You are on page 1of 17

Received: 10 November 2020 | Revised: 7 December 2020 | Accepted: 8 December 2020

DOI: 10.1002/jmv.26720

REVIEW

Cellular mechanisms underlying neurological/


neuropsychiatric manifestations of COVID‐19

Brittany Bodnar1,2,3 | Kena Patel1,2,4 | Wenzhe Ho1 | Jin Jun Luo5 |


Wenhui Hu1,2,3

1
Department of Pathology and Laboratory
Medicine, Lewis Katz School of Medicine, Abstract
Temple University, Philadelphia,
Patients with severe acute respiratory syndrome coronavirus‐2 (SARS‐CoV‐2) infection
Pennsylvania, USA
2
Center for Metabolic Disease Research,
manifest mainly respiratory symptoms. However, clinical observations frequently iden-
Lewis Katz School of Medicine, Temple tified neurological symptoms and neuropsychiatric disorders related to COVID‐19
University, Philadelphia, Pennsylvania, USA
(Neuro‐SARS2). Accumulated robust evidence indicates that Neuro‐SARS2 may play an
3
Biomedical Sciences Graduate Program,
Lewis Katz School of Medicine, Temple important role in aggravating the disease severity and mortality. Understanding the
University, Philadelphia, Pennsylvania, USA neuropathogenesis and cellular mechanisms underlying Neuro‐SARS2 is crucial for both
4
Undergraduate Research Program, College basic research and clinical practice to establish effective strategies for early detection/
of Science and Technology, Temple
University, Philadelphia, Pennsylvania, USA diagnosis, prevention, and treatment. In this review, we comprehensively examine cur-
5
Department of Neurology and Department rent evidence of SARS‐CoV‐2 infection in various neural cells including neurons, micro-
of Pharmacology, Lewis Katz School of
glia/macrophages, astrocytes, pericytes/endothelial cells, ependymocytes/choroid
Medicine, Temple University, Philadelphia,
Pennsylvania, USA 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
Correspondence
Wenhui Hu, Center for Metabolic Disease
routes (transneuronal and hematogenous) of the virus and the cellular/molecular
Research, Department of Pathology and mechanisms underlying the development/progression of this disease. Future and ongoing
Laboratory Medicine, Temple University
Lewis Katz School of Medicine, 3500 N Broad
studies require the establishment of more clinically relevant and suitable neural cell
St, Philadelphia, PA 19140, USA. models using human induced pluripotent stem cells, brain organoids, and postmortem
Email: whu@temple.edu
specimens.

Funding information
KEYWORDS
NIH, Grant/Award Numbers: R01AI145034,
brain organoids, COVID‐19, iPS cells, neural cells, neuroinvasion, neurology, neuropsychiatry,
R01DA050505
neurovirulence, SARS‐CoV‐2

1 | INTRODUCTION coronavirus), and large numbers of new cases still occur daily. The
unprecedented scale of the pandemic continues to push the bound-
The severe acute respiratory syndrome coronavirus‐2 (SARS‐CoV‐2) aries of scientific research and development as the community pro-
virus has caused a global pandemic affecting 218 countries and ceeds to further understand the virus and its complexity involving
territories with 51,791,296 confirmed cases and 1,278,460 con- multiple organ systems with varying severities. The most common
firmed deaths, as of November 10, 2020 (www.worldometers.info/ and predominant presentations of illness are respiratory symptoms.

Abbreviations: ACE2, angiotensin‐converting enzyme‐2; BBB, blood–brain barrier; BCSFB, blood‐CSF barrier; CNS, central nervous system; CO, cerebral organoid; CoV, coronavirus;
COVID‐19, coronavirus disease 2019; 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.

J Med Virol. 2021;93:1983–1998. wileyonlinelibrary.com/journal/jmv © 2020 Wiley Periodicals LLC | 1983


1984 | BODNAR ET AL.

However, there are increasing reports of neurological and neu- syndrome, and myopathies (manifested as rhabdomyolysis).10
ropsychiatric symptoms.1–8 Such symptoms may be nonspecific, such Autopsies from several SARS1 patients revealed the presence of
as somatoform disorders including muscle soreness, dizziness, head- SARS‐CoV‐1 in the brain, primarily in the cytoplasm of neurons in the
ache, anxiety, sleep disturbances, depression, fatigue, anger/stress, cortex and hypothalamus.11 Additionally, SARS‐CoV‐1 could be de-
loneliness, stigmatization; or specific, such as central nervous system tected in the cerebrospinal fluid (CSF) of SARS1 patients who were
(CNS) disorders including stroke, seizures, encephalitis, ataxia, and experiencing seizures.10 Similarly, neurological complications were
myelitis; peripheral nervous system (PNS) disorders including loss of reported in some patients with MERS, including peripheral neuro-
smell and taste (anosmia/hyposmia and ageusia/dysgeusia), Guillain‐ pathies, encephalopathies, intracerebral hemorrhage, stroke, sei-
Barré syndrome and Miller Fisher syndrome; or psychiatric and neu- zures, and confusion.12 However, there is no confirmatory evidence
ropsychiatric syndromes including major depressive disorder, bipolar from post‐mortem or CSF viral detection studies to support
disorder, obsessive‐compulsive disorder and posttraumatic stress MERS‐CoV directly infecting the brain.
disorder. The exact mechanisms of SARS‐CoV‐2 underlying neurolo- In SARS2 cases, SARS‐CoV‐2 has been detected by reverse‐
gical/neuropsychiatric dysfunctions are not fully understood but seem transcription polymerase chain reaction (RT‐PCR) assay in the CSF of
to be due to a combination of potentially direct viral infection, or numerous patients with various neurological symptoms13–17 al-
indirect systemic disease complication plus para/post‐infectious in- though many anecdotal clinical reports showed a negative test for
flammation in the nervous system. Notably, knowledge of the long‐ SARS‐CoV‐2 by RT‐PCR assays of CSF samples.18 The reason why no
term effects of SARS‐CoV‐2 on the nervous system in surviving viral load was detectable in the CSF of the majority of SARS2 pa-
COVID‐19 patients is virtually absent. tients with an active neurological condition remains unclear. This
In this review, we focus on the current understanding of cellular discrepancy could be due to the differences in sensitivity of RT‐PCR
mechanisms underlying COVID‐19 neurological/neuropsychiatric assays, the stages of infection, and the timing when the CSF sample
pathogenesis and symptoms. Accumulated evidence has shown that is manipulated and the assay is performed, or the clearance of
SARS‐CoV‐2 may directly infect various neural cells. Additionally, SARS‐CoV‐2 from the CSF. Nonetheless, detection of SARS‐CoV‐2
systemic immune activation and chronic neuroinflammation may also by RT‐PCR in CSF from patients with active neurological symptoms
contribute to neurological dysfunctions. Although severe neurological has provided undeniable evidence that SARS‐CoV‐2 could invade the
complications seem to be rare, the global scale of the pandemic can nervous system. Undoubtedly, more studies are needed to secure
amount to a large number of cases in the long‐term as symptoms and convincible evidence.
latent effects of the virus continue to manifest, potentially leaving A case series study of 18 post‐mortem SARS2 patients with
patients with prolonged existing neurological and/or neuropsychiatric various neurological symptoms has shown evidence of acute hypoxic‐
disturbances.7,8 Presumably, neurological and neuropsychiatric man- ischemic damages in the brains of all patients, including neuronal
ifestations in surviving COVID‐19 patients may have equal, if not loss.19 Of these patients, the virus was detected by RT‐PCR in five
more significant, adverse consequences when compared to pulmon- brains. SARS‐CoV‐2 spike (S) protein was detected in cortical neu-
ary manifestations. Understanding the cellular mechanisms will pave rons and endothelial cells in SARS2 brain samples20 but not the viral
the way for basic research directions and clinical therapeutic stra- nucleocapsid (N) protein by immunohistochemical analyses.19
tegies. For better clarity and understanding, we tentatively assign Other neuropathological case studies have found histological
SARS1 for the original 2003 SARS caused by SARS‐CoV‐1 and SARS2 evidence of vascular etiologies, encephalitis (both global and
for the 2019 COVID caused by SARS‐CoV‐2, while SARS defines brainstem‐specific) and/or meningitis, demyelination, and other en-
both SARS1 and SARS2, and SARS‐CoV represents SARS‐CoV‐1 and cephalopathies.21,22 Post‐mortem analysis using transmission electron
SARS‐CoV‐2. 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 lobe.23 However, the question remains
2 | CL I NI CA L EVI D E N C E OF SAR S ‐ COV ‐ 2 whether the outcome neuropathology resulted from direct infection
I N F E C T I O N IN TH E N E R V O U S SY S T E M or a downstream event following a systemic infection; or whether the
lack of evidence of vital direct infection was an outcome of the low
Neurologic and neuropsychiatric dysfunctions in SARS2 patients sensitivity of the detection threshold in methodologies. New tech-
have recently been well discussed.1–6,9 However, many questions nologies such as RNAscope have been utilized to detect SARS‐CoV‐2
remain to be clarified, particularly if these neurologic/psychiatric RNA in cultured cells24 and lung specimens of SARS2 patients, and are
disturbances are caused by direct infection of SARS‐CoV‐2 to the expected to validate the existence of SARS‐CoV‐2 in brain tissues.
brain or the downstream consequences via indirect mechanisms Digital droplet PCR, viral outgrowth assay, and single‐cell RNA se-
caused by SARS‐CoV‐2 systematically. The previous clinical findings quencing of brain samples from SARS2 patients may also help to va-
of the brains from patients infected with SARS‐CoV‐1 have sug- lidate the neurotropism of SARS‐CoV‐2. Further multilabeled
gested that these SARS‐CoVs are able to invade the brain. Sub- immunohistochemical analyses are needed to better understand the
sequent clinical reports have documented both CNS and PNS cellular distribution and mechanisms of SARS‐CoV‐2 in the nervous
involvement including stroke, seizures, smell loss, Guillain‐Barré system.
BODNAR ET AL.
| 1985

