You are on page 1of 11

Original Article

American Journal of Rhinology & Allergy


2021, Vol. 35(3) 323–333
A Systematic Review of the ! The Author(s) 2020
Article reuse guidelines:
Neuropathologic Findings of Post-Viral sagepub.com/journals-permissions
DOI: 10.1177/1945892420957853
Olfactory Dysfunction: Implications and journals.sagepub.com/home/ajr

Novel Insight for the COVID-19 Pandemic

Jason C. Lee, MD, PhD1 , Rohit Nallani, MD1 ,


Lauren Cass, MD, MPH1, Vidur Bhalla, MD2,
Alexander G. Chiu, MD1, and Jennifer A. Villwock, MD1

Abstract
Background: Post-viral olfactory dysfunction is a common cause of both short- and long-term smell alteration. The
coronavirus pandemic further highlights the importance of post-viral olfactory dysfunction. Currently, a comprehensive
review of the neural mechanism underpinning post-viral olfactory dysfunction is lacking.
Objectives: To synthesize the existing primary literature related to olfactory dysfunction secondary to viral infection, detail
the underlying pathophysiological mechanisms, highlight relevance for the current COVID-19 pandemic, and identify high
impact areas of future research.
Methods: PubMed and Embase were searched to identify studies reporting primary scientific data on post-viral olfactory
dysfunction. Results were supplemented by manual searches. Studies were categorized into animal and human studies for
final analysis and summary.
Results: A total of 38 animal studies and 7 human studies met inclusion criteria and were analyzed. There was significant
variability in study design, experimental model, and outcome measured. Viral effects on the olfactory system varies signif-
icantly based on viral substrain but generally include damage or alteration in components of the olfactory epithelium and/or
the olfactory bulb.
Conclusions: The mechanism of post-viral olfactory dysfunction is highly complex, virus-dependent, and involves a com-
bination of insults at multiple levels of the olfactory pathway. This will have important implications for future diagnostic and
therapeutic developments for patients infected with COVID-19.

Keywords
anosmia, hyposmia, smell dysfunction, olfactory dysfunction, post-viral olfactory dysfunction, coronavirus, SARS, influenza,
respiratory syncytial virus, herpes simplex virus, neuropathology

Introduction
The pandemic of COVID-19, caused by a novel strain of
coronavirus (SARS-CoV-2), is rapidly spreading global-
ly. In the acute phase, symptoms commonly reported 1
Department of Otolaryngology—Head and Neck Surgery, University of
include fatigue, cough, dyspnea, myalgia, joint pain, Kansas School of Medicine, Kansas City, Kansas
anosmia, dysgeusia, and lack of appetite.1 Mounting evi- 2
Saint Luke’s Hospital of Kansas City, Kansas City, Missouri
dence suggests that isolated chemosensory dysfunction –
Corresponding Author:
namely hyposmia, anosmia, and dysgeusia – is frequent.2
Jason C. Lee, Department of Otolaryngology—Head and Neck Surgery,
Recent reports indicate the incidence of anosmia in University of Kansas School of Medicine, Kansas City, KS 66160, USA.
COVID-19 positive patients is as high as 80–98%, with Email: jlee22@kumc.edu
324 American Journal of Rhinology & Allergy 35(3)

Figure 1. A simplified illustration of the olfactory pathway, from initial odorant binding to signal transduction to higher olfactory cortical
centers.

