are more practical and feasible for elucidating virus pathogenicity and therapeutic targets. However, both SARS-CoVs exhibit a poor cellular tropism in wild type mice due to significant species difference in ACE2 sequence and function. Thus, various types of human ACE2 (hACE2)-expressing mouse models have been established for SARS-CoV-2 infection (Table 2). Interestingly, only hACE2 mouse models among the currently available SARS2 animal models fully recapitulated SARS2 clinical scenarios, particularly the severity and mortality of the illness. The successfully established hACE2-transgenic mouse model with high susceptibility to both SARS-CoV-1 and SARS-CoV-2 infection provided a useful tool in SARS-CoVs studies3134, particularly the K18-hACE2 transgenic mice, in which hACE2 expression is driven by the epithelial cell-specific promoter human cytokeratin 18 (K18) and is capable of recapitulating many features of SARS-CoV-2 infections32, 33, 35, even better than the other hACE2-expressing mouse models32, 35.

Although numerous reports on SARS2 animal models are available, the understanding of neuroinvasive pathogenesis and resultant neurological changes caused by the virus remain limited. In SARS2 animal studies, the virus is delivered intranasally, which may readily infect brain via olfactory nervous system. The brain cellular expression pattern of ACE2 in these animal models may address the susceptibility of brain tissues/cells to SARS-CoV-2. SARS-CoV-2 RNAs are detected in the brain of hACE2 mice after intranasal inoculation31, 34, 36.

b. Human brain organoid models

To get the most clinically relevant data for whether and how SARS-CoV-2 may infect human brain, it is important to use materials from humans. The simplest and most direct approach to test human cellular susceptibility to SARS-CoV-2 is to utilize immortalized human neural cell lines or primary neural cells. SARS-CoV-2 can moderately infect and replicate in the human glioblastoma cell line U25137. Primary olfactory sensory neurons of human4 and hamsters38 are highly susceptible to SARS-CoV-2 infection. However, another study did not detect any presence of SARS-CoV-2 in primary olfactory neurons in hamsters, but did identify viral infection in a large proportion of the supporting sustentacular cells39. In cultured human neural stem cells (NSC)/neural progenitor cells (NPC), ACE2, TMPRSS2, cathepsin L, and furin were readily detected and cells were highly susceptible to infection with SARS-CoV-240, but not SARS-CoV-140. Human primary astrocytes are rarely (0.18%) infected by SARS-CoV-241. Other human primary neural cells have not yet been examined for their susceptibility to SARS-CoV-2 infection.

Fortunately, the rapidly evolving technology surrounding stem cell culture has allowed for the generation of human induced pluripotent stem cells (iPSCs) that can be differentiated into virtually any cell lineage. This has led to the generation of pure human neural cells such as NSCs/NPCs, neurons, astrocytes, microglia, oligodendrocytes, endothelial cells, etc. Human iPSC-derived neurons, particularly dopaminergic neurons, are highly susceptible to S-pseudotyped SARS-CoV-2 infection, but iPSC-derived microglia and macrophages are moderately infected by S-pseudovirions42. In contrast, another report showed that iPSC-derived neurons and astrocytes were sparsely infected, and microglia had no infection; however, interestingly, the choroid plexus epithelial cells underwent robust infection by live SARS-CoV-2 virus41. The inconsistent reports in neural cell viral susceptibility may result from variable differentiation protocols, the neural cell maturity, and virus infection dosage.

Three-dimensional tissue cultures known as organoids contain multiple cell types and recapitulate tissue structures and development. Cerebral organoids (COs) have been used to model several neurodevelopmental and neuropathological disorders as they accurately resemble the molecular, cellular, and structural features of the developing brain (Figure 1a). COs can be readily infected with neurotropic viruses and develop phenotypes similar to that seen clinically, as has been shown in several models studying Zika virus (ZIKV)43.

Figure 1: Human brain organoids to model SARS-CoV-2 infection.

Figure 1:

(a) Example timeline of an experiment to generate cerebral organoids (COs) with SARS-CoV-2 infection. Human fibroblasts or peripheral blood mononuclear cells (PBMC) are reprogrammed to generate induced pluripotent stem cells (iPSCs), which are then grown in 3D suspension culture as embryoid bodies (EBs). After neural induction, COs are embedded in Matrigel to promote formation of neuroepithelial lobes and then transferred to spinning culture, using an orbital shaker or similar method. At around one month, microglia begin to mature in microglia-containing COs. By around 50–60 days, astrocytes can be detected in COs. At this point, COs are in a state of growth and neural cell maturation; this is typically the timepoint where COs are infected with SARS-CoV-2 for experiments. (b) Examples of different types of COs. After pre-patterning to a neuroectodermal fate using dual SMAD inhibition, EBs can be induced into region-specific organoids by adding different patterning factors. Several studies have used this technique to examine the effect of SARS-CoV-2 infection on different brain regions, particularly the choroid plexus. MPC, microglia progenitor cells; NSC/NPC, neural stem/progenitor cells; OPC, oligodendrocyte progenitor cells.

As clinical samples from SAR2 patients are scarce, COs make an attractive experimental model for studying the neuropathogenic effects of SARS-CoV-2 infections in human live brain. To date, several labs have utilized COs to study the neurotropic potential of SARS-CoV-2; all have consistently found that the virus can readily infect and replicate within COs derived from human iPSCs20, 4042, 4446. In most studies, SARS-CoV-2 preferentially infected neurons and viral particles were primarily localized to the soma, sometimes extending into neurites and axons. This infection of COs was further confirmed in two studies using electron microscopy, which detected viral particles in neurons and viral budding in the endoplasmic reticulum20, 40. An additional study using a SARS-CoV-2 pseudovirus instead of live virus also found that neurons of COs readily uptake viral particles, with a similar expression pattern47. While this evidence does not take into consideration how SARS-CoV-2 would cross the BBB to enter the brain, it does show that human brain organoids are susceptible to the SARS-CoV-2 infection.

3. Neural cell susceptibility to SARS-CoV-2

SARS-CoV-2 is primed by proteases such as transmembrane protease serine 2 (TMPRSS2) and requires expression of its receptor angiotensin-converting enzyme-2 (ACE2) to gain entry into host cells26. However, several other receptors have been identified that serve as potential receptors or co-receptors for SARS-CoV-2 infection such as neuropilin-148, CD14749, 50, CD265153, CD13353, CD20954, furin40, 50, and cadherin-1753. Therefore, studying the expression of ACE2 and other potential receptors throughout the body helps bring understanding to cell susceptibility to SARS-CoV-2 infection. Earlier studies found ACE2 mRNA expression nearly ubiquitous but its protein expression more restricted, with high levels found in the heart, kidneys, lung epithelium, small intestine, and vascular endothelium55. In the brain, neural cells include neurons, astrocytes, oligodendrocytes, microglia/macrophages, NSCs/NPCs, ependymal cells, pericyte, endothelial cells and various type of immune cells (Table 3). These neural cells also exhibit enormous diversity in their subtypes, sizes, shapes, regional locations, and functions (Figure 1b). The expression pattern of ACE2 and other potential receptors in various neural cells remains to be determined and their susceptibility to SARS-CoV-2 may vary with physiological and pathological conditions. Exploration of SARS-CoV-2 neurotropism and cellular localization is important in elucidating SARS2 neuropathogenesis and developing therapeutic and prophylactic countermeasures.