In summary, accumulated evidence from clinical studies has animal studies, the virus is delivered intranasally, which may readily
shown a variety of neurological and neuropsychiatric manifestations infect the brain via the olfactory nervous system. The brain cellular
in SARS2 patients (Table 1). However, more convincible evidence for expression pattern of ACE2 in these animal models may address the
direct viral infection to the nervous system remains to be found in susceptibility of brain tissues/cells to SARS‐CoV‐2. SARS‐CoV‐2
addition to the indirect contribution of systemic immune/in- RNAs are detected in the brain of hACE2 mice after intranasal
flammatory responses after SARS‐CoV‐2 infection.46 The availability inoculation.49,54,55
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. 3.2 | Human brain organoid models

To get the most clinically relevant data for whether and how
3 | THE N EU ROPATHOGENIC MODELING SARS‐CoV‐2 may infect the human brain, it is important to use ma-
F OR SA R S 2 terials from humans. The simplest and most direct approach to test
human cellular susceptibility to SARS‐CoV‐2 is to utilize immortalized
Due to the limited sources of human brain specimen from SARS2 human neural cell lines or primary neural cells. SARS‐CoV‐2 can
patients, various models have been developed to explore the neu- moderately infect and replicate in the human glioblastoma cell line
rovirulent infection of SARS‐CoV‐2 in various types of neural cells U251.73 Primary olfactory sensory neurons of human4 and hamsters74
and recapitulate the neuropathogenesis and neurological manifes- are highly susceptible to SARS‐CoV‐2 infection. However, another
tation as well as their correlations with viral neuroinvasion (Table 2). study did not detect any presence of SARS‐CoV‐2 in primary olfactory
neurons in hamsters but did identify viral infection in a large pro-
portion of the supporting sustentacular cells.75 In cultured human
3.1 | Preclinical animal models neural stem cells (NSC)/neural progenitor cells (NPCs), ACE2,
TMPRSS2, cathepsin L, and furin were readily detected and cells
Angiotensin‐converting enzyme‐2 (ACE2) has been well recognized to were highly susceptible to infection with SARS‐CoV‐2,61 but not
serve as the major receptor for SARS‐CoV‐2 to gain entry into host SARS‐CoV‐1.61 Human primary astrocytes are rarely (0.18%) infected
cells.68 The homology conservation and the expression patterns of ACE2 by SARS‐CoV‐2.58 Other human primary neural cells have not yet
69
in various species have been identified. Therefore, numerous non‐ been examined for their susceptibility to SARS‐CoV‐2 infection.
rodent species have been tested as natural animal models for SARS2, Fortunately, the rapidly evolving technology surrounding stem
including monkey, ferret, hamster, cat, bat, and pig.70 Cynomolgus ma- cell culture has allowed for the generation of human‐induced plur-
caques infected with SARS‐CoV‐2 exhibit virus shedding and replication ipotent stem cells (iPSCs) that can be differentiated into virtually any
in lung tissues and certain pathological changes, but no clinical signs,71 cell lineage. This has led to the generation of pure human neural cells
while another study showed moderate clinical symptoms in rhesus ma- such as NSCs/NPCs, neurons, astrocytes, microglia, oligoden-
caques infected with SARS‐CoV‐2.72 Thus, nonhuman primate models drocytes, endothelial cells, etc. Human iPSC‐derived neurons,
may be useful for further preclinical tests; however, monkey models, in particularly dopaminergic neurons, are highly susceptible to
general, fail to manifest clinical illness and are very expensive and in- S‐pseudotyped SARS‐CoV‐2 infection, but iPSC‐derived microglia
convenient. Mouse models are more practical and feasible for elucidating and macrophages are moderately infected by S‐pseudovirions.59 In
virus pathogenicity and therapeutic targets. However, both SARS‐CoVs contrast, another report showed that iPSC‐derived neurons and as-
exhibit a poor cellular tropism in wild‐type mice due to significant species trocytes were sparsely infected, and microglia had no infection;
difference in ACE2 sequence and function. Thus, various types of human however, interestingly, the choroid plexus epithelial cells underwent
ACE2 (hACE2)‐expressing mouse models have been established for robust infection by live SARS‐CoV‐2 virus.58 The inconsistent reports
SARS‐CoV‐2 infection (Table 2). Interestingly, only hACE2 mouse models in neural cell viral susceptibility may result from variable differ-
among the currently available SARS2 animal models fully recapitulated entiation protocols, neural cell maturity, and virus infection dosage.
SARS2 clinical scenarios, particularly the severity and mortality of the Three‐dimensional tissue cultures known as organoids contain
illness. The successfully established hACE2‐transgenic mouse model with multiple cell types and recapitulate tissue structures and develop-
high susceptibility to both SARS‐CoV‐1 and SARS‐CoV‐2 infection ment. Cerebral organoids (COs) have been used to model several
provided a useful tool in SARS‐CoVs studies,47,52,54,55 particularly neurodevelopmental and neuropathological disorders as they accu-
K18‐hACE2 transgenic mice, in which hACE2 expression is driven by the rately resemble the molecular, cellular, and structural features of the
epithelial cell‐specific promoter human cytokeratin 18 (K18) and is cap- developing brain (Figure 1A). COs can be readily infected with
able of recapitulating many features of SARS‐CoV‐2 infections,47,48,52 neurotropic viruses and develop phenotypes similar to that seen
47,48
even better than the other hACE2‐expressing mouse models. clinically, as has been shown in several models studying the Zika
Although numerous reports on SARS2 animal models are avail- virus (ZIKV).76
able, the understanding of neuroinvasive pathogenesis and resultant As it is very difficult to obtain clinical brain samples from patients
neurological changes caused by the virus remains limited. In SARS2 who died of SARS2, the COs are an attractive live human brain model for
1986 | BODNAR ET AL.

T A B L E 1 Clinical features of
Manifestation type Symptom/feature Percentage Reference
neurological and neuropsychiatric
25–27
symptoms in SARS2 patients (percentages General neurological Any nervous symptoms 36.4%–82.3%
derived from the clinical studies with >100 symptoms Headache 6.5%–70.3% 25–34

patients; the others noted the case Muscle pain (myalgia) 22%–62.5% 25–34

numbers) Dizziness 8%–29.7% 26–28,30,34

28,30,32,34
Fatigue 50%–69.6%
26,27,35–37
Seizures <1%
26,27,30,38
Impaired/disordered 4.3%–9%
consciousness
26
Rhabdomyolysis 3.5%
25–28,32,33,39
Sensory impairments Anosmia/hyposmia 5.1%–70.2%
25–28,32,33,39
Ageusia/dysgeusia 5.6%–54.2%
25,27,32
Vision impairment 1.4%–7.4%
26,27,36,37
Neuropathies Peripheral neuropathy 0.4%–8.9%
36,37,40
Gullian–Barre syndrome and 23 cases
variants
27
Nerve pain 2.3%
25,26
Focal weakness and/or motor 0.6%–2.5%
deficit
26,36,37
Encephalopathies Encephalopathy (undefined) 7.2%–31.8%
25,29,35,37,38
Confusion/disorientation/altered 2.0%–27.9%
mental state
17,41
Acute necrotizing 3 cases
encephalopathy (ANE)
21,36
Leukoencephalopathy 2 cases
30
Hypoxic encephalopathy 9%
22,36
Inflammations Acute demyelinating 10 cases
encephalomyelitis (ADEM)
36
Myelitis (CNS) 3 cases
26,36,37,42
Encephalitis 15 cases
42
Lymphocytic meningitis 6 cases
21,27,35–37,42,43
Acute cerebrovascular Ischemic stroke 1.4%–5.0%
disease Intracerebral hemorrhage 25 cases 21,26,35,37,42,43

38
Neuropsychiatric Depression 32.6%
presentations Anxiety 35.7% 38

25,38
Sleep disorders/insomnia 37.2%–41.9%
38
Anger/stress/irritability 3.9%
37,38
Psychosis 0.8%–4.4%
25
Behavioral disorder 2.0%
(not specified)
38
Frequent recall of traumatic 30.4%
memories
37
Cognitive disorder 0.5%
19,30
Neuropathologic Acute hypoxic‐ischemic damage
features Astrogliosis 22,44,45

19,45
Microgliosis
22
Demyelination
17,19,21,22,42,44,45
Neuronal cell loss/injury
45
Cytotoxic T‐cell infiltration
22
Macrophage infiltration
BODNAR
ET AL.