olfactory dysfunction (OD) being the primary symptom hypothesis for the development and deployment of novel
in nearly 12% of patients.3,4 therapeutics, especially those that may have immediate
Anosmia related to viral upper respiratory tract infec- relevance during the current COVID-19 pandemic.
tion (URI) is not unique to COVID-19. Post-viral olfac-
tory dysfunction (PVOD) has long been recognized as a
Methods
major cause of clinically significant olfactory loss,
accounting for up to 40% of the overall incidence in Literature Search
OD.5,6 Nevertheless, the precise neuropathological
mechanism underpinning PVOD in general is poorly The Preferred Reporting Items for Systematic Reviews
understood. and Meta-Analyses (PRISMA) guidelines were followed
The olfactory system is complex, and its organization for this study. We conducted a broad literature search in
consists of distinct structural and functional cellular PubMed and Embase from inception through January
components (Figure 1). Briefly, odorants must reach 31st, 2020 for studies reporting primary scientific data
the olfactory epithelium (OE) and bind to odorant bind- investigating the mechanism of PVOD. The comprehen-
ing proteins (OBPs). Activation of olfactory receptor sive search terms used, including Mesh Terms, was the
neurons (ORNs) then transmits the resultant signal to following: “olfaction disorders”[MeSH Terms] OR
the olfactory bulb (OB). The summation of this signal is “olfactory dysfunction” OR “olfactory disorder” OR
subsequently transduced to higher olfactory cortices and “anosmia” OR “hyposmia” OR “smell disorder”
central sensory areas in the brain, where olfactory proc- OR “olfactory loss” OR “hyposmia” OR “smell”
essing becomes increasingly more complex. AND “respiratory tract infections”[MeSH Terms]
Additionally, there are multiple supporting and regener- OR “Virus” OR “viral” OR “postviral” OR “post-
ative cells critical to olfactory function that continually viral” OR “post-infectious” OR “postinfectious” OR
replenish and maintain this system. A viral insult to any “URI” OR “URTI” OR “severe acute respiratory syn-
of these interconnected components may potentially drome” OR “SARS” OR “covid” OR “coronavirus” OR
result in OD. “influenza” OR “rhinovirus” OR “respiratory syncytial
Here, we present a systematic review that summarizes virus” OR “human metapneumovirus” OR “herpes sim-
and synthesizes the current scientific literature on PVOD plex virus” OR “parainfluenza virus” OR
from both animal and human studies, with an emphasis “cytomegalovirus” OR “Epstein-barr virus” OR
on pathophysiologic mechanisms. Based on the available “adenovirus”. Additional searches were conducted for
data, we propose a novel “Two-Hit Hypothesis” on the each aforementioned URI virus type with AND
development of PVOD and highlight implications of this “olfactory” OR “olfaction” OR “smell”.
Lee et al. 325

Figure 2. Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) flowchart based on our predefined inclusion
and exclusion criteria.

Two authors independently completed the literature expected. Furthermore, studies that did not examine
search and reviewed titles and abstracts, eliminating inel- either the olfactory epithelium (OE) or OB, or their com-
igible articles. Disagreements in article selection were ponent parts, were excluded as pathophysiologic mech-
discussed in depth, and a third independent reviewer anisms could not be determined.
was involved if a tie-breaking vote was needed. Human studies were limited to those assessing PVOD.
Figure 2 displays initial studies identified and excluded Studies that investigate viral detection or serology with-
studies at different screening points. out further investigation of any mechanistic component
(i.e. anatomical or physiological changes within the
Study Selection olfactory system) were excluded as well. Studies of sino-
nasal inflammatory disorders or OD secondary to head
Only primary, retrievable scientific literature available in
trauma were excluded. Additionally, studies examining
the English language were included. Only literature
prognosis and treatment of PVOD were also excluded
examining viruses that are known to cause acute URI
from the formal systematic review. For both animal and
in humans or are analogous in the experimental species
human studies, references within the selected articles
were included. Many viruses, such as vesicular stomatitis
were also reviewed.
virus, rabies virus, and poliovirus, are also neurotropic
and may target the olfactory system.7 However, studies
of these viruses were beyond the scope of this review Data Extraction
focusing on viral URI-induced olfactory loss and thus A wide variety of methods were utilized in selected stud-
were excluded. ies. For animal studies, data extracted included animal
Overall exclusion criteria for animal studies was as model, type of virus examined, structural olfactory unit
follows. Studies that examined only viral titers as param- examined, morphological changes, functional cellular
eters were excluded. In animal studies, experimental unit examined, involvement of inflammation, activation
models utilizing viral injection directly into the olfactory of microglia, any role of apoptosis or programmed cell
bulb (OB) were excluded as such models contrast with death, and any olfactory testing employed in the study.
the natural mode of URI transmission and induce cyto- For human studies, data extraction assessed study type,
toxicity and inflammation beyond what would be sample sizes, imaging modality, anatomical site or brain
326 American Journal of Rhinology & Allergy 35(3)