Table 3:

Neural Cell Susceptibility to SARS-CoV-2 Infection

Cell Type Receptor Expression Susceptibility to Infection Consequences of Infection in vitro Consequences of Infection in vivo
Neurons Varied expression of ACE2 (subtype and region specific) Varying reports of susceptibility Increased cell death20, 41, 44, 45; impaired synaptogenesis20, 40, 44; Tau mislocalization45 Evidence of direct SARS-CoV2 infection in neurons of K18-hACE2 and hACE2 transgenic mice20, 31, 36, 80 and select clinical SARS2 cases20, 23; Neuronal injury17, 19, 21, 22, 59, 112, 114, 121, 122, neuronal necrosis and/or neuronophagia19, 22, 115, 121, 123 detected in clinical SARS2 cases
Microglia Inconsistent reports Inconsistent reports Not available due to low susceptibility to SARS-CoV-2 infection Reactive microglia/microgliosis detected in SARS-CoV-2 infected K18-hACE2 transgenic mice36 and clinical SARS2 cases19, 59, 115, 121123, but no evidence of direct infection of microglia; Presence of microglial nodules in select SARS2 cases19, 115, 121, 123
Pericytes/ Endothelial Cells High expression of ACE255, 62, 68 and TMPRSS2 High susceptibility High levels of viral replication and shedding Vasculitis and other cerebrovascular etiologies detected in K18-hACE2 transgenic mice20, 36, 80; Acute cerebrovascular injuries in numerous clinical SARS2 cases (see Table 1)
Choroid Plexus Cells High expression of ACE2 and TMPRSS241, 46, 68, 69, 124 High susceptibility41, 46 Increased cell death, downregulation of genes related to BCSFB integrity and upregulation of inflammatory genes41; Impaired barrier function46 One clinical case report of direct SARS-CoV2 infection in endothelial cells of the choroid plexus and tanycytes of the median emminence124
NSCs/NPCs Moderate expression of ACE2; expression of TMPRSS2, furin, and cathepsin L40 Conflicting reports of low to moderate susceptibility20, 40, 44, 46 Increased cell death20, 40, 44 No evidence available
Astrocytes Weak to little ACE2 expression69 Low susceptibility36, 41, 46, 58 Not available due to low susceptibility to SARS-CoV-2 infection Reactive astrocytes/astrocytosis detected in SARS-CoV-2 infected K18-hACE2 transgenic mice36, 80 and clinical SARS2 cases17, 22, 114, 115 but no evidence of direct infection of astrocytes

a. Neurons

Both in vitro and in vivo studies have demonstrated the neuronal susceptibility to SARS-CoV-2, although the infection efficiency varied with neuronal subtypes, regional difference, and entry receptor expression patterns. Database mining showed weak expression of hACE2 in human brain cortical neurons but strong expression of CD147 and neuropilin-1. Experiments using RT-PCR, immunostaining and immunoblotting validated hACE2 expression in human neurons to various extents. The expression of entry receptors destinates neuronal infectivity of SARS-CoV-2. Human neuronal cell line U25137 and primary olfactory sensory neurons4 are susceptible to SARS-CoV-2 infection. In hamsters, one report identified high infection of primary olfactory neurons, but another study found viral infection only in sustentacular cells instead of olfactory neurons (Figure 2a)39. In mice, pervasive expression of Ace2 protein in olfactory epithelial sustentacular cells and olfactory bulb pericytes but not olfactory sensory neurons was detected by immunohistochemistry, suggesting that olfactory bulb neurons are likely not a primary site of infection, but that vascular pericytes may be sensitive to SARS-CoV-256. Interestingly, SARS-CoV-2 infected COs at Day 60 showing a significantly higher rate than at Day 15, suggesting that the virus prefers infecting more mature neural cells45 (Figure 1). Neurons in hypothalamus and with dopaminergic properties are highly susceptible to SARS-CoV-2, indicating its possibly potential affinity to some neuroantomic structures underlying pathogenesis for clinical Neuro-SARS symptoms57. The S protein-immunoreactivity is detected in brain neurons of K18-hACE2 mice after SARS-CoV-2 infection36 by intranasal delivery, suggesting the possibility that SARS-CoV-2 live virus can inffect brain neurons via the intranasal route.

Figure 2: Diagrams for some potential routes of SARS-CoV-2 neuroinvasion.

Figure 2:

There are two primary routes by which SARS-CoV-2 may invade the brain: (1) transneuronally or (2) hematogenously. Potential transneuronal routes include nasally via the olfactory system, orally via the gustatory/trigeminal nerves, spinally via the dorsal root ganglia (either via sensory receptors in the skin or in the gut), or through the enteric nervous system via the vagus nerve. Potential hematogenous routes can occur with an intact blood-brain barrier (BBB) via the choroid plexus or circumventricular organs (CVOs), or with an impaired BBB via damaged endothelial cells. (a) SARS-CoV-2 may invade the brain via the olfactory system. Viral particles enter the olfactory epithelium after passage through the nasal cavity. Within the olfactory epithelium, SARS-CoV-2 can create a reservoir within support cells, such as sustentacular cells. The virus may enter the olfactory bulb transneuronally, via axons of olfactory receptor neurons, or by alternative routes, such as infection of vascular pericytes (not pictured) or entry into the cerebrospinal fluid (CSF) in the subarachnoid space. Basal cells of the olfactory epithelium and olfactory ensheathing cells within the lamina propria and subarachnoid space are also susceptible to SARS-CoV-2 infection. (b) SARS-CoV-2 may invade the brain by crossing the BBB or blood-CSF barrier. Viral particles travel through the blood and exit through fenestrated capillaries within the choroid plexus. SARS-CoV-2 may infect choroid plexus epithelial cells and be shed into the CSF, from which they could cross into brain parenchyma. Additionally, viral particles may pass through fenestrated capillaries of CVOs directly into brain parenchyma and/or CSF. (c) SARS-CoV-2 may invade the brain by crossing the BBB. Viral particles travel through the blood and exit into the brain parenchyma by infecting or passing through damaged endothelial cells and/or their surrounding pericytes or hijacking by infected immune cells (not pictured). Within the parenchyma, SARS-CoV-2 may directly infect neural cells or elicit an immune/inflammatory response. Schematic created using BioRender.com.

b. Microglia/macrophages

Microglia in the brain play a critical role in neurodevelopment, neural innate immunity, and neuroinflammation. Microglia, like macrophages, may predominantly contribute to the inflammatory response during Neuro-SARS258, as significant activation of microglia and neuronophagia exists in the brainstem (pons, medulla oblongata), and olfactory bulb of SARS2 patients59. Whether SARS-CoV-2 can infect and replicate in microglia remain elusive. In iPSC-derived microglia, one study showed no infection of live SARS-CoV-2 virus, but another study demonstrated moderate susceptibility to S-pseudovirions42. SARS-CoV-2 has been shown to infect monocytes and macrophages, resulting in host immunoparalysis60. In human macrophages from lymph nodes and lung, SARS-CoV-2 was detected by immunohistochemistry and electron microscopy61. Whether infected macrophages infiltrate the brain, serving as a hijacking mechanism for SARS-CoV-2, remain to be determined.

c. Pericytes and endothelium cells (EC)