TABLE 2 Experimental models for SARS2 neuropathogenesis

Model Description Model characteristics References


3 5 20,47–53
In vivo Mouse Models Transgenic Human ACE2 expression driven by the epithelial High susceptibility to SARS‐CoV‐2 infection (2 × 10 –1 × 10 plague
K18‐hACE2 mice cell‐specific K18 promoter 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 mice20,47–51
54
Transgenic Human ACE2 expression driven by the lung ciliated Susceptibility to SARS‐CoV‐2 infection and presentation of interstitial
HFH4‐hACE2 mice epithelial cell‐specific HFH4/FOXJ1 promoter pneumonia; High viral RNA levels were detected in the brain of a
portion of mice and was highly correlated with increased mortality
55
Transgenic CRISPR/Cas9 edited knock‐in mice that express human Human ACE2 replaces mouse Ace2 expression; high susceptibility to
hACE2 mice ACE2 under the mouse genomic Ace2 promoter SARS‐CoV‐2 infection, with particularly high viral RNA levels found
in the brain
47,56,57
Adenovirus delivery of Human ACE2 packaged in adenoviral particles under the Human ACE2 expressing cells show susceptibility to SARS‐CoV‐2
hACE2 CMV promoter and delivered to wild‐type mice for infection, with high viral titers in lung tissues; Mice develop some
exogenous expression of hACE2 pathologies but do not display severe SARS2 features
20,58–62
In vitro human Monolayer neural cell Human iPSC‐derived neural cell cultures patterned into Monolayer cultures of mature neurons58‐60 of various subtypes
iPSC‐derived culture cultures specific cell types (e.g., dopaminergic, cortical, etc.), NSCs/NPCs20,61,62 and glial cell
models cultures58,59 including microglia and astrocytes; Susceptibility to
SARS‐CoV‐2 infection varies by cell type
20,60,61,63–65
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
58
Region‐specific 3D cerebral organoids generated from human iPSCs that Organoids of hippocampal, midbrain, hypothalamic, and forebrain
cerebral organoids are patterned to specific regional lineages subtypes with heterogenous mixture of neural cells; Varied
susceptibility to SARS‐CoV‐2 infection
58,66
Choroid plexus 3D cerebral organoids generated from human iPSCs High susceptibility to SARS‐CoV‐2 infection; SARS‐CoV‐2 infection
organoids primarily composed of choroid plexus epithelial cells leads to transcriptional dysregulation and impaired barrier function
67
Blood vessel organoids 3D organoid structures derived from human iPSCs High susceptibility to SARS‐CoV‐2 infection that can be inhibited in an
composed of vascular networks of endothelial cells ACE2‐dependent manner
|
1987
1988 | BODNAR ET AL.

F I G U R E 1 Human brain organoids to model SARS‐CoV‐2 infection. A, Example timeline of an experiment to generate COs with SARS‐CoV‐2
infection. Human fibroblasts or PBMCs are reprogrammed to generate iPSCs, which are then grown in 3D suspension culture as 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 time point 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. CO, cerebral organoid;
EB, embryoid body; iPSC, induced pluripotent stem cell; MPC, microglia progenitor cell; NSC/NPC, neural stem/progenitor cell; OPC,
oligodendrocyte progenitor cell; PBMC, peripheral blood mononuclear cell; SARS‐CoV‐2, severe acute respiratory syndrome coronavirus‐2

studying the impact of SARS‐CoV‐2 infection on the CNS. Utilizing CO showing viral particles in neurons and viral budding in the endoplasmic
models, several labs have demonstrated that the virus can readily infect reticulum.20,61 An additional study using a SARS‐CoV‐2 pseudovirus in-
20,58,59,61,63,64,66
and replicate within COs derived from human iPSCs. The stead of a living virus also found that neurons of COs readily uptake viral
mojority of these studies showed that SARS‐CoV‐2 preferentially in- particles, with a similar expression pattern.60 Although these findings
fected neurons and that viral particles were primarily localized to the provide little clue about how SARS‐CoV‐2 crosses the BBB and enters
soma, sometimes extending into neurites and axons. This infection of the brain, they demonstrate that iPSC‐derived human brain organoids
COs was further confirmed in two studies using electron microscopy, are susceptible to the SARS‐CoV‐2 infection.
BODNAR ET AL.
| 1989

4 | N E U R A L C E L L S U S C EP T I B I L I T Y T O K18‐hACE2 mice after SARS‐CoV‐2 infection49 by intranasal delivery,


SARS ‐C O V‐ 2 suggesting the possibility that SARS‐CoV‐2 live virus can infect brain
neurons via the intranasal route.
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 cells.68 However, 4.2 | Microglia/macrophages
several other receptors have been identified that serve as potential re-
ceptors or co‐receptors for SARS‐CoV‐2 infection such as neuropilin‐1,77 Microglia in the brain play a critical role in neurodevelopment, neural
CD147,78,79 CD26,80–82 CD133,82 CD209,83 furin,61,79 and cadherin‐ innate immunity, and neuroinflammation. Microglia, like macrophages,
17.82 Therefore, studying the expression of ACE2 and other potential may predominantly contribute to the inflammatory response during
receptors throughout the body helps bring understanding to cell sus- Neuro‐SARS2,93 as significant activation of microglia and neuronophagia
ceptibility to SARS‐CoV‐2 infection. Earlier studies found ACE2 mRNA exists in the brainstem (pons, medulla oblongata), and olfactory bulb of
expression nearly ubiquitous but its protein expression more restricted, SARS2 patients.85 Whether SARS‐CoV‐2 can infect and replicate in mi-
with high levels found in the heart, kidneys, lung epithelium, small in- croglia remain elusive. In iPSC‐derived microglia, one study showed no
testine, and vascular endothelium.84 In the brain, neural cells include infection of live SARS‐CoV‐2 virus, but another study demonstrated
neurons, astrocytes, oligodendrocytes, microglia/macrophages, moderate susceptibility to S‐pseudovirions.59 SARS‐CoV‐2 has been
NSCs/NPCs, ependymal cells, pericytes, endothelial cells, and various shown to infect monocytes and macrophages, resulting in host im-
types of immune cells (Table 3). These neural cells also exhibit an en- munoparalysis.96 In human macrophages from lymph nodes and lung,
ormous diversity in their subtypes, sizes, shapes, regional locations, and SARS‐CoV‐2 was detected by immunohistochemistry and electron mi-
functions (Figure 1B). The expression pattern of ACE2 and other po- croscopy.97 Whether infected macrophages infiltrate the brain, serving as
tential receptors in various neural cells remains to be determined and a hijacking mechanism for SARS‐CoV‐2, remains to be determined.
their susceptibility to SARS‐CoV‐2 may vary with physiological and pa-
thological conditions. Exploration of SARS‐CoV‐2 neurotropism and cel-
lular localization is important in elucidating SARS2 neuropathogenesis 4.3 | Pericytes and endothelium cells (EC)
and developing therapeutic and prophylactic countermeasures.
The blood–brain barrier (BBB) functions to protect the brain against
harmful invasion such as viruses. The BBB permeability is exquisitely
4.1 | Neurons regulated by the neurovascular unit formed by highly specialized
endothelial cells, pericytes, astrocytes, and others (Figure 2C). Early
Both in vitro and in vivo studies have demonstrated the neuronal sus- studies with immunohistochemistry showed that ACE2 expression
ceptibility to SARS‐CoV‐2, although the infection efficiency varied with was limited to vascular endothelial cells in the human brain.84 In-
neuronal subtypes, regional difference, and entry receptor expression terestingly, a recent study demonstrated that ACE2 in the brain is
patterns. Database mining showed weak expression of hACE2 in human not expressed in the vascular endothelial cells but instead in
brain cortical neurons but strong expression of CD147 and neuropilin‐1. pericytes.89 Several single‐cell RNA‐seq studies have identified a
Experiments using RT‐PCR, immunostaining, and immunoblotting high level of ACE2 in pericytes.98 Pericytes wrap themselves around
validated hACE2 expression in human neurons to various extents. The the vascular endothelium and play a critical role in maintaining
expression of entry receptors destinates the neuronal infectivity of vascular integrity. However, it is unlikely that the virus would be able
SARS‐CoV‐2. The human glioblastoma cell line U25173 and primary ol- to cross the BBB by passing from the blood to pericytes unless
4
factory sensory neurons are susceptible to SARS‐CoV‐2 infection. In the vascular endothelium itself was damaged in the first place
hamsters, one report identified high infection of primary olfactory neu- (Figure 2C), whether it be from pre‐existing conditions, direct viral
rons, but another study found viral infection only in sustentacular cells infection, or post‐infectious immune response. There has been direct
instead of olfactory neurons (Figure 2A).75 In mice, pervasive expression evidence for SARS‐CoV‐2 infection of vascular endothelial cells and
of Ace2 protein in olfactory epithelial sustentacular cells and olfactory endothelialitis reported in clinical cases.99 This evidence is further
bulb pericytes but not olfactory sensory neurons was detected by im- corroborated by a recent study in which SARS‐CoV‐2 was able to
munohistochemistry, suggesting that olfactory bulb neurons are likely readily infect engineered human blood vessel organoids, which could
not a primary site of infection, but that vascular pericytes may be sen- be reduced using clinical‐grade soluble ACE2.67 iPSC‐derived
94
sitive to SARS‐CoV‐2. Interestingly, SARS‐CoV‐2 infected COs at Day endothelial cells are also susceptible to SARS‐CoV‐2 infection.59
60 at a significantly higher rate than at Day 15, suggesting that the virus The severity of vasculopathy in the brain correlates with the sever
prefers infecting more mature neural cells64 (Figure 1). Neurons in the ity of SARS2 neurological symptoms such as stroke or mental
hypothalamus and with dopaminergic properties are highly susceptible to alterations.100 A clinical case report identified profound pericyte
SARS‐CoV‐2, indicating its potential affinity to some neuroanatomic apoptosis in the lungs of SARS2 patients, which might be the initial
structures underlying pathogenesis for clinical Neuro‐SARS symptoms.95 trigger of micro‐vasculopathy.101 Direct evidence for SARS‐CoV‐2
The S protein‐immunoreactivity is detected in brain neurons of infection in brain pericytes and the sequela is still lacking.
1990
|

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 Varying reports of susceptibility Increased cell death20,58,63,64; impaired Evidence of direct SARS‐CoV2 infection in neurons of K18‐
(subtype and region specific) synaptogenesis20,61,63; Tau mislocalization64 hACE2 and hACE2 transgenic mice20,49,50,55 and select
clinical SARS2 cases20,23; neuronal
injury,17,19,21,22,42,44,85–87 neuronal necrosis and/or
neuronophagia19,22,45,86,88 detected in clinical SARS2
cases