region studied, brief summary of results, olfactory test- Human Studies


ing used, and any limitations of the study relevant to
Human studies of PVOD also varied widely in method-
understanding the pathophysiology of PVOD.
ology, outcomes, and approach. At the level of the OE,
viral insult caused variable degeneration and death of
Results the ORNs on histopathology with biopsy.52 Increase in
The systematic search resulted in a total of 45 primary olfactory cleft (OC) width and volume was noted to be a
scientific articles specific to the investigation of PVOD potential risk factor for PVOD.46 Three studies demon-
(Figure 2). A subtotal of 38 animal studies were found strated significant reduction in OB volume in patients
and are summarized. Data from animal studies are listed with PVOD,48,50,51 and that greater duration of PVOD
led to greater reduction in OB volume.48,51 Finally, one
and summarized in Table 1. A subtotal of 7 human stud-
study showed that metabolic activities of higher olfac-
ies were identified and are summarized in Table 2.
tory centers were reduced following PVOD.49
Animal Studies
Discussion
There was high variability with respect to methodology
and outcome measured in experimental models of OD Our systematic review revealed that PVOD is attributable
induced by viral infection. Of the 38 animal studies to disruption at many levels of the olfactory pathway by
included in the review, 29 (76.3%) were conducted the cumulative effects of direct cell damage, inflamma-
using mice or rats, 7 (18.5%) were done using ferrets, tion, and cytokine effects. Most animal studies demon-
one (2.6%) was done using dogs, and one (2.6%) using strated that the numbers of ORNs are reduced in the OE
chicken. Of the different type of viruses used, influenza and OB, which is likely the major cause of olfactory dis-
was the most widely studied, followed by coronavirus, ruption following most types of viral infections.
herpes simplex virus (HSV), parainfluenza virus, cyto- Importantly, our review also highlighted the fact that tar-
megalovirus (CMV), and respiratory syncytial virus gets of virus-mediated cellular disruption in the olfactory
(RSV). system are viral substrain dependent. This has significant
In influenza virus, all studies that examined OE dem- implications and emphasizes the need to study and direct
onstrated lesional changes in the OE,19,21–23,29,32,33 with patient care in a highly virus-specific manner following
exception of a non-neurovirulent subtype H3N2.21 PVOD. Our review also found that human studies con-
Olfactory bulb was examined specifically in 12 studies, sistently suggest that both peripheral and central olfac-
with 9 studies (75%) showing histological evidence of tory structures are negatively impacted in relation to
lesional changes.20,22,23,25–28,31,33 Neural damages in structural integrity, volume, or metabolism. Many impor-
higher cortical centers were also noted in 9 stud- tant studies in humans have shown putative viral agents
ies.19,20,22,23,25–28,33 Damages to various cell types in PVOD. For instance, high levels of parainfluenza virus
within the olfactory system were observed, including 3 have been detected in patients with PVOD.54 However,
ORNs,22,23,27,32,33 supporting cells (SC),19 basal cells direct demonstration of specific pathophysiological
(BC),19 granule cells (GC),22,23,33 and mitral cells changes within the olfactory system following viral sub-
(MC).22–24,33 type insults in humans has yet to be accomplished.
In coronavirus, none of the studies demonstrated
gross lesioning in the OE.38–40 However, the number or Viral Specific Effects on the Olfactory System
turnover of ORNs were negatively impacted.37–40 The Two important aspects of the influenza virus emerged
OB was specifically examined histologically in 4 studies, during our review: (1) histologic changes in the OE
with 3 studies (75%) showing some evidence of lesional and OB are relatively small, consisting of small foci of
changes.35,38,53 Three studies that investigated higher degeneration, and (2) hemagglutinin plays a critical role
cortical centers also noted neuronal changes.35,36,38 in influenza neurotropism, or the ability to infect certain
In parainfluenza virus, 2 of the 5 studies noted gross neural cells. The OE and OB are consistently affected by
changes in the OE,42,45 while 3 others did not.41,43,44 the influenza virus but exhibit only small foci of histo-
Notably, no gross changes in the olfactory bulb or logic abnormalities in all included studies. Involvement
higher cortical centers were demonstrated.42,44,45 The of higher cortical centers within the olfactory pathway
ORNs were damaged or negatively impacted in all avail- appears to be subtype specific.28 Direct comparison of
able studies.41,42,44,45 H5N1 and H1N1 showed that infection with H5N1
In HSV, neuronal lesions were identified throughout resulted in lesion and inflammation of higher cortical
the olfactory system, including the OE, OB, as well as centers with poor animal survival, while H1N1 infection
higher cortical centers in all 6 included studies,13–18 with was strictly limited to the OE.28 Adding to the complex-
the exception of a neuro-attenuated L1BR1 substrain.18 ity, different substrains of H5N1 influenza also exhibited
Lee et al. 327

Table 1. Animal Studies.