The blood–brain barrier (BBB) functions to protect the brain against harmful invasion such as viruses. The BBB permeability is exquisitely regulated by the neurovascular unit formed by highly specialized endothelial cells, pericytes, astrocytes, and others (Figure 2c). Early studies with immunohistochemistry showed that ACE2 expression was limited to vascular endothelial cells in the human brain55. Interestingly, a recent study demonstrated that ACE2 in the brain is not expressed in the vascular endothelial cells but instead in pericytes62. Several single cell RNA-seq studies have identified high level of ACE2 in pericytes63. Pericytes wrap themselves around vascular endothelium and play a critical role in maintaining vascular integrity. However, it is unlikely that the virus would be able to cross the BBB by passing from the blood to pericytes unless the vascular endothelium itself was damaged in the first place (Figure 2c), whether it be from pre-existing conditions, direct viral infection, or post-infection immune response. There has been direct evidence for SARS-CoV-2 infection of vascular endothelial cells and endothelialitis reported in clinical cases64. This evidence is further corroborated by a recent study in which SARS-CoV-2 was able to readily infect engineered human blood vessel organoids, which could be reduced using clinical-grade soluble ACE265. iPSC-derived endothelial cells are also susceptible to SARS-CoV-2 infection42. The severity of vasculopathy in the brain correlates with the severity of SARS2 neurological symptoms such as in stroke or mental alterations66. A clinical case report identified profound pericyte apoptosis in the lungs of SARS2 patients, which might be the initial trigger of micro-vasculopathy67. The direct evidence for SARS-CoV-2 infection in brain pericytes and the sequela is still lacking.

d. Choroid plexus epithelial cells and ependymal cells

The choroid plexus is a complex network of epithelial cells surrounding capillaries in the ventricles of the brain and is the primary region of CSF production. The epithelial cells of the choroid plexus also have a unique role as serving as the major barrier between the blood and CSF (termed the blood-CSF-barrier or BCSFB) (Figure 2b). The BCSFB acts similarly to the BBB as a selectively permeable barrier for transport to and from the brain, but likewise is also susceptible to pathogen invasion. One study looking at SARS-CoV-2 infectability in region-specific COs (including hippocampal, hypothalamic, midbrain, and cortical) (Figure 1b) found that choroid plexus epithelial cells expressed in hippocampal organoids were susceptible to SARS-CoV-2 viral infection at a much higher rate than that of neurons and other neural cell subtypes41. This led the authors to generate choroid plexus specific organoids, which displayed very high rates of infection even at low viral titer, strongly suggesting that SARS-CoV-2 may be able to invade the brain by infecting choroid plexus epithelial cells in humans. Another study validated the high susceptibility of the choroid plexus to SARS-CoV-2 S-pseudovirus and live virus in iPSC-derived brain organoids and demonstrated the leakage of BCSFB due to SARS-CoV-2-induced choroid plexus epithelial damages46. This possibility is further supported by recent evidence finding high expression of ACE2 and TMPRSS2 in the choroid plexus of both humans and mice46, 68, 69.

The ependymal cells are unique neuroepithelial cells lining the brain ventricles and are critical for CSF generation and regulation. Their basal membranes contact astrocyte endfeet via tentacle-like extensions. SARS-CoV-2 has been detected in the CSF of SARS2 patients1317, 70, 71. Whether SARS-CoV-2 in the CSF can infect ependymal cells and render neuroinvasion remains to be determined (Figure 2b). Given ependymal cells have weak or no expression of ACE269 and TMPRSS269, they may express other viral entry receptors for SARS-CoV-2, or may allow neuroinvasion under pathological conditions.

e. NSCs/NPCs

Neurogenesis occurs throughout life in both animals and humans. NSCs are self-renewing, multipotent, and long-lived cells that generate the main cell types of the nervous system. NSCs represent a small population of quiescent and slowly dividing cells, whereas their intermediate NPCs are a large population of amplifying, rapidly dividing cells. NSCs/NPCs are susceptible to various virus infections as such as ZIKV, Dengue virus, CMV, etc.43, 72. ACE2 is highly expressed in human iPSC-derived NSCs/NPCs73; thus, three studies validated infection of SARS-CoV-2 in these cells40, 41. However, another study did not detect ACE2 expression or SARS-CoV-2 infection in NSCs/NPCs46. NSCs share properties with pericytes, ependymal cells, and tanycytes. In circumventricular organs (CVO), NSCs may have direct contact with circulating pathogens. The functional states and cell fate decisions of NSCs/NPCs after SARS-CoV-2 infection warrant further investigation.

f. Astrocytes

Since astrocytes are the most abundant cell type in the CNS, their role in neural virulence has been well studied for different viruses. The endfeet of astrocytes play a key role in the formation and function of the BBB. However, SARS-CoV-2 infection efficiency is very low or absent in human primary astrocytes41, 46, 58, which may be due to high heterogeneity of astrocyte subtypes as well as weak/rare expression of ACE2 and TMPRSS269. As is the case with HIV infection in astrocytes, a minority of GFAP-positive cells infected by SARS-CoV-2 may represent NSCs. A better understanding of the similarity and diversity of SARS-CoV-2 infection between NSCs and astrocytes may provide new therapeutic routes for Neuro-SARS2.

4. Potential neuroinvasive routes for SARS-CoV-2 infection

Neurological symptoms and neuropsychiatric disorders in SARS2 patients have been extensively reported and evaluated in numerous review papers16, 9. The viral entry receptor expression and infection susceptibility of neural cells strongly support the preliminary conclusion that SARS-CoV-2 can effectively infect various types of neural cells in human brain and cause neurological/neuropsychiatric pathogenesis and symptoms. However, the routes for SARS-CoV-2 neuroinvasive infection remain to be determined. The possibility of the following potential routes is explored based on currently available literature (Figure 2).

a. Retrograde/anterograde axonal transport and transneuronal invasion:

Neurotropic viruses may invade neurons via virion trafficking in a retrograde or/and anterograde manner by interacting neuronal cytoskeletal proteins. After neuronal infection, the virions may be released and transneuronally spread to neighboring or presynaptic neurons. Many RNA viruses have been shown to invade the CNS via this transneuronal pathway including SARS-CoV-1, thus SARS-CoV-2 is extrapolated to follow similar transneuronal routes to infect the nervous system.

a-1. Intranasal epithelium and olfactory nervous system

One of the most plausible proposed routes of neuroinvasion of SARS-CoV-2 is via the olfactory system. Invasion by this route would involve the virus infecting cells of the olfactory epithelium, potentially establishing a viral reservoir (Figure 2a) 4, 74. This is because (1) olfactory epithelial cells have high ACE2 and TMPRSS2 expression68; (2) several clinical studies identified SARS-CoV-2 infection in the olfactory nerve system19, including MRI hyperintensity in the olfactory bulb of SARS2 patient75; (3) a large majority of SARS2 patients present loss of smell/taste7679; and (4) intranasal delivery of SARS-CoV-2 effectively induces brain infection in animal models80. Once cells in the olfactory epithelium are infected, the virus could either travel transneuronally via axons of olfactory receptor neurons or by non-neuronal cells up to the olfactory bulb74. However, it is still less clear if SARS-CoV-2 can infect neuronal cells within the olfactory epithelium in order to invade the brain via the olfactory bulb, as olfactory receptor neurons express relatively low levels of ACE281. If the olfactory bulb and/or olfactory nerves are infected by SARS-CoV-2, it may provide the mechanisms for the widely reported loss of smell and/or taste in SARS2 patients7679, 82. Alternatively, impaired sense of smell may be the result of an immune response in the olfactory epithelium, as these cells also co-express high levels of innate immune response genes and olfactory epithelial biopsies from SARS2 patients show significantly increased levels of proinflammatory cytokines83.