Microglia Inconsistent reports Inconsistent reports Not available due to low susceptibility to SARS‐ Reactive microglia/microgliosis detected in SARS‐CoV‐2
CoV‐2 infection infected K18‐hACE2 transgenic mice49 and clinical
SARS2 cases,19,45,85–88 but no evidence of direct
infection of microglia; presence of microglial nodules in
select SARS2 cases19,45,86,88

Pericytes/ High expression of ACE284,89,90 High susceptibility67 High levels of viral replication and shedding67 Vasculitis and other cerebrovascular etiologies detected in
Endothelial and TMPRSS2 K18‐hACE2 transgenic mice20,49,50; acute
Cells cerebrovascular injuries in numerous clinical SARS2
cases (see Table 1)

Choroid Plexus High expression of ACE2 and High susceptibility58,66 Increased cell death, downregulation of genes One clinical case report of direct SARS‐CoV2 infection in
Cells TMPRSS258,66,90–92 related to BCSFB integrity and upregulation of endothelial cells of the choroid plexus and tanycytes of
inflammatory genes58; Impaired barrier the median emminence92
function66

NSCs/NPCs Moderate expression of ACE2; Conflicting reports of low to Increased cell death20,61,63 No evidence available
expression of TMPRSS2, furin, moderate
and cathepsin L61 susceptibility20,61,63,66

Astrocytes Weak to little ACE2 expression91 Low susceptibility49,58,66,93 Not available due to low susceptibility to SARS‐ Reactive astrocytes/astrocytosis detected in SARS‐CoV‐2
CoV‐2 infection infected K18‐hACE2 transgenic mice49,50 and clinical
SARS2 cases17,22,44,45 but no evidence of direct
infection of astrocytes
BODNAR
ET AL.
BODNAR ET AL.
| 1991

F I G U R E 2 Diagrams for some potential routes of SARS‐CoV‐2 neuroinvasion. 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 BBB via the choroid plexus or 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 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. BBB, blood–brain barrier; CSF, cerebrospinal fluid; CVO, circumventricular organ; SARS‐CoV‐2,
severe acute respiratory syndrome coronavirus‐2

4.4 | Choroid plexus epithelial cells and The BCSFB acts similarly to the BBB as a selectively permeable
ependymal cells barrier for transport to and from the brain but likewise is also sus-
ceptible to pathogen invasion. One study looking at SARS‐CoV‐2
The choroid plexus is a complex network of epithelial cells sur- infectability in region‐specific COs (including hippocampal, hy-
rounding capillaries in the ventricles of the brain and is the primary pothalamic, midbrain, and cortical) (Figure 1B) found that choroid
region of CSF production. The epithelial cells of the choroid plexus plexus epithelial cells expressed in hippocampal organoids were
also have a unique role as serving as the major barrier between the susceptible to SARS‐CoV‐2 viral infection at a much higher rate than
blood and CSF (termed the blood–CSF‐barrier or BCSFB) (Figure 2B). that of neurons and other neural cell subtypes.58 This led the authors
1992 | BODNAR ET AL.

to generate choroid plexus specific organoids, which displayed very 5 | POTENTIAL NEUROINVASIVE ROUTES
high rates of infection even at low viral titer, strongly suggesting that FO R SARS ‐ COV ‐2 IN F EC T IO N
SARS‐CoV‐2 may be able to invade the brain by infecting choroid
plexus epithelial cells in humans. Another study validated the high Neurological symptoms and neuropsychiatric disorders in SARS2
susceptibility of the choroid plexus to SARS‐CoV‐2 S‐pseudovirus patients have been extensively reported and evaluated in numerous
and live virus in iPSC‐derived brain organoids and demonstrated the review papers.1–6,9 The viral entry receptor expression and infection
leakage of BCSFB due to SARS‐CoV‐2‐induced choroid plexus epi- susceptibility of neural cells strongly support the preliminary con-
thelial damages.66 This possibility is further supported by recent clusion that SARS‐CoV‐2 can effectively infect various types of
evidence finding high expression of ACE2 and TMPRSS2 in the neural cells in the human brain and cause neurological/neu-
choroid plexus of both humans and mice.66,90,91 ropsychiatric pathogenesis and symptoms. However, the routes for
The ependymal cells are unique neuroepithelial cells lining the brain SARS‐CoV‐2 neuroinvasive infection remain to be determined. The
ventricles and are critical for CSF generation and regulation. Their basal possibility of the following potential routes is explored based on
membranes contact astrocyte endfeet via tentacle‐like extensions. currently available literature (Figure 2).
SARS‐CoV‐2 has been detected in the CSF of SARS2
patients.13–17,102,103 If SARS‐CoV‐2 in the CSF can infect ependymal
cells and render neuroinvasion remains to be determined (Figure 2B). 5.1 | Retrograde/anterograde axonal transport
Given ependymal cells have weak or no expression of ACE291 and transneuronal invasion
and TMPRSS2,91 they may express other viral entry receptors for
SARS‐CoV‐2, or may allow neuroinvasion under pathological conditions. Neurotropic viruses may invade neurons via virion trafficking in a
retrograde or/and anterograde manner by interacting with neuronal
cytoskeletal proteins. After neuronal infection, the virions may be
4.5 | NSCs/NPCs released and transneuronally spread to neighboring or presynaptic
neurons. Many RNA viruses have been shown to invade the CNS via
Neurogenesis occurs throughout life in both animals and humans. NSCs this transneuronal pathway including SARS‐CoV‐1, thus SARS‐CoV‐2
are self‐renewing, multipotent, and long‐lived cells that generate the is extrapolated to follow similar transneuronal routes to infect the
main cell types of the nervous system. NSCs represent a small population nervous system.
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 such as ZIKV, Dengue virus, CMV, 5.1.1 | Intranasal epithelium and olfactory nervous
76,104 62
etc. ACE2 is highly expressed in human iPSC‐derived NSCs/NPCs ; system
thus, three studies validated infection of SARS‐CoV‐2 in these cells.58,61
However, another study did not detect ACE2 expression or SARS‐CoV‐2 One of the most plausible proposed routes of neuroinvasion of
infection in NSCs/NPCs.66 NSCs share properties with pericytes, epen- SARS‐CoV‐2 is via the olfactory system. Invasion by this route would
dymal cells, and tanycytes. In circumventricular organs (CVOs), NSCs involve the virus infecting cells of the olfactory epithelium, poten-
may have direct contact with circulating pathogens. The functional states tially establishing a viral reservoir (Figure 2A).4,105 This is because (1)
and cell fate decisions of NSCs/NPCs after SARS‐CoV‐2 infection war- olfactory epithelial cells have high ACE2 and TMPRSS2 expression90;
rant further investigation. (2) several clinical studies identified SARS‐CoV‐2 infection in the
olfactory nerve system,19 including MRI hyperintensity in the olfac-
tory bulb of SARS2 patient106; (3) a large majority of SARS2 patients
4.6 | Astrocytes present loss of smell/taste28,39,107,108; and (4) intranasal delivery of
SARS‐CoV‐2 effectively induces brain infection in animal models.50
As astrocytes are the most abundant cell type in the CNS, their role Once cells in the olfactory epithelium are infected, the virus could
in neural virulence has been well studied for different viruses. The either travel transneuronally via axons of olfactory receptor neurons
endfeet of astrocytes play a key role in the formation and function of or by non‐neuronal cells up to the olfactory bulb.105 However, it is
the BBB. However, SARS‐CoV‐2 infection efficiency is very low or still less clear if SARS‐CoV‐2 can infect neuronal cells within the
absent in human primary astrocytes,58,66,93 which may be due to high olfactory epithelium to invade the brain via the olfactory bulb, as
heterogeneity of astrocyte subtypes as well as weak/rare expression olfactory receptor neurons express relatively low levels of ACE2.109
91
of ACE2 and TMPRSS2. As is the case with HIV infection in as- If the olfactory bulb and/or olfactory nerves are infected by
trocytes, a minority of GFAP‐positive cells infected by SARS‐CoV‐2 SARS‐CoV‐2, it may provide the mechanisms for the widely reported
may represent NSCs. A better understanding of the similarity and loss of smell and/or taste in SARS2 patients.28,39,107,108,110 Alter-
diversity of SARS‐CoV‐2 infection between NSCs and astrocytes may natively, an impaired sense of smell may be the result of an immune
provide new therapeutic routes for Neuro‐SARS2. response in the olfactory epithelium, as these cells also co‐express
BODNAR ET AL.
| 1993

high levels of innate immune response genes and olfactory epithelial infect enteric neurons and travel along the well‐established gut‐brain
biopsies from SARS2 patients show significantly increased levels of axis such as the vagus or splanchnic nerve routing into the CNS
pro‐inflammatory cytokines.111 (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 influenza.118 Additionally, infection in the enteric
5.1.2 | Oral health and gustatory/trigeminal glial cells could induce various degrees of inflammatory responses,
nervous system which may aggravate systemic and neurological symptoms.