Virus Model Examined Region Lesioned Site Affected Cell Microglia Inflammation Cytokine Apoptosis
8 8
RSV Mice OE, OB OE ORNþ/BSþ N/A N/A N/A N/A
Mice9 OE, OB OEþ, OB ORNþ N/A Yes IL-17C N/A
CMV Mice10 OE OEþ ORNþ/SCþ N/A N/A N/A N/A
Mice11 OE N/A ORNþ/SCþ N/A N/A N/A N/A
Mice12 OE N/A ORNþ/SCþ N/A N/A N/A N/A
HSV Mice13 OB, HCR OBþ, HCRþ MCþ/GCþ Yes Yes TNF-a IFN-c Unchanged
Neuronsþ
Mice14 OB, HCR N/A MCþ N/A Yes N/A N/A
Rats15 OB, HCR OBþ, HCRþ N/A N/A Yes N/A N/A
Mice16 OB, HCR OBþ, HCRþ MCþ/GCþ Activated Yes N/A N/A
Mice17 OE, OB, HCR OBþ, HCRþ MCþ/GCþ N/A Yes N/A N/A
Mice18¥ OE, OB 186 - OEþ/OBþ/HCRþ ORNþ N/A N/A N/A 186 -
L1BR1 - unchangedL1BR1 -
OEþ/OB/HCR increased
Influenza Chicken19 OE, OB, HCR OEþ/OB/HCRþ ORN/SCþ/BCþ; Activated N/A N/A N/A
Neuronsþ,
glial cellsþ
Ferrets20 OB, HCR OBþ, HCRþ Neuronsþ Activated Yes N/A N/A
Ferrets21¥ OE H5N1 - OEþ N/A N/A Yes N/A N/A
H3N2 - OE
H1N1 - OEþ
Ferrets22 OE, OB, HCR OEþ/OBþ/HCRþ ORNþ/MCþ/GCþ N/A Yes N/A N/A
Ferrets23¥ OE, OB, HCR H5N1 - OEþ ORNþ/MCþ/GCþ Activated Yes N/A N/A
/OBþ/HCRþ
MBCS -
OEþ/OB/HCR
Mice24 OB, HCR N/A MCþ/Neuronþ Activated N/A N/A Increased
Ferrets25¥ OB, HCR VN1203 - OBþ/HCRþ N/A N/A Yes N/A N/A
HK483 - OBþ/HCRþ
Ferrets26¥ OB, HCR HK486 - OBþ/HCRþ N/A N/A Yes N/A N/A
HK483 - OBþ/HCRþ
Mice27 OB, HCR OBþ/HCRþ ORNþ/Neuronsþ N/A N/A N/A N/A
Mice28¥ OB, HCR HK483 - OBþ/HCRþ HK483 -Neuronsþ N/A Yes N/A N/A
PR8 - Epithelial cells /Glial cells
PR8 -
Epithelial cells
Ferrets29 OE, OB OEþ/OB N/A N/A Yes TNF-a N/A
IL-6
Mice30 OB N/A Neuron-/Glial cellsþ Activated N/A TNF-a IL1-b N/A
Mice31 OB OBþ N/A N/A N/A TNF-a IL1-b N/A
Mice32 OE, OB OEþ, OB ORNþ Activated No N/A Increased
Mice Tg33 OE, OB, HCR OEþ, OBþ, HCRþ ORNþ/SC/MCþ Activated Yes N/A N/A
/TCþ/GCþ
Coronavirus Mice34 OB N/A N/A Activated N/A IFN N/A
Mice35 OB OBþ/HCRþ MCþ N/A Yes N/A N/A
Mice Tg36 OB, HCR OB/HCRþ N/A N/A No N/A N/A
Mice37 OB OBþ ORNþ N/A N/A N/A Increased
Mice38 OE, OB, HCR OE/OBþ/HCRþ ORNþ/MCþ/GCþ/TCþ N/A Yes N/A N/A
Mice39 OE, OB OE/OBþ ORNþ/MCþ/GCþ/TCþ N/A N/A N/A N/A
Mice40 OE OE ORNþ N/A N/A N/A N/A
Parainfluenza Mice41 OE, OB OE ORNþ/BCþ N/A No N/A Decreased
Mice42 OE, OB OEþ/OB ORNþ/MC/TC N/A N/A N/A N/A
Dogs43 OE OE N/A N/A No N/A N/A
Mice44 OE, OB, HCR OE/OB/HCR ORNþ/SCþ N/A No N/A N/A
Mice45 OE, OB OEþ/OB ORNþ/SC/MC/TC N/A N/A N/A N/A