a-2. Oral health and gustatory/trigeminal nervous system

Oral health plays an important role in the severity of SARS2 development. Oral bacterial infection may increase the risk of SARS-CoV-2 infection. The majority of SARS2 patients present taste disturbances such as ageusia, dysgeusia, xerostomia82, 84. In some cases this may present as a prodromal symptom or as the sole manifestation of SARS2, particularly at the early stage85. A large cohort study which enrolled > 2 million participants concluded that the loss of smell and taste is more predictive of SARS-CoV-2 infection than all other symptoms, including fatigue, fever, or cough. Similar to olfactory neuronal terminals, the gustatory and trigeminal nerve endings may be highly susceptible to SARS-CoV-2 infection, especially at the early stage of infection (Figure 2)84. Understanding the mechanism of these neuroinvasions will provide a better strategy for the early diagnosis, prevention, and treatment of SARS2 patients, particularly in regard to neurological manifestations.

a-3. Skin and the peripheral nervous system

The human dorsal root ganglion (DRG) neurons express high levels of ACE2 at both mRNA and protein levels86. Database mining showed broad expression of entry receptors in human DRG at the lumbar and thoracic level. In a subset of nociceptive neurons, ACE2 is coexpressed with MRGPRD (Mas-related G-protein coupled receptor member D), a polymodal nociceptive receptor on the nerve endings at the outermost layers of skin and luminal organs. SARS-CoV-2 can survive on the skin for 9 hours87. Thus, SARS-CoV-2 may infect human nociceptors, causing not only peripheral pain but also neuroinvasion into the CNS via DRG sensory neurons (Figure 2). However, the direct evidence for the DRG infection is still lacking.

a-4. Gut infection and enteric nervous system

In addition to respiratory symptoms, gastrointestinal symptoms are also paramount in SARS2 patients due to abundant expression of ACE2 and other potential entry receptors in mature enterocytes88. Presence of SARS-CoV-2 viral RNA can be identified in the stool of SARS2 patients89. The intrinsic enteric nervous system is enriched with neurons and glial cells and exquisitely innervates mucosal epithelial cells. Its location makes the enteric nerve endings be readily exposed to the invading viruses. Notably, high level of ACE2 and TMPRSS2 expression have been identified in a large number of enteric neurons and enteric glia cells69. Thus, the enteric nervous system is assumed to be susceptible to SARS-CoV-2 infection. The virus infects enteric neurons and travels along with the well-established gut-brain axis such as the vagus or splanchnic nerve routing into the CNS (Figure 2). Several viruses have been shown to take the gut-brain transneuronal pathway to infect the CNS, such as herpes, varicella zoster virus, and influenza90. Additionally, infection in the enteric glial cells could induce various degree of inflammatory responses, which may aggravate systemic and neurological symptoms.

b. Hematogenous invasion to the nervous system

b-1. SARS-CoV-2 hematogenous dissemination to circumventricular organ (CVO) and choroid plexus under physiological condition

Another potential route of SARS-CoV-2 infection into the brain could be via circumventricular organs (CVOs) and/or choroid plexus, where the BBB is absent, particularly at the early stage of SARS2 wherein the BBB and BCSFB remains intact elsewhere (Figure 2b)58. The BBB under physiological conditions functions to prevent harmful pathogens from entering the brain. However, CVOs have no BBB structure, allowing the circulating pathogens, along with nutrients and metabolites, cytokines and hormones freely travel through to and out from the brain91. Specific CVOs include the median eminence, subfornical organ, and organum vasculosum lamina terminalis (around the third ventricles at the mid-level hypothalamus), as well as the area postrema in the brain stem. Direct imaging to track SARS-CoV-2 reporter virus passing through the CVO into the brain in human brain organoids or animal models is warranted.

The theory of the choroid plexus serving as an invasion route for neurotropic viruses was recently highlighted in a study showing that ZIKV can infect pericytes and cross the BCSFB via the choroid plexus in mice92. Interestingly, ZIKV’s presence in the CSF and infection of the choroid plexus preceded infection in the rest of the brain, which could be attenuated by neutralizing the virus in the CSF. If SARS-CoV-2 utilized a similar neuroinvasion mechanism, it could explain the inconsistency in CSF detection of the virus. SARS-CoV-2 may enter the brain via the CSF prior to infection of CNS neurons and then the CSF viral levels may decrease over time as it is cleared by the immune system; however, this could be when neurological symptoms begin to manifest as the virus spreads in the brain. Both CVOs and the choroid plexus are characterized by fenestrated and highly permeable capillaries (Figure 2b)58. SARS-CoV-2 in the CSF may infect ependymal cells and subsequently infect CVOs then spread to neural cells.

b-2. Transendothelial invasion via damaged BBB under pathological condition

The BBB is formed by the highly specialized endothelial cells, pericytes, and astrocytes, and is regulated by the neurovascular unit. BBB could be damaged by virus infection of any cell components of BBB or the post-infectious inflammatory responses. SARS-CoV-2 virus in the blood may directly infect brain endothelial cells and transendothelially disseminate virus into brain parenchyma via the damaged BBB, which could be aggravated by viral infection and inflammatory storm (Figure 2c)93.

b-3. Hijacking (trajectory) route via infected immune cells

Several neurotropic viruses such as HIV and Western Nile virus have been shown to enter brain through a “Trojan horse” mechanism, in which infected immune cells traffic across the permeabilized BBB and spread the hidden viruses to neurons and glial cells94. SARS-CoVs have been shown to infect monocytes/macrophages, lymphocytes, and leukocytes60, 95. Thus, SARS-CoV-2 is likely able to traffic across the BBB and BCSFB to infect neural cells93.

5. Neural cell consequence after SARS-CoV-2 infection

While elucidating the mechanisms of how SARS-CoV-2 may enter the brain is highly important to our understanding of neurological/neuropsychiatric manifestations of SARS2, it is equally critical to understand the cellular consequences that result from the virus once in the brain, such as neural cell survival/death, proliferation, migration, virus production, viral spread, etc. Several studies report increased neuronal cell death in COs after SARS-CoV-2 infection as determined by TUNNEL staining and Caspase-3 immunostaining20, 41, 44, 45. Histopathological examination of human brain autopsy specimens also characterized significant neuronal damage19. NSCs/NPCs also experience significant cell death20, 40, 44. While less data is available for glial cells, such as astrocytes and microglia, if infected, they may produce various extent of immune/inflammatory responses or neuroprotective responses. Intriguingly, cells infected with SARS-CoV-2 may promote the death of uninfected neighboring cells20, 41. Robust syncytial formation of infected cells found in the lung of SARS2 patients96 may also occur to neural cells, being a source neuropathogenesis in the brain.

Transcriptomics analysis identified large scale transcriptional dysregulation, particularly in metabolic genes and cytokine storm genes as well as genes related to hypoxia response in SARS-CoV-2-infected cells or choroid plexus organoids20, 41, 44, 45, confirming the local hypoxic environment in neurons surrounding SARS-CoV-2 infected cells via HIF1α expression. The upregulation of vascular remodeling genes and the downregulation of genes related to CSF secretory function as well as cell junction genes suggest potentially impaired function of the BCSFB and BBB.