Oral health plays an important role in the severity of SARS2 devel-


opment. Oral bacterial infection may increase the risk of SARS‐CoV‐2 5.2 | Hematogenous invasion to the nervous
infection. The majority of SARS2 patients present taste disturbances system
such as ageusia, dysgeusia, xerostomia.110,112 In some cases this may
present as a prodromal symptom or as the sole manifestation of 5.2.1 | SARS‐CoV‐2 hematogenous dissemination
113
SARS2, particularly at the early stage. A large cohort study that to circumventricular organ (CVO) and choroid plexus
enrolled >2 million participants concluded that the loss of smell and under physiological condition
taste is more predictive of SARS‐CoV‐2 infection than all other
symptoms, including fatigue, fever, or cough. Similar to olfactory Another potential route of SARS‐CoV‐2 infection into the brain could
neuronal terminals, the gustatory and trigeminal nerve endings may be via circumventricular organs (CVOs) and/or choroid plexus, where
be highly susceptible to SARS‐CoV‐2 infection, especially at the early the BBB is absent, particularly at the early stage of SARS2 wherein
stage of infection (Figure 2).112 Understanding the mechanism of the BBB and BCSFB remain intact elsewhere (Figure 2B).93 The BBB
these neuroinvasions will provide a better strategy for the early di- under physiological conditions functions to prevent harmful patho-
agnosis, prevention, and treatment of SARS2 patients, particularly in gens from entering the brain. However, CVOs have no BBB struc-
regard to neurological manifestations. ture, allowing the circulating pathogens, along with nutrients and
metabolites, cytokines and hormones to freely travel through and
out from the brain.119 Specific CVOs include the median eminence,
5.1.3 | Skin and the peripheral nervous system subfornical organ, and organum vasculosum lamina terminalis
(around the third ventricles at the mid‐level hypothalamus), as well
The human dorsal root ganglion (DRG) neurons express high levels of as the area postrema in the brain stem. Direct imaging to track the
ACE2 at both mRNA and protein levels.114 Database mining showed SARS‐CoV‐2 reporter virus passing through the CVO into the brain
a broad expression of entry receptors in human DRG at the lumbar in human brain organoids or animal models is warranted.
and thoracic levels. In a subset of nociceptive neurons, ACE2 is co- The theory of the choroid plexus serving as an invasion route for
expressed with MRGPRD (Mas‐related G‐protein coupled receptor neurotropic viruses was recently highlighted in a study showing that
member D), a polymodal nociceptive receptor on the nerve endings ZIKV can infect pericytes and cross the BCSFB via the choroid plexus
at the outermost layers of skin and luminal organs. SARS‐CoV‐2 can in mice.120 Interestingly, ZIKV's presence in the CSF and infection of
115
survive on the skin for 9 hours. Thus, SARS‐CoV‐2 may infect the choroid plexus preceded infection in the rest of the brain,
human nociceptors, causing not only peripheral pain but also neu- which could be attenuated by neutralizing the virus in the CSF. If
roinvasion into the CNS via DRG sensory neurons (Figure 2). How- SARS‐CoV‐2 utilized a similar neuroinvasion mechanism, it could
ever, direct evidence for DRG infection is still lacking. explain the inconsistency in CSF detection of the virus. SARS‐CoV‐2
may enter the brain via the CSF before infection of CNS neurons and
then the CSF viral levels may decrease over time as it is cleared by
5.1.4 | Gut infection and enteric nervous system the immune system; however, this could be when neurological
symptoms begin to manifest as the virus spreads in the brain. Both
In addition to respiratory symptoms, gastrointestinal symptoms are CVOs and the choroid plexus are characterized by fenestrated and
also paramount in SARS2 patients due to the abundant expression of highly permeable capillaries (Figure 2B).93 SARS‐CoV‐2 in the CSF
116
ACE2 and other potential entry receptors in mature enterocytes. may infect ependymal cells and subsequently infect CVOs then
Presence of SARS‐CoV‐2 viral RNA can be identified in the stool of spread to neural cells.
117
SARS2 patients. The intrinsic enteric nervous system is enriched
with neurons and glial cells and exquisitely innervates mucosal epi-
thelial cells. Its location makes the enteric nerve endings be readily 5.2.2 | Transendothelial invasion via damaged BBB
exposed to the invading viruses. Notably, a high level of ACE2 and under pathological condition
TMPRSS2 expression has been identified in a large number of enteric
neurons and enteric glia cells.91 Thus, the enteric nervous system is The BBB is formed by the highly specialized endothelial cells, peri-
assumed to be susceptible to SARS‐CoV‐2 infection. The virus could cytes, and astrocytes, and is regulated by the neurovascular unit.
1994 | BODNAR ET AL.

BBB could be damaged by virus infection of any cell components of SARS‐CoV‐2‐infected mature neurons as opposed to the typical lo-
BBB or the post‐infectious inflammatory responses. The SARS‐CoV‐2 calization within axons of normal neurons.64 Further investigation
virus in the blood may directly infect brain endothelial cells and showed that the phospho‐Tau mislocalization is specific to
transendothelially disseminate virus into brain parenchyma via the pT231Tau, which correlated with a fraction of cells that also ex-
damaged BBB, which could be aggravated by viral infection and in- pressed caspase‐3. Thus, SARS‐CoV‐2 infection results in aberrant
flammatory storm (Figure 2C).121 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 re-
5.2.3 | Hijacking (trajectory) route via infected sponse (IFNγ), but not Type I interferon response (IFNα/β).20,58,63,64
immune cells

Several neurotropic viruses such as HIV and Western Nile virus have 7 | CONCLUSIONS A ND FUTURE
been shown to enter brain through a “Trojan horse” mechanism, in DIRECTIONS
which infected immune cells traffic across the permeabilized BBB
and spread the hidden viruses to neurons and glial cells.122 Accumulated evidence from clinical observations and laboratory
SARS‐CoVs have been shown to infect monocytes/macrophages, studies helps to understand the neuropathogenesis underlying
lymphocytes, and leukocytes.96,123 Thus, SARS‐CoV‐2 is likely able to SARS‐CoV‐2 infection‐causing various neurological and neu-
121
traffic across the BBB and BCSFB to infect neural cells. ropsychiatric conditions in SARS2 patients. It remains to be de-
termined whether SARS‐CoV‐2 directly or indirectly induces
neurological/psychiatric damages. Studies have shown that many of
6 | NEURAL CELL CONSEQUENCE AFTER SARS2 patients with an active neurological symptom had no detect-
SARS ‐C O V‐ 2 IN F E C TIO N able virus in the CSF. In addition, no virus could be recovered from the
brain tissues of patients who died of SARS2. These observations
While elucidating the mechanisms of how SARS‐CoV‐2 may enter the suggest the possibility that indirect effects such as inflammation in-
brain is highly important to our understanding of neurological/neu- duced by SARS‐Cov‐2 infection contribute to the neurological/neu-
ropsychiatric manifestations of SARS2, it is equally critical to understand ropsychiatric manifestations. However, failure to detect the virus may
the cellular consequences that result from the virus once in the brain, be due, at least partly, to the low sensitivity of the assays, stages of
such as neural cell survival/death, proliferation, migration, virus produc- infection, and timing when the samples were examined. Notably,
tion, viral spread, etc. Several studies report increased neuronal cell studies from different groups reported the significant differences in
death in COs after SARS‐CoV‐2 infection as determined by TUNNEL SARS‐CoV‐2 infection of various neural cells, which may be re-
staining and caspase‐3 immunostaining.20,58,63,64 Histopathological ex- sponsible for the conflicting data reported. Hopefully, with the avail-
aminations of human brain autopsy specimens have characterized sig- ability of more advanced technologies such as RNAscope,84,94 digital
19
nificant neuronal damage. NSCs/NPCs also experience significant cell droplet PCR,85,93,95 viral outgrowth assay,96,97 single‐cell RNA se-
death.20,61,63 Although less data is available for glial cells, such as astro- quencing,89,98 and multilabeled immunohistochemical analysis as well
cytes and microglia, if infected, they may produce various extents of as better in vivo and in vitro models such as preclinical animals, human
immune/inflammatory responses or neuroprotective responses. Intrigu- iPSCs, and human brain organoids for various neural cells, we will be
ingly, cells infected with SARS‐CoV‐2 may promote the death of unin- able to determine the mechanisms for SARS‐Cov‐2 infection‐caused
fected neighboring cells.20,58 Robust syncytial formation of infected cells neurological injury, which is crucial in developing strategies for
found in the lung of SARS2 patients124 may also occur to neural cells, treatment of SARS2 patients with the neurological/neuropsychiatric
being a source neuropathogenesis in the brain. manifestations. Although several new studies using brain organoids
Transcriptomics analysis identified large scale transcriptional dys- offer strong and straightforward evidence for the neuroinvasive effect
regulation, particularly in metabolic genes and cytokine storm genes as on different neural cells with particularly high susceptibility of choroid
well as genes related to the hypoxia response in SARS‐CoV‐2‐infected plexus epithelial cells, there are still limitations of current brain
cells or choroid plexus organoids,20,58,63,64 confirming the local hypoxic organoid technology to recapitulate clinical patients, including
environment in neurons surrounding SARS‐CoV‐2 infected cells via immaturity of neural cells, unclear dose of neurovirulent infection in
HIF1α expression. The upregulation of vascular remodeling genes and vivo, and lack of additional cell types such as microglia, pericytes,
the downregulation of genes related to CSF secretory function as well endothelial cells, oligodendrocytes, and immune cells.58 Given that the
as cell junction genes suggest a potentially impaired function of the SARS2 pandemic remains raging and seasonal resurgence is impend-
BCSFB and BBB. ing, more extended efforts and investigation into Neuro‐SARS2
Significantly decreased expression of vGLUT1, an excitatory pre‐ mechanisms remain urgently needed. Long‐term follow‐up for poten-
synapse marker, has been identified in human brain organoids, sug- tial neurological/neuropsychiatric manifestations as well as the de-
gesting impaired synaptogenesis in SARS‐CoV‐2 infected cells.20,61,63 velopment of novel prophylactic and therapeutic treatments for
Another study identified Tau mislocalization in the neuronal soma of Neuro‐SARS2 are equally critical.
BODNAR ET AL.
| 1995