Abbreviations: OE, olfactory epithelium; OB, olfactory bulb; HCR, higher cortical centers; ORN, olfactory receptor neuron; SC, supporting cell; BS, basal
cell; MC, mitrial cell; GC, granule cell; TC, tufted cell; Neurons, neural cells not otherwise specified in the article; þ, denotes lesion or cell infected cells; 
denotes no lesion or uninfected cells; Tg, transgenic; N/A, not applicable; ¥, studies that used multiple viral strains.

variable neurovirulence. H5N1 VN1203 substrain exhib- mutation of a multi-basic cleavage site within the hem-
ited a rapid and widespread CNS transmission that agglutinin impaired the ability of H5N1 to spread within
resulted in more extensive neuronal death and inflamma- the olfactory system and the CNS.23 These together sug-
tory response, while infection with its counterpart H5N1 gest that hemagglutinin has a critical role in neurotrop-
HK483 showed a much slower spread within the CNS ism. In general, microglial activation, inflammatory
with less notable cellular damage.25 Interestingly, response, and cytokine production appear to be elevated
328
Table 2. Human Studies.

Anatomical Site
Sample Imaging or Brain Region
Authors Year Study Type Size Modality Studied Results Olfactory Testing
46
Altundag et al. 2020 Retrospective Case– n ¼ 71 CT Scan OC Significantly greater OC width and volume Sniffin Sticks
Control in OD group compared to controls.
No difference in olfactory fossa depth
Wolf et al.47 2018 Cross-sectional n ¼ 21 N/A OC 1117 different proteins detected with 10 Sniffin Sticks
being most abundant
No differences between OD patients and
healthy controls in olfactory cleft
proteome
Yao et al.48 2018 Cross-sectional n ¼ 38 Voxel-based OB/OFC Significant decrease in right OFC and total T&T Olfactometry &
MRI OB volume in PVOD. Significant negative Sniffin Sticks
correlation between right OFC and OB
volume and duration of olfactory loss
Kim et al.49 2012 Cross-sectional n ¼ 18 FDG-PET OFC Significant hypometabolism in olfactory Olfactory Butanol
sensory and integration brain regions in Threshold Test &
PVOD patients compared to controls Olfactory
Regional metabolism in cortical regions Identification Test
inversely correlated with olfactory tests
Rombaux et al.50 2009 Review þ Cross-sectional n ¼ 122 MRI OB Reduced OB volume in all patients Sniffin Sticks
Cohort OB volume significantly correlated with Retronasal Testing
TDI and retronasal scores.
Rombaux et al.51 2006 Retrospective Cohort n ¼ 26 MRI OB/Olfactory OB volume smaller in anosmic patients Sniffin Sticks
Sulcus compared to hyposmia. No difference in
olfactory sulcus depth in hyposmia vs.
anosmia patients. Significant negative
correlation between OB volume and
duration of olfactory loss
Yamagishi et al.52 1994 Cross-sectional n ¼ 70 N/A Olfactory Variable damage to and degeneration of T&T Olfactometry &
Epithelium peripheral ORNs identified on mucosal IV Alinamin Injection
samples Test

Abbreviations: PVOD, post-viral olfactory dysfunction; OC, olfactory cleft; OD, olfactory dysfunction; OFC, orbitofrontal cortex; OB, olfactory bulb; T&T, odor-threshold & identification; TDI, Threshold,
Discrimination, Identification; ORN, Olfactory Receptor Neuron; NSE, neuron-specific enolase.
Lee et al. 329