Significantly decreased expression of vGLUT1, an excitatory pre-synapse marker, has been identified in human brain organoids, suggesting impaired synaptogenesis in SARS-CoV-2 infected cells20, 40, 44. Another study identified Tau mislocalization in the neuronal soma of SARS-CoV-2-infected mature neurons as opposed to the typical localization within axons of normal neurons45. Further investigation showed that the phospho-Tau mislocalization is specific to pT231Tau, which correlated with a fraction of cells that also expressed caspase-3. Thus, SARS-CoV-2 infection results in aberrant pT231Tau phosphorylation, which may trigger programmed cell death pathways. Intriguingly, the neuronal apoptosis induced by SARS-CoV-2 infection appears dependent on Type II interferon response (IFNγ), but not Type I interferon response (IFNα/β)20, 41, 44, 45.

6. Conclusions and future directions

Accumulated evidence from clinical observations and laboratory studies helps approach to understanding the neuropathogenesis underlying SARS-CoV-2 infection causing various neurological and neuropsychiatric conditions in SARS2 patients. It remains to be determined whether SARS-CoV-2 directly or indirectly induces neurological/psychiatric damages. Studies have shown that many of SARS2 patients with an active neurological symptom had no detectable virus in the CSF. In addition, no virus could be recovered from the brain tissues of patients who died of SARS2. These observations suggest the possibility that indirect effects such as inflammation induced by SARS-Cov-2 infection contribute to the neurological/neuropsychiatric manifestations. However, failure to detect the virus may be due, at least partly, to low sensitivity of the assays; stages of infection and timing when the samples were examined. Notably, studies from different groups reported the significant differences in SARS-CoV-2 infection of various neural cells, which may be responsible for the conflicting data reported. Hopefully, with the availability of more advanced technologies such as RNAscope55, 56, digital droplet PCR5759, viral outgrowth assay60, 61, single cell RNA sequencing62, 63, and multilabeled immunohistochemical analysis as well as better in vivo and in vitro models such as preclinical animals, human induced pluripotent stem cells, and human brain organoids for various neural cells, we will be able to determine the mechanisms for SARS-Cov-2 infection-caused neurological injury, which is crucial in developing strategies for treatment of SARS2 patients with the neurological/neuropsychiatric manifestations. Although several new studies using brain organoids offer strong and straightforward evidence for the neuroinvasive effect on different neural cells with particularly high susceptibility of choroid plexus epithelial cells, there are still limitations of current brain organoid technology to recapitulate clinical patients, including immaturity of neural cells, unclear dose of neurovirulent infection in vivo, and lack of additional cell types such as microglia, pericytes, endothelial cells, oligodendrocytes, and immune cells41. Given that the SARS2 pandemic remains raging and seasonal resurgence is impending, more extended efforts and investigation into Neuro-SARS2 mechanisms remain urgently needed. Long-term follow-up for potential neurological/neuropsychiatric manifestations as well as development of novel prophylactic and therapeutic treatments for Neuro-SARS2 are equally critical.

Acknowledgement:

This work was supported by the National Institute of Health 1R01DA050505-01 (WZH and WHH) and 1R01AI145034 (WHH). The content of this review article is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health. All authors have read the journal’s authorship agreement and policy on disclosure of potential conflicts of interest. All authors declare that they have no conflict of interest. No English writing service was sought for this manuscript.

Abbreviations:

ACE2

angiotensin-converting enzyme-2

BBB

blood-brain barrier

BCSFB

blood-CSF barrier

CoV

coronavirus

COVID-19

coronavirus disease 2019

CNS

central nervous system

CO

cerebral organoid

CSF

cerebrospinal fluid

CVO

circumventricular organ

DRG

dorsal root ganglion

iPSC

induced pluripotent stem cell

K18

human cytokeratin 18

MERS

Middle East respiratory syndrome

MERS-CoV

Middle East respiratory syndrome coronavirus

Neuro-SARS

neurological/neuropsychiatric manifestation of severe acute respiratory syndrome

NPC

neural progenitor cell

NSC

neural stem cell

PNS

peripheral nervous system

RT-PCR

reverse transcription polymerase chain reaction

SARS

severe acute respiratory syndrome

SARS-CoV

severe acute respiratory syndrome coronavirus

TMPRSS2

transmembrane protease serine 2

ZIKV

Zika virus

Footnotes

Conflict of interest

The authors declare that there are no conflict of interests.

Data availability statement

The data that support the findings of this study are available from the corresponding author upon reasonable request.