A C K N O W L E D GM E N T 12. Kim J‐E, Heo J‐H, Kim H, et al. Neurological complications during
This study was supported by the National Institute of Health treatment of Middle East respiratory syndrome. J Clin Neurol.
2017;13:227‐233.
1R01DA050505‐01 (WZH and WHH) and 1R01AI145034 (WHH).
13. Andriuta D, Roger P‐A, Thibault W, et al. COVID‐19 encephalo-
The content of this review article is solely the responsibility of the pathy: detection of antibodies against SARS‐CoV‐2 in CSF. J Neurol.
authors and does not necessarily represent the official views of the 2020;267:2810‐2811.
National Institutes of Health. All authors have read the journal's 14. Cebrián J, Gonzalez‐Martinez A, García‐Blanco MJ, et al. Headache
and impaired consciousness level associated with SARS‐CoV‐2 in
authorship agreement and policy on disclosure of potential conflicts
CSF. Neurology. 2020;95:266‐268.
of interest. No English writing service was sought for this manuscript. 15. Fadakar N, Ghaemmaghami S, Masoompour SM, et al. A first case of
acute cerebellitis associated with coronavirus disease (COVID‐19): a
C O NF L IC T O F IN T E R ES T S case report and literature review. Cerebellum. 2020;19:911‐914.
16. Kremer S, Lersy F, de Sèze J, et al. Brain MRI findings in severe
The authors declare that there are no conflict of interests.
COVID‐19: a retrospective observational study. Radiology. 2020;
297:E242‐E251.
AUTHOR CONTRIBUTIONS 17. Virhammar J, Kumlien E, Fällmar D, et al. Acute necrotizing en-
Brittany Bodnar and Wenhui Hu: Conceptualization and design, cephalopathy with SARS‐CoV‐2 RNA confirmed in cerebrospinal
literature search, drafting, editing, and critical revision. Jin Jun Luo fluid. Neurology. 2020;95:445‐449.
18. Bellon M, Schweblin C, Lambeng N, et al. Cerebrospinal fluid fea-
and Wenzhe Ho: reviewing, editing, and critical revision. Kena Patel:
tures in SARS‐CoV‐2 RT‐PCR positive patients. Clin Infect Dis.
Literature search, table preparation, and drafting. All approved the 2020. https://doi.org/10.1093/cid/ciaa1165
final version of the manuscript. 19. Solomon IH, Normandin E, Bhattacharyya S, et al. Neuropatholo-
gical features of COVID‐19. N Engl J Med. 2020;383:989‐992.
20. Song E, Zhang C, Israelow B, et al. Neuroinvasion of SARS‐CoV‐2 in
DATA AV AILA BILITY STATEMENT
human and mouse brain. bioRxiv. 2020. https://doi.org/10.1101/
The data that support the findings of this study are available from 2020.06.25.169946
the corresponding author upon reasonable request. 21. Hernández‐Fernández F, Sandoval Valencia H, Barbella‐Aponte RA,
et al. Cerebrovascular disease in patients with COVID‐19: neuroima-
ging, histological and clinical description. Brain. 2020;143:3089‐3103.
ORCID
22. Reichard RR, Kashani KB, Boire NA, Constantopoulos E, Guo Y,
Brittany Bodnar http://orcid.org/0000-0003-0467-5219 Lucchinetti CF. Neuropathology of COVID‐19: a spectrum of vas-
Wenhui Hu http://orcid.org/0000-0001-8152-6116 cular and acute disseminated encephalomyelitis (ADEM)‐like pa-
thology. Acta Neuropathol. 2020;140:1‐6.
23. Paniz‐Mondolfi A, Bryce C, Grimes Z, et al. Central nervous system
R EF E RE N C E S
involvement by severe acute respiratory syndrome coronavirus‐2
1. Assari S. COVID‐19 Pandemic and neurological disease: a critical
(SARS‐CoV‐2). J Med Virol. 2020;92:699‐702.
review of the existing literature. Hosp Pract Res. 2020;5:81‐86.
24. Carossino M, Ip HS, Richt JA, et al. Detection of SARS‐CoV‐2 by
2. Ellul MA, Benjamin L, Singh B, et al. Neurological associations of
RNAscope((R)) in situ hybridization and immunohistochemistry
COVID‐19. Lancet Neurol. 2020;19:767‐783.
techniques. Arch Virol. 2020;165:2373‐2377.
3. Kumar M, Thakur AK. Neurological manifestations and comorbid-
25. Kacem I, Gharbi A, Harizi C, et al. Characteristics, onset, and
ity associated with COVID‐19: an overview. Neurol Sci. 2020;41:
evolution of neurological symptoms in patients with COVID‐19.
3409‐3418.
Neurol Sci. 2020. https://doi.org/10.1007/s10072-020-04866-9
4. Cooper KW, Brann DH, Farruggia MC, et al. COVID‐19 and the
26. Liotta EM, Batra A, Clark JR, et al. Frequent neurologic manifes-
chemical senses: supporting players take center stage. Neuron.
tations and encephalopathy‐associated morbidity in COVID‐19
2020;107:219‐233.
patients. Ann Clin Transl Neurol. 2020;7:2221‐2230.
5. Iadecola C, Anrather J, Kamel H. Effects of COVID‐19 on the
27. Mao L, Jin H, Wang M, et al. Neurologic manifestations of hospi-
nervous system. Cell. 2020;183:16‐27 e1.
talized patients with coronavirus disease 2019 in Wuhan, China.
6. Oliveira R, Sotero FD, Teodoro T. NeuroCOVID: critical review of
JAMA Neurology. 2020;77:683.
neuropsychiatric manifestations of SARS‐CoV‐2 infection. Ir J Med
28. Spinato G, Fabbris C, Polesel J, et al. Alterations in smell or taste in
Sci. 2020. https://doi.org/10.1007/s11845-020-02367-4
mildly symptomatic outpatients with SARS‐CoV‐2 infection. JAMA.
7. Sinanovic O, Muftic M, Sinanovic S. COVID‐19 pandemia: neu-
2020;323:2089‐2091.
ropsychiatric comorbidity and consequences. Psychiatr Danub.
29. Chen N, Zhou M, Dong X, et al. Epidemiological and clinical char-
2020;32:236‐244.
acteristics of 99 cases of 2019 novel coronavirus pneumonia in
8. Roy D, Ghosh R, Dubey S, Dubey MJ, Benito‐León J, Kanti Ray B.
Wuhan, China: a descriptive study. Lancet. 2020;395:507‐513.
Neurological and neuropsychiatric impacts of COVID‐19 pan-
30. Chen T, Wu D, Chen H, et al. Clinical characteristics of 113 de-
demic. Can J Neurol Sci. 2020:1‐16.
ceased patients with coronavirus disease 2019: retrospective
9. Nalleballe K, Reddy Onteddu S, Sharma R, et al. Spectrum of
study. BMJ. 2020;368:m1091.
neuropsychiatric manifestations in COVID‐19. Brain Behav Immun.
31. Huang C, Wang Y, Li X, et al. Clinical features of patients infected with
2020;88:71‐74.
2019 novel coronavirus in Wuhan, China. Lancet. 2020;395:497‐506.
10. Lau K‐K, Yu W‐C, Chu C‐M, Lau ST, Sheng B, Yuen KY. Possible
32. Klopfenstein T, Kadiane‐Oussou NJ, Toko L, et al. Features of
Central nervous system infection by SARScoronavirus. Emerging
anosmia in COVID‐19. Med Mal Infect. 2020;50:436‐439.
Infect Dis. 2004;10:342‐344.
33. Lechien JR, Chiesa‐Estomba CM, Place S, et al. Clinical and epidemio-
11. Gu J, Gong E, Zhang B, et al. Multiple organ infection and the
logical characteristics of 1420 European patients with mild‐to‐
pathogenesis of SARS. J Exp Med. 2005;202:415‐424.
moderate coronavirus disease 2019. J Intern Med. 2020;288:335‐344.
1996 | BODNAR ET AL.