References

  • [1].Assari S COVID-19 Pandemic and Neurological Disease: A Critical Review of the Existing Literature. Hosp Pract Res. 2020;5:81–6. [PubMed] [Google Scholar]
  • [2].Ellul MA, Benjamin L, Singh B, et al. Neurological associations of COVID-19 Lancet Neurol. 2020;19:767–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [3].Kumar M, Thakur AK. Neurological manifestations and comorbidity associated with COVID-19: an overview. Neurol Sci. 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [4].Cooper KW, Brann DH, Farruggia MC, et al. COVID-19 and the Chemical Senses: Supporting Players Take Center Stage Neuron. 2020;107:219–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [5].Iadecola C, Anrather J, Kamel H. Effects of COVID-19 on the Nervous System Cell. 2020;183:16–27 e1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [6].Oliveira R, Sotero FD, Teodoro T. NeuroCOVID: critical review of neuropsychiatric manifestations of SARS-CoV-2 infection Ir J Med Sci. 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [7].Sinanovic O, Muftic M, Sinanovic S. COVID-19 Pandemia: Neuropsychiatric Comorbidity and Consequences. Psychiatr Danub. 2020;32:236–44. [DOI] [PubMed] [Google Scholar]
  • [8].Roy D, Ghosh R, Dubey S, et al. Neurological and Neuropsychiatric Impacts of COVID-19 Pandemic Can J Neurol Sci. 2020:1–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [9].Nalleballe K, Reddy Onteddu S, Sharma R, et al. Spectrum of neuropsychiatric manifestations in COVID-19. Brain Behav Immun. 2020;88:71–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [10].Lau K-K, Yu W-C, Chu C-M, et al. Possible Central Nervous System Infection by SARS Coronavirus Emerging Infectious Diseases. 2004;10:342–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [11].Gu J, Gong E, Zhang B, et al. Multiple organ infection and the pathogenesis of SARS Journal of Experimental Medicine. 2005;202:415–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [12].Kim J-E, Heo J-H, Kim H-o, et al. Neurological Complications during Treatment of Middle East Respiratory Syndrome Journal of Clinical Neurology. 2017;13:227–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [13].Andriuta D, Roger P-A, Thibault W, et al. COVID-19 encephalopathy: detection of antibodies against SARS-CoV-2 in CSF Journal of Neurology. 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [14].Cebrián J, Gonzalez-Martinez A, García-Blanco MJ, et al. Headache and impaired consciousness level associated with SARS-CoV-2 in CSF. Neurology. 2020;95:266–8. [DOI] [PubMed] [Google Scholar]
  • [15].Fadakar N, Ghaemmaghami S, Masoompour SM, et al. A First Case of Acute Cerebellitis Associated with Coronavirus Disease (COVID-19): a Case Report and Literature Review. The Cerebellum. 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [16].Kremer S, Lersy F, de Sèze J, et al. Brain MRI Findings in Severe COVID-19: A Retrospective Observational Study. Radiology. 2020:202222–. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [17].Virhammar J, Kumlien E, Fällmar D, et al. Acute necrotizing encephalopathy with SARS-CoV-2 RNA confirmed in cerebrospinal fluid Neurology. 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [18].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]
  • [19].Solomon IH, Normandin E, Bhattacharyya S, et al. Neuropathological Features of Covid-19 N Engl J Med. 2020;383:989–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [20].Song E, Zhang C, Israelow B, et al. Neuroinvasion of SARS-CoV-2 in human and mouse brain. bioRxiv. 2020:2020.06.25.169946. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [21].Hernández-Fernández F, Sandoval Valencia H, Barbella-Aponte RA, et al. Cerebrovascular disease in patients with COVID-19: neuroimaging, histological and clinical description Brain. 2020;143:3089–103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [22].Reichard RR, Kashani KB, Boire NA, et al. Neuropathology of COVID-19: a spectrum of vascular and acute disseminated encephalomyelitis (ADEM)-like pathology. Acta Neuropathologica. 2020;140:1–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [23].Paniz-Mondolfi A, Bryce C, Grimes Z, et al. Central nervous system involvement by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). Journal of Medical Virology. 2020;92:699–702. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [24].Carossino M, Ip HS, Richt JA, et al. Detection of SARS-CoV-2 by RNAscope((R)) in situ hybri dization and immunohistochemistry techniques Arch Virol. 2020;165:2373–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [25].Abdelaziz OS, Waffa Z. Neuropathogenic human coronaviruses: A review. Rev Med Virol. 2020;30:e2118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [26].Hoffmann M, Kleine-Weber H, Schroeder S, et al. SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor Cell. 2020:1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [27].Alexander MR, Schoeder CT, Brown JA, et al. Predicting susceptibility to SARS-CoV-2 infection based on structural differences in ACE2 across species FASEB J. 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [28].Cleary SJ, Pitchford SC, Amison RT, et al. Animal models of mechanisms of SARS-CoV-2 infection and COVID-19 pathology Br J Pharmacol. 2020;177:4851–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [29].Rockx B, Kuiken T, Herfst S, et al. Comparative pathogenesis of COVID-19, MERS, and SARS in a nonhuman primate model Science. 2020;368:1012–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [30].Munster VJ, Feldmann F, Williamson BN, et al. Respiratory disease in rhesus macaques inoculated with SARS-CoV-2 Nature. 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [31].Sun SH, Chen Q, Gu HJ, et al. A Mouse Model of SARS-CoV-2 Infection and Pathogenesis. Cell Host Microbe. 2020;28:124–33 e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [32].Rathnasinghe R, Strohmeier S, Amanat F, et al. Comparison of transgenic and adenovirus hACE2 mouse models for SARS-CoV-2 infection Emerg Microbes Infect. 2020;9:2433–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [33].Moreau GB, Burgess SL, Sturek JM, et al. Evaluation of K18-hACE2 Mice as a Model of SARS-CoV-2 Infection Am J Trop Med Hyg. 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [34].Jiang R-D, Liu M-Q, Chen Y, et al. Pathogenesis of SARS-CoV-2 in Transgenic Mice Expressing Human Angiotensin-Converting Enzyme 2 Cell. 2020;182:50–8.e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [35].Winkler ES, Bailey AL, Kafai NM, et al. SARS-CoV-2 infection of human ACE2-transgenic mice causes severe lung inflammation and impaired function Nature Immunology. 2020:1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [36].Golden JW, Cline CR, Zeng X, et al. Human angiotensin-converting enzyme 2 transgenic mice infected with SARS-CoV-2 develop severe and fatal respiratory disease. JCI Insight. 2020;5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [37].Chu H, Chan JF, Yuen TT, et al. Comparative tropism, replication kinetics, and cell damage profiling of SARS-CoV-2 and SARS-CoV with implications for clinical manifestations, transmissibility, and laboratory studies of COVID-19: an observational study. Lancet Microbe. 2020;1:e14–e23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [38].Zhang AJ, Lee AC, Chu H, et al. SARS-CoV-2 infects and damages the mature and immature olfactory sensory neurons of hamsters Clin Infect Dis. 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [39].Bryche B, St Albin A, Murri S, et al. Massive transient damage of the olfactory epithelium associated with infection of sustentacular cells by SARS-CoV-2 in golden Syrian hamsters Brain Behav Immun. 2020;89:579–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [40].Zhang BZ, Chu H, Han S, et al. SARS-CoV-2 infects human neural progenitor cells and brain organoids Cell Res. 2020;30:928–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [41].Jacob F, Pather SR, Huang W-K, et al. Human Pluripotent Stem Cell-Derived Neural Cells and Brain Organoids Reveal SARS-CoV-2 Neurotropism Predominates in Choroid Plexus Epithelium. Cell Stem Cell. 2020;27:1–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [42].Yang L, Han Y, Nilsson-Payant BE, et al. A Human Pluripotent Stem Cell-based Platform to Study SARS-CoV-2 Tropism and Model Virus Infection in Human Cells and Organoids. Cell Stem Cell. 2020;27:125–36 e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [43].Qian X, Nguyen HN, Song MM, et al. Brain-Region-Specific Organoids Using Mini-bioreactors for Modeling ZIKV Exposure Cell. 2016;165:1238–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [44].Mesci P, Macia A, Saleh A, et al. Sofosbuvir protects human brain organoids against SARS-CoV-2. bioRxiv. 2020. [Google Scholar]
  • [45].Ramani A, Muller L, Ostermann PN, et al. SARS-CoV-2 targets neurons of 3D human brain organoids EMBO J. 2020;39:e106230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [46].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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [47].Yi SA, Nam KH, Yun J, et al. Infection of Brain Organoids and 2D Cortical Neurons with SARS-CoV-2 Pseudovirus. Viruses. 2020;12:1004–. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [48].Cantuti-Castelvetri L, Ojha R, Pedro LD, et al. Neuropilin-1 facilitates SARS-CoV-2 cell entry and infectivity Science. 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [49].Helal MA, Shouman S, Abdelwaly A, et al. Molecular basis of the potential interaction of SARS-CoV-2 spike protein to CD147 in COVID-19 associated-lymphopenia J Biomol Struct Dyn. 2020:1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [50].Latini A, Agolini E, Novelli A, et al. COVID-19 and Genetic Variants of Protein Involved in the SARS-CoV-2 Entry into the Host Cells. Genes (Basel). 2020;11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [51].Strollo R, Pozzilli P. DPP4 inhibition: Preventing SARS-CoV-2 infection and/or progression of COVID-19? Diabetes Metab Res Rev. 2020;36:e3330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [52].Raha AA, Chakraborty S, Henderson J, et al. Investigation of CD26, a potential SARS-CoV-2 receptor, as a biomarker of age and pathology Biosci Rep. 2020;40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [53].Kotani N, Nakano T. Candidate screening of host cell membrane proteins involved in SARS-CoV-2 entry. BioRxiv. 2020. [Google Scholar]
  • [54].Amraie R, Napoleon MA, Yin W, et al. CD209L/L-SIGN and CD209/DC-SIGN act as receptors for SARS-CoV-2 and are differentially expressed in lung and kidney epithelial and endothelial cells. bioRxiv. 2020. [Google Scholar]
  • [55].Hamming I, Timens W, Bulthuis MLC, et al. Tissue distribution of ACE2 protein, the functional receptor for SARS coronavirus. A first step in understanding SARS pathogenesis The Journal of Pathology. 2004;203:631–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [56].Brann DH, Tsukahara T, Weinreb C, et al. Non-neuronal expression of SARS-CoV-2 entry genes in the olfactory system suggests mechanisms underlying COVID-19-associated anosmia. Sci Adv. 2020;6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [57].Chiappelli F Towards Neuro-CoViD-19. Bioinformation. 2020;16:288–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [58].Murta V, Villarreal A, Ramos AJ. Severe Acute Respiratory Syndrome Coronavirus 2 Impact on the Central Nervous System: Are Astrocytes and Microglia Main Players or Merely Bystanders? ASN Neuro. 2020;12:1759091420954960. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [59].Deigendesch N, Sironi L, Kutza M, et al. Correlates of critical illness-related encephalopathy predominate postmortem COVID-19 neuropathology Acta Neuropathol. 2020;140:583–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [60].Merad M, Martin JC. Pathological inflammation in patients with COVID-19: a key role for monocytes and macrophages Nat Rev Immunol. 2020;20:355–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [61].Martines RB, Ritter JM, Matkovic E, et al. Pathology and Pathogenesis of SARS-CoV-2 Associated with Fatal Coronavirus Disease, United States Emerg Infect Dis. 2020;26:2005–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [62].He L, Mäe MA, Sun Y, et al. Pericyte-specific vascular expression of SARS-CoV-2 receptor ACE2- implications for microvascular inflammation and hypercoagulopathy in COVID-19 patients. bioRxiv. 2020. [Google Scholar]
  • [63].Ziegler CGK, Allon SJ, Nyquist SK, et al. SARS-CoV-2 Receptor ACE2 Is an Interferon-Stimulated Gene in Human Airway Epithelial Cells and Is Detected in Specific Cell Subsets across Tissues Cell. 2020;181:1016–35 e19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [64].Varga Z, Flammer AJ, Steiger P, et al. Endothelial cell infection and endotheliitis in COVID-19 The Lancet. 2020;395:1417–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [65].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;181:905–13.e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [66].Avula A, Nalleballe K, Narula N, et al. COVID-19 presenting as stroke Brain Behav Immun. 2020;87:115–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [67].Cardot-Leccia N, Hubiche T, Dellamonica J, Burel-Vandenbos F, Passeron T. Pericyte alteration sheds light on micro-vasculopathy in COVID-19 infection. Intensive Care Med. 2020;46:1777–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [68].Chen R, Wang K, Yu J, et al. The spatial and cell-type distribution of SARS-CoV-2 receptor ACE2 in human and mouse brain. bioRxiv. 2020:2020.04.07.030650–2020.04.07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [69].Deffner F, Scharr M, Klingenstein S, et al. Histological Evidence for the Enteric Nervous System and the Choroid Plexus as Alternative Routes of Neuroinvasion by SARS-CoV2. Front Neuroanat. 2020;14:596439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [70].Domingues RB, Mendes-Correa MC, de Moura Leite FBV, et al. First case of SARS-COV-2 sequencing in cerebrospinal fluid of a patient with suspected demyelinating disease Journal of Neurology. 2020:2–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [71].Huang YH, Jiang D, Huang JT. SARS-CoV-2 Detected in Cerebrospinal Fluid by PCR in a Case of COVID-19 Encephalitis Brain, Behavior, and Immunity. 2020;87:149–. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [72].Muffat J, Li Y, Omer A, et al. Human induced pluripotent stem cell-derived glial cells and neural progenitors display divergent responses to Zika and dengue infections Proc Natl Acad Sci U S A. 2018;115:7117–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [73].Kase Y, Okano H. Expression of ACE2 and a viral virulence-regulating factor CCN family member 1 in human iPSC-derived neural cells: implications for COVID-19-related CNS disorders. Inflamm Regen. 2020;40:32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [74].Butowt R, Bilinska K. SARS-CoV-2: Olfaction, Brain Infection, and the Urgent Need for Clinical Samples Allowing Earlier Virus Detection. ACS Chem Neurosci. 2020;11:1200–3. [DOI] [PubMed] [Google Scholar]
  • [75].Politi LS, Salsano E, Grimaldi M. Magnetic Resonance Imaging Alteration of the Brain in a Patient With Coronavirus Disease 2019 (COVID-19) and Anosmia. JAMA Neurology. 2020;77:1028–9. [DOI] [PubMed] [Google Scholar]
  • [76].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 Clinical Infectious Diseases. 2020;71:889–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [77].Kaye R, Chang CWD, Kazahaya K, Brereton J, Denneny JC. COVID-19 Anosmia Reporting Tool: Initial Findings. Otolaryngology–Head and Neck Surgery. 2020;163:132–4. [DOI] [PubMed] [Google Scholar]
  • [78].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. European Archives of Oto-Rhino-Laryngology. 2020;277:2251–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [79].Spinato G, Fabbris C, Polesel J, et al. Alterations in Smell or Taste in Mildly Symptomatic Outpatients With SARS-CoV-2 Infection. JAMA. 2020;323:2089–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [80].Yinda CK, Port JR, Bushmaker T, et al. K18-hACE2 mice develop respiratory disease resembling severe COVID-19. bioRxiv. 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [81].Bilinska K, Jakubowska P, Von Bartheld CS, Butowt R. Expression of the SARS-CoV-2 Entry Proteins, ACE2 and TMPRSS2, in Cells of the Olfactory Epithelium: Identification of Cell Types and Trends with Age. ACS Chem Neurosci. 2020;11:1555–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [82].Parma V, Ohla K, Veldhuizen MG, et al. More Than Smell-COVID-19 Is Associated With Severe Impairment of Smell, Taste, and Chemesthesis Chem Senses. 2020;45:609–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [83].Torabi A, Mohammadbagheri E, Akbari Dilmaghani N, et al. Proinflammatory Cytokines in the Olfactory Mucosa Result in COVID-19 Induced Anosmia. ACS Chemical Neuroscience. 2020;11:1909–13. [DOI] [PubMed] [Google Scholar]
  • [84].Samaranayake LP, Fakhruddin KS, Panduwawala C. Sudden onset, acute loss of taste and smell in coronavirus disease 2019 (COVID-19): a systematic revi ew Acta Odontol Scand. 2020;78:467–73. [DOI] [PubMed] [Google Scholar]