34. Wang D, Hu B, Hu C, et al. Clinical characteristics of 138 hospi- 55. Sun SH, Chen Q, Gu HJ, et al. A mouse model of SARS‐CoV‐2
talized patients with 2019 novel coronavirus–infected pneumonia infection and pathogenesis. Cell Host Microbe. 2020;28:124‐133.
in Wuhan, China. JAMA. 2020;323:1061. 56. Hassan AO, Case JB, Winkler ES, et al. A SARS‐CoV‐2 infection
35. Benussi A, Pilotto A, Premi E, et al. Clinical characteristics and model in mice demonstrates protection by neutralizing antibodies.
outcomes of inpatients with neurologic disease and COVID‐19 in Cell. 2020;182:744‐753.
Brescia, Lombardy, Italy. Neurology. 2020;95:e910‐e920. 57. Sun J, Zhuang Z, Zheng J, et al. Generation of a broadly useful
36. Paterson RW, Brown RL, Benjamin L, et al. The emerging spectrum model for COVID‐19 pathogenesis, vaccination, and treatment.
of COVID‐19 neurology: clinical, radiological and laboratory find- Cell. 2020;182:734‐43.e5.
ings. Brain. 2020;143:3104‐3120. 58. Jacob F, Pather SR, Huang W‐K, et al. Human pluripotent stem cell‐
37. Varatharaj A, Thomas N, Ellul MA, et al. Neurological and derived neural cells and brain organoids reveal SARS‐CoV‐2 neu-
neuropsychiatric complications of COVID‐19 in 153 patients: a rotropism predominates in choroid plexus epithelium. Cell Stem
UK‐wide surveillance study. Lancet Psychiatry. 2020;7:875‐882. Cell. 2020;27(6):937‐950.
38. Rogers JP, Chesney E, Oliver D, et al. Psychiatric and neu- 59. Yang L, Han Y, Nilsson‐Payant BE, et al. A human pluripotent stem cell‐
ropsychiatric presentations associated with severe coronavirus based platform to study SARS‐CoV‐2 tropism and model virus infection
infections: a systematic review and meta‐analysis with comparison in human cells and organoids. Cell Stem Cell. 2020;27:125‐136.e7.
to the COVID‐19 pandemic. Lancet Psychiatry. 2020;7:611‐627. 60. Yi SA, Nam KH, Yun J, et al. Infection of brain organoids and 2D
39. Giacomelli A, Pezzati L, Conti F, et al. Self‐reported olfactory and cortical neurons with SARS‐CoV‐2 pseudovirus. Viruses. 2020;12:
taste disorders in patients with severe acute respiratory cor- 1004.
onavirus 2 infection: a cross‐sectional study. Clin Infect Dis. 2020; 61. Zhang BZ, Chu H, Han S, et al. SARS‐CoV‐2 infects human neural
71:889‐890. progenitor cells and brain organoids. Cell Res. 2020;30:928‐931.
40. Dalakas MC. Guillain‐Barré syndrome: the first documented 62. Kase Y, Okano H. Expression of ACE2 and a viral virulence‐
COVID‐19–triggered autoimmune neurologic disease. Neurology regulating factor CCN family member 1 in human iPSC‐derived
Neuroimmun Neuroinflam. 2020;7:e781. neural cells: implications for COVID‐19‐related CNS disorders.
41. Poyiadji N, Shahin G, Noujaim D, Stone M, Patel S, Griffith B. Inflamm Regen. 2020;40:32.
COVID‐19‐associated acute hemorrhagic necrotizing encephalo- 63. Mesci P, Macia A, Saleh A, et al. Sofosbuvir protects human brain
pathy: imaging features. Radiology. 2020;296:E119‐E120. organoids against SARS‐CoV‐2. bioRxiv. 2020. https://doi.org/10.
42. Weyhern CH, von Kaufmann I, Neff F, Kremer M. Early evidence of 1101/2020.05.30.125856
pronounced brain involvement in fatal COVID‐19 outcomes. 64. Ramani A, Müller L, Ostermann PN, et al. SARS‐CoV‐2 targets
Lancet. 2020;395:e109e. neurons of 3D human brain organoids. EMBO J. 2020;39:e106230.
43. Li Y, Li M, Wang M, et al. Acute cerebrovascular disease following 65. Bullen CK, Bullen K. Infectability of human brainsphere neurons
COVID‐19: a single center, retrospective, observational study. suggests neurotropism of SARS‐CoV‐2. ALTEX. 2020;37(3):665–671.
Stroke Vasc Neurol. 2020;5:279‐284. 66. Pellegrini L, Albecka A, Mallery DL, et al. SARS‐CoV‐2 infects the
44. Kanberg N, Ashton NJ, Andersson L‐M, et al. Neurochemical evi- brain choroid plexus and disrupts the blood‐CSF barrier in human
dence of astrocytic and neuronal injury commonly found in brain organoids. Cell Stem Cell. 2020;27:951‐961.e5.
COVID‐19. Neurology. 2020;95:e1754‐e1759. 67. Monteil V, Kwon H, Prado P, et al. Inhibition of SARS‐CoV‐2 in-
45. Matschke J, Lütgehetmann M, Hagel C, et al. Neuropathology of fections in engineered human tissues using clinical‐grade soluble
patients with COVID‐19 in Germany: a post‐mortem case series. human ACE2. Cell. 2020;181:905‐913.
Lancet Neurol. 2020;19:919‐929. 68. Hoffmann M, Kleine‐Weber H, Schroeder S, et al. SARS‐CoV‐2 cell
46. Abdelaziz OS, Waffa Z. Neuropathogenic human coronaviruses: entry depends on ACE2 and TMPRSS2 and is blocked by a clini-
a review. Rev Med Virol. 2020;30:e2118. cally proven protease inhibitor. Cell. 2020;181(2):271‐280.
47. Rathnasinghe R, Strohmeier S, Amanat F, et al. Comparison of 69. Alexander MR, Schoeder CT, Brown JA, et al. Predicting suscept-
transgenic and adenovirus hACE2 mouse models for SARS‐CoV‐2 ibility to SARS‐CoV‐2 infection based on structural differences in
infection. Emerg Microbes Infect. 2020;9:2433‐2445. ACE2 across species. FASEB J. 2020;34:15946‐15960.
48. Winkler ES, Bailey AL, Kafai NM, et al. SARS‐CoV‐2 infection of 70. Cleary SJ, Pitchford SC, Amison RT, et al. Animal models of me-
human ACE2‐transgenic mice causes severe lung inflammation and chanisms of SARS‐CoV‐2 infection and COVID‐19 pathology. Br
impaired function. Nature Immunol. 2020;21:1‐9. J Pharmacol. 2020;177:4851‐4865.
49. Golden JW, Cline CR, Zeng X, et al. Human angiotensin‐converting 71. Rockx B, Kuiken T, Herfst S, et al. Comparative pathogenesis of
enzyme 2 transgenic mice infected with SARS‐CoV‐2 develop se- COVID‐19, MERS, and SARS in a nonhuman primate model.
vere and fatal respiratory disease. JCI Insight. 2020;5(19):e142032. Science. 2020;368:1012‐1015.
https://doi.org/10.1172/jci.insight.142032 72. Munster VJ, Feldmann F, Williamson BN, et al. Respiratory disease
50. Yinda CK, Port JR, Bushmaker T, et al. K18‐hACE2 mice develop in rhesus macaques inoculated with SARS‐CoV‐2. Nature. 2020;
respiratory disease resembling severe COVID‐19. bioRxiv. 2020. 585:268‐272.
https://doi.org/10.1101/2020.08.11.246314 73. Chu H, Chan JFW, Yuen TTT, et al. Comparative tropism, replica-
51. Zheng J, Wong LYR, Li K, et al. COVID‐19 treatments and patho- tion kinetics, and cell damage profiling of SARS‐CoV‐2 and
genesis including anosmia in K18‐hACE2 mice. Nature. 2020. SARS‐CoV with implications for clinical manifestations, transmis-
https://doi.org/10.1038/s41586-020-2943-z sibility, and laboratory studies of COVID‐19: an observational
52. Moreau GB, Burgess SL, Sturek JM, Donlan AN, Petri WA, Mann BJ. study. Lancet Microbe. 2020;1(1):e14‐e23.
Evaluation of K18‐hACE2 mice as a model of SARS‐CoV‐2 infection. 74. Zhang AJ, Lee ACY, Chu H, et al. SARS‐CoV‐2 infects and damages
Am J Trop Med Hyg. 2020;103:1215‐1219. the mature and immature olfactory sensory neurons of hamsters.
53. Bao L, Deng W, Huang B, et al. The pathogenicity of SARS‐CoV‐2 in Clin Infect Dis. 2020. https://doi.org/10.1093/cid/ciaa995
hACE2 transgenic mice. Nature. 2020;583:830‐833. 75. Bryche B, St Albin A, Murri S, et al. Massive transient damage of
54. Jiang R‐D, Liu M‐Q, Chen Y, et al. Pathogenesis of SARS‐CoV‐2 in the olfactory epithelium associated with infection of sustentacular
transgenic mice expressing human angiotensin‐converting enzyme cells by SARS‐CoV‐2 in golden Syrian hamsters. Brain Behav Immun.
2. Cell. 2020;182:50‐58. 2020;89:579‐586.
BODNAR ET AL.
| 1997