  • [85].Biadsee A, Biadsee A, Kassem F, et al. Olfactory and Oral Manifestations of COVID-19: Sex-Related Symptoms-A Potential Pathway to Early Diagnosis. Otolaryngol Head Neck Surg. 2020;163:722–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [86].Shiers S, Ray PR, Wangzhou A, et al. ACE2 and SCARF expression in human dorsal root ganglion nociceptors: implications for SARS-CoV-2 virus neurological effects. Pain. 2020;161:2494–501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [87].Hirose R, Ikegaya H, Naito Y, et al. Survival of SARS-CoV-2 and influenza virus on the human skin: Importance of hand hygiene in COVID-19. Clin Infect Dis. 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [88].Agarwal A, Chen A, Ravindran N, To C, Thuluvath PJ. Gastrointestinal and Liver Manifestations of COVID-19. J Clin Exp Hepatol. 2020;10:263–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [89].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;159:81–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [90].Shahbazi R, Yasavoli-Sharahi H, Alsadi N, Ismail N, Matar C. Probiotics in Treatment of Viral Respiratory Infections and Neuroinflammatory Disorders. Molecules. 2020;25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [91].Ferguson AV. Circumventricular Organs: Integrators of Circulating Signals Controlling Hydration, Energy Balance, and Immune Function In: De Luca LA Jr., Menani JV, Johnson AK, editors. Neurobiology of Body Fluid Homeostasis: Transduction and Integration. Boca Raton (FL)2014. [PubMed] [Google Scholar]
  • [92].Kim J, Alejandro B, Hetman M, et al. Zika virus infects pericytes in the choroid plexus and enters the central nervous system through the blood-cerebrospinal fluid barrier. PLOS Pathogens. 2020;16:e1008204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [93].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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [94].Kramer-Hammerle S, Rothenaigner I, Wolff H, Bell JE, Brack-Werner R. Cells of the central nervous system as targets and reservoirs of the human immunodeficiency virus. Virus Res. 2005;111:194–213. [DOI] [PubMed] [Google Scholar]
  • [95].Codo AC, Davanzo GG, Monteiro LB, et al. Elevated Glucose Levels Favor SARS-CoV-2 Infection and Monocyte Response through a HIF-1alpha/Glycolysis-Dependent Axis. Cell Metab. 2020;32:437–46 e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [96].Oprinca GC, Muja LA. Postmortem examination of three SARS-CoV-2-positive autopsies including histopathologic and immunohistochemical analysis. Int J Legal Med. 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [97].Kacem I, Gharbi A, Harizi C, et al. Characteristics, onset, and evolution of neurological symptoms in patients with COVID-19. Neurological Sciences. 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [98].Liotta EM, Batra A, Clark JR, et al. Frequent neurologic manifestations and encephalopathy-associated morbidity in Covid-19 patients. Annals of Clinical and Translational Neurology. 2020:acn3.51210–acn3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [99].Mao L, Jin H, Wang M, et al. Neurologic Manifestations of Hospitalized Patients With Coronavirus Disease 2019 in Wuhan, China. JAMA Neurology. 2020;77:683–. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [100].Chen N, Zhou M, Dong X, et al. Epidemiological and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: a descriptive study. The Lancet. 2020;395:507–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [101].Chen T, Wu D, Chen H, et al. Clinical characteristics of 113 deceased patients with coronavirus disease 2019: retrospective study. BMJ. 2020;368:m1091–m. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [102].Huang C, Wang Y, Li X, et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. The Lancet. 2020;395:497–506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [103].Klopfenstein T, Kadiane-Oussou NJ, Toko L, et al. Features of anosmia in COVID-19. Médecine et Maladies Infectieuses. 2020;50:436–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [104].Lechien JR, Chiesa-Estomba CM, Place S, et al. Clinical and epidemiological characteristics of 1420 European patients with mild-to-moderate coronavirus disease 2019. Journal of Internal Medicine. 2020;288:335–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [105].Wang D, Hu B, Hu C, et al. Clinical Characteristics of 138 Hospitalized Patients With 2019 Novel Coronavirus–Infected Pneumonia in Wuhan, China. JAMA. 2020;323:1061–. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [106].Benussi A, Pilotto A, Premi E, et al. Clinical characteristics and outcomes of inpatients with neurologic disease and COVID-19 in Brescia, Lombardy, Italy. Neurology. 2020;95:e910–e20. [DOI] [PubMed] [Google Scholar]
  • [107].Paterson RW, Brown RL, Benjamin LA, et al. The emerging spectrum of COVID-19 neurology: clinical, radiological and laboratory findings. Brain. 2020;143:3104–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [108].Varatharaj A, Thomas N, Ellul MA, et al. Neurological and neuropsychiatric complications of COVID-19 in 153 patients: a UK-wide surveillance study. The Lancet Psychiatry. 2020;7:875–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [109].Rogers JP, Chesney E, Oliver D, et al. Psychiatric and neuropsychiatric presentations associated with severe coronavirus infections: a systematic review and meta-analysis with comparison to the COVID-19 pandemic. The Lancet Psychiatry 2020;7:611–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [110].Dalakas MC. Guillain-Barré syndrome: The first documented COVID-19–triggered autoimmune neurologic disease. Neurology: Neuroimmunology & Neuroinflammation. 2020;7:e781–e. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [111].Poyiadji N, Shahin G, Noujaim D, et al. COVID-19–associated Acute Hemorrhagic Necrotizing Encephalopathy: Imaging Features. Radiology. 2020;296:E119–E20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [112].von Weyhern CH, Kaufmann I, Neff F, Kremer M. Early evidence of pronounced brain involvement in fatal COVID-19 outcomes. The Lancet. 2020;395:e109–e. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [113].Li Y, Li M, Wang M, et al. Acute cerebrovascular disease following COVID-19: a single center, retrospective, observational study. Stroke and Vascular Neurology. 2020;5:279–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [114].Kanberg N, Ashton NJ, Andersson L-M, et al. Neurochemical evidence of astrocytic and neuronal injury commonly found in COVID-19. Neurology. 2020;95:e1754–e9. [DOI] [PubMed] [Google Scholar]
  • [115].Matschke J, Lütgehetmann M, Hagel C, et al. Neuropathology of patients with COVID-19 in Germany: a post-mortem case series. The Lancet Neurology. 2020;19:919–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [116].Zheng J, Roy Wong L-Y, Li K, et al. COVID-19 treatments and pathogenesis including anosmia in K18-hACE2 mice. Nature. 2020:2020.08.07.242073–2020.08.07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [117].Bao L, Deng W, Huang B, et al. The pathogenicity of SARS-CoV-2 in hACE2 transgenic mice. Nature. 2020;583:830–3. [DOI] [PubMed] [Google Scholar]
  • [118].Hassan AO, Case JB, Winkler ES, et al. A SARS-CoV-2 Infection Model in Mice Demonstrates Protection by Neutralizing Antibodies. Cell. 2020;182:744–53.e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [119].Sun J, Zhuang Z, Zheng J, et al. Generation of a Broadly Useful Model for COVID-19 Pathogenesis, Vaccination, and Treatment. Cell. 2020;182:734–43.e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [120].Bullen CK, Hogberg HT, Bahadirli-Talbott A, et al. Infectability of Human BrainSphere Neurons Suggests Neurotropism of SARS-CoV-2. ALTEX. 2020. [DOI] [PubMed] [Google Scholar]
  • [121].Al-Dalahmah O, Thakur KT, Nordvig AS, et al. Neuronophagia and microglial nodules in a SARS-CoV-2 patient with cerebellar hemorrhage. Acta Neuropathol Commun. 2020;8:147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [122].Kantonen J, Mahzabin S, Mäyränpää MI, et al. Neuropathologic features of four autopsied COVID-19 patients. Brain Pathology. 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [123].Jensen MP, Le Quesne J, Officer-Jones L, et al. Neuropathological Findings in Two Patients with Fatal COVID-19. Neuropathology and Applied Neurobiology. 2020. [DOI] [PubMed] [Google Scholar]
  • [124].Nampoothiri S, Sauve F, Ternier G, et al. The hypothalamus as a hub for SARS-CoV-2 brain infection and pathogenesis. bioRxiv. 2020:2020.06.08.139329. [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

The data that support the findings of this study are available from the corresponding author upon reasonable request.

RESOURCES