76. Qian X, Nguyen HN, Song MM, et al. Brain‐region‐specific orga- 95. Chiappelli F. Towards neuro‐COVID‐19. Bioinformation. 2020;16:
noids using mini‐bioreactors for modeling ZIKV exposure. Cell. 288‐292.
2016;165:1238‐1254. 96. Merad M, Martin JC. Pathological inflammation in patients with
77. Cantuti‐Castelvetri L, Ojha R, Pedro LD, et al. Neuropilin‐1 facil- COVID‐19: a key role for monocytes and macrophages. Nat Rev
itates SARS‐CoV‐2 cell entry and infectivity. Science. 2020;370: Immunol. 2020;20:355‐362.
856‐860. 97. Martines RB, Ritter JM, Matkovic E, et al. Pathology and patho-
78. Helal MA, Shouman S, Abdelwaly A, et al. Molecular basis of the genesis of SARS‐CoV‐2 associated with fatal coronavirus disease,
potential interaction of SARS‐CoV‐2 spike protein to CD147 in United States. Emerg Infect Dis. 2020;26:2005‐2015.
COVID‐19 associated‐lymphopenia. J Biomol Struct Dyn. 2020:1‐11. 98. Ziegler CGK, Allon SJ, Nyquist SK, et al. SARS‐CoV‐2 receptor
https://doi.org/10.1080/07391102.2020.1822208 ACE2 is an interferon‐stimulated gene in human airway epithelial
79. Latini A, Agolini E, Novelli A, et al. COVID‐19 and genetic variants cells and is detected in specific cell subsets across tissues. Cell.
of protein involved in the SARS‐CoV‐2 entry into the host cells. 2020;181:1016‐1035.
Genes. 2020;11(9). 99. Varga Z, Flammer AJ, Steiger P, et al. Endothelial cell infection and
80. Strollo R, Pozzilli P. DPP4 inhibition: preventing SARS‐CoV‐2 in- endotheliitis in COVID‐19. Lancet. 2020;395:1417‐1418.
fection and/or progression of COVID‐19? Diabetes Metab Res Rev. 100. Avula A, Nalleballe K, Narula N, et al. COVID‐19 presenting as
2020;36:e3330. stroke. Brain Behav Immun. 2020;87:115‐119.
81. Raha AA, Chakraborty S, Henderson J, et al. Investigation of CD26, a 101. Cardot‐Leccia N, Hubiche T, Dellamonica J, Burel‐Vandenbos F,
potential SARS‐CoV‐2 receptor, as a biomarker of age and pathology. Passeron T. Pericyte alteration sheds light on micro‐vasculopathy
Biosci Rep. 2020;40(12). https://doi.org/10.1042/BSR20203092 in COVID‐19 infection. Intensive Care Med. 2020;46:1777‐1778.
82. Kotani N, Nakano T. Candidate screening of host cell membrane 102. Domingues RB, Mendes‐Correa MC, de Moura Leite FBV, et al. First
proteins involved in SARS‐CoV‐2 entry. bioRxiv. 2020. https://doi. case of SARS‐COV‐2 sequencing in cerebrospinal fluid of a patient
org/10.1101/2020.09.09.289488 with suspected demyelinating disease. J Neurol. 2020;267:2‐4.
83. Amraei R, Napoleon M, Yin W, et al. CD209L/L‐SIGN and CD209/ 103. Huang YH, Jiang D, Huang JT. SARS‐CoV‐2 detected in cere-
DC‐SIGN act as receptors for SARS‐CoV‐2 and are differentially brospinal fluid by PCR in a case of COVID‐19 encephalitis. Brain
expressed in lung and kidney epithelial and endothelial cells. Behav Immun. 2020;87:149.
bioRxiv. 2020. https://doi.org/10.1101/2020.06.22.165803 104. Muffat J, Li Y, Omer A, et al. Human induced pluripotent stem cell‐
84. Hamming I, Timens W, Bulthuis M, Lely A, Navis G, van Goor H. derived glial cells and neural progenitors display divergent re-
Tissue distribution of ACE2 protein, the functional receptor for sponses to Zika and dengue infections. Proc Natl Acad Sci U S A.
SARS coronavirus. A first step in understanding SARS pathogen- 2018;115:7117‐7122.
esis. J Pathol. 2004;203:631‐637. 105. Butowt R, Bilinska K. SARS‐CoV‐2: Olfaction, brain infection, and
85. Deigendesch N, Sironi L, Kutza M, et al. Correlates of critical illness‐ the urgent need for clinical samples allowing earlier virus detec-
related encephalopathy predominate postmortem COVID‐19 tion. ACS Chem Neurosci. 2020;11:1200‐1203.
neuropathology. Acta Neuropathol. 2020;140:583‐586. 106. Politi LS, Salsano E, Grimaldi M. Magnetic resonance imaging al-
86. Al‐Dalahmah O, Thakur KT, Nordvig AS, et al. Neuronophagia and teration of the brain in a patient with coronavirus disease 2019
microglial nodules in a SARS‐CoV‐2 patient with cerebellar he- (COVID‐19) and anosmia. JAMA Neurology. 2020;77:1028‐1029.
morrhage. Acta Neuropathol Commun. 2020;8:147. 107. Kaye R, Chang CWD, Kazahaya K, Brereton J, Denneny JC.
87. Kantonen J, Mahzabin S, Mäyränpää MI, et al. Neuropathologic COVID‐19 anosmia reporting tool: initial findings. Otolaryngol Head
features of four autopsied COVID‐19 patients. Brain Pathol. 2020. Neck Surg. 2020;163:132‐134.
https://doi.org/10.1111/bpa.12889 108. Lechien JR, Chiesa‐Estomba CM, De Siati DR, et al. Olfactory and
88. Jensen MP, Le Quesne J, Officer‐Jones L, et al. Neuropathological gustatory dysfunctions as a clinical presentation of mild‐to‐moderate
findings in two patients with fatal COVID‐19. Neuropathol Appl forms of the coronavirus disease (COVID‐19): a multicenter European
Neurobiol. 2020;nan12662. https://doi.org/10.1111/nan.12662 study. Eur Arch Otrhinolaryngol. 2020;277:2251‐2261.
89. He L, Mäe MA, Sun Y, et al. Pericyte‐specific vascular expression of 109. Bilinska K, Jakubowska P, Von Bartheld CS, Butowt R. Expression
SARS‐CoV‐2 receptor ACE2‐ implications for microvascular in- of the SARS‐CoV‐2 entry proteins, ACE2 and TMPRSS2, in cells of
flammation and hypercoagulopathy in COVID‐19 patients. bioRxiv. the olfactory epithelium: identification of cell types and trends
2020. https://doi.org/10.1101/2020.05.11.088500 with age. ACS Chem Neurosci. 2020;11:1555‐1562.
90. Chen R, Wang K, Yu J, et al. The spatial and cell‐type distribution of 110. Parma V, Ohla K, Veldhuizen MG, et al. More than smell‐COVID‐19
SARS‐CoV‐2 receptor ACE2 in human and mouse brain. bioRxiv. 2020. is associated with severe impairment of smell, taste, and che-
https://doi.org/10.1101/2020.04.07.030650 mesthesis. Chem Senses. 2020;45:609‐622.
91. Deffner F, Scharr M, Klingenstein S, et al. Histological evidence for 111. Torabi A, Mohammadbagheri E, Akbari Dilmaghani N, et al. Proin-
the enteric nervous system and the choroid plexus as alternative flammatory cytokines in the olfactory mucosa result in COVID‐19
routes of neuroinvasion by SARS‐CoV2. Front Neuroanat. 2020;14: induced anosmia. ACS Chem Neurosci. 2020;11:1909‐1913.
596439. 112. Samaranayake LP, Fakhruddin KS, Panduwawala C. Sudden onset,
92. Nampoothiri S, Sauve F, Ternier G, et al. The hypothalamus as a hub acute loss of taste and smell in coronavirus disease 2019 (COVID‐19):
for SARS‐CoV‐2 brain infection and pathogenesis. bioRxiv. 2020. a systematic review. Acta Odontol Scand. 2020;78:467‐473.
https://doi.org/10.1101/2020.06.08.139329 113. Biadsee A, Biadsee A, Kassem F, Dagan O, Masarwa S, Ormianer Z.
93. Murta V, Villarreal A, Ramos AJ. Severe acute respiratory syn- Olfactory and oral manifestations of COVID‐19: sex‐related
drome coronavirus 2 impact on the central nervous system: are symptoms—a potential pathway to early diagnosis. Otolaryngol
astrocytes and microglia main players or merely bystanders? ASN Head Neck Surg. 2020;163:722‐728.
Neuro. 2020;12:1759091420954960. 114. Shiers S, Ray PR, Wangzhou A, et al. ACE2 and SCARF expression
94. Brann DH, Tsukahara T, Weinreb C, et al. Non‐neuronal expression in human dorsal root ganglion nociceptors: implications for
of SARS‐CoV‐2 entry genes in the olfactory system suggests me- SARS‐CoV‐2 virus neurological effects. Pain. 2020;161:2494‐2501.
chanisms underlying COVID‐19‐associated anosmia. Sci Adv. 2020; 115. Hirose R, Ikegaya H, Naito Y, et al. Survival of SARS‐CoV‐2 and
6(31):eabc5801. influenza virus on the human skin: importance of hand hygiene in
1998 | BODNAR ET AL.

COVID‐19. Clin Infect Dis. 2020. ciaa1517. https://doi.org/10. pathogen of the central nervous system. Eur J Neurol. 2020;27:
1093/cid/ciaa1517 2348‐2360.
116. Agarwal A, Chen A, Ravindran N, To C, Thuluvath PJ. Gastro- 122. Kramer‐Hammerle S, Rothenaigner I, Wolff H, Bell JE, Brack‐
intestinal and liver manifestations of COVID‐19. J Clin Exp Hepatol. Werner R. Cells of the central nervous system as targets and re-
2020;10:263‐265. servoirs of the human immunodeficiency virus. Virus Res. 2005;
117. Cheung KS, Hung IFN, Chan PPY, et al. Gastrointestinal manifes- 111:194‐213.
tations of SARS‐CoV‐2 Infection and virus load in fecal samples 123. Codo AC, Davanzo GG, Monteiro LB, et al. Elevated glucose levels
from a Hong Kong cohort: systematic review and meta‐analysis. favor SARS‐CoV‐2 infection and monocyte response through a
Gastroenterology. 2020;159:81‐95. HIF‐1alpha/glycolysis‐dependent axis. Cell Metab. 2020;32:
118. Shahbazi R, Yasavoli‐Sharahi H, Alsadi N, Ismail N, Matar C. Pro- 437‐446.
biotics in treatment of viral respiratory infections and neuroin- 124. Oprinca GC, Muja LA. Postmortem examination of three
flammatory disorders. Molecules. 2020;25:4891. SARS‐CoV‐2‐positive autopsies including histopathologic and im-
119. Ferguson AV Circumventricular organs: integrators of circulating munohistochemical analysis. Int J Legal Med. 2020. https://doi.org/
signals controlling hydration, energy balance, and immune func- 10.1007/s00414-020-02406-w
tion. In: De Luca LA, Jr, Menani JV, Johnson AK, editors. Neuro-
biology of Body Fluid Homeostasis: Transduction and Integration. Boca
Raton, FL: CRC Press/Taylor & Francis; 2014.
120. Kim J, Alejandro B, Hetman M, et al. Zika virus infects pericytes in How to cite this article: Bodnar B, Patel K, Ho W, Luo JJ, Hu
the choroid plexus and enters the central nervous system through W. Cellular mechanisms underlying neurological/
the blood‐cerebrospinal fluid barrier. PLOS Pathog. 2020;16:
neuropsychiatric manifestations of COVID‐19. J Med Virol.
e1008204.
121. Alam SB, Willows S, Kulka M, Sandhu JK. Severe acute re- 2021;93:1983–1998. https://doi.org/10.1002/jmv.26720
spiratory syndrome coronavirus 2 may be an underappreciated

You might also like