following neuroinvasion of influenza, which collectively within the olfactory system is important in the patho-
may contribute to viral clearance and cell death. A nota- genesis of OD. According to single cell sequencing anal-
ble exception is influenza A R404BP which induces min- ysis of RNA profiles from existing databases, only a
imal inflammation in the olfactory system but significant subset of sustentacular cells, horizontal basal cells, and
apoptosis of the ORNs.32 Bowman’s gland cells co-express the genes encoding the
Coronaviruses similarly displayed strain specific dif- ACE2 receptor and TMPRSS2 protease, both of which
ferences in neurovirulence. For example, SARS-CoV-1 are essential for SARS-CoV-2 viral entry.55–57
in transgenic mice expressing human angiotensin- Therefore, SARS-CoV-2 may disrupt homeostasis and
converting enzyme 2 (ACE2) in epithelial cell lines dem- regeneration of ORNs in the OE, leading to clinically
onstrated a high degree of neurovirulence towards observable OD. Alternatively, SARS-CoV-2 may pro-
higher cortical regions in the olfactory pathway while duce extensive neuronal damage in higher cortical cen-
sparing much of the morphology of the OB.36 This con- ters of the olfactory pathway, like its SARS-CoV-1
trasts with the highly neurovirulent strain of MHV-JHM counterpart.36
and MHV-OBLV—both of which are murine coronavi-
rus counterpart—that significantly disrupt the OB and Filling the Knowledge Gap in PVOD
higher cortical regions with resultant cell death.35,38
Available animal and human studies demonstrate that
Consistent with other studies, disruption of the architec-
measurable and observable damages to the olfactory
ture within the OB was shown to lead to olfactory
system following viral infection are apparent: reduction
impairment after coronavirus infection in vivo.53
in progenitor cells, cell death of ORNs, damage to sup-
Parainfluenza virus had unique tropism—or the abil-
porting cells, deciliation of neuroepithelium, and lesions
ity of specific virus to target and infect different cells—
of higher olfactory cortical centers. However, most stud-
within the olfactory system. All studies to date have
ies also showed some degree of regeneration and repair
shown that, regardless of subtypes, the neurovirulence
following viral clearance. Notably, the olfactory system
of parainfluenza is restricted to primary ORNs and their
has a remarkable ability to regenerate. For instance,
surrounding cells in the OE and OB. Infectivity or lesion
bilaterally bulbectomized rats have been shown to
of higher cortical lesions has yet to be demonstrated. The
retain the ability to smell via newly generated connec-
component that restricts parainfluenza tropism within
tions to the forebrain.58 Furthermore, induced chemical
the olfactory system remains elusive. It is also unclear
lesion of the OE—by as much as 95%—and the OB in
if viral components are transported to higher cortical
rats showed little to no impairment of olfactory function
regions. Two studies objectively evaluated olfaction
and failed to produce anosmia.53,59,60 Additionally,
after parainfluenza infection and found deficits.41,43
anosmia is reported in 65% of patients who survive
Following infection of Sendai virus (murine parain-
herpes encephalitis, which inflicts devastating neuronal
fluenza), mice showed increased latency to find buried
damage in multiple brain areas, including the olfactory
food pallet even in a food deprived state.41 ORNs also
pathway.61 This highlights the remarkable permissibility
showed diminished calcium signaling to odorant bind-
of the olfactory pathway to neuronal damage and its
ing, suggesting impairment in depolarization and signal
regenerative ability. Finally, normal and subnormal
transduction on a cellular level.41 Interestingly, parain-
olfaction in humans has been reported in the absence
fluenza virus also produces minimal morphologic
of the OB.62,63 Therefore, the mechanism of how viral
changes or inflammatory response when compared to
insults account for the lack of olfactory perception on a
other viruses, such as influenza or herpes. This may
long-term or even permanent basis remains elusive.
account for the prolonged presence of viral protein
For a previously normosmic individual to become
and viral RNA within the OB.41,45
truly anosmic, the damage would theoretically need to
Studies demonstrated high neurovirulence of HSV for
be quite extensive and long lasting, which does not
the CNS. In vivo studies of HSV-1 and HSV-2 consis-
appear to be the case based on the available animal lit-
tently showed high degree of neuroinvasion within the
erature. Yet, PVOD is consistently a major cause of
olfactory pathway as well as in higher cortical centers,
long-term OD despite differences in the mechanism of
resulting in encephalitis and cell death.13,15,16 ORNs,
initial insults. Furthermore, human studies that broadly
mitral cells, and granule cells have all been shown to
studied PVOD consistently show volumetric changes in
undergo cell death following HSV infection.
olfactory related brain regions.50,51 Barring complete
disruption of regenerative ability and destruction of
Implications for COVID-19 the olfactory pathway, this evidence taken together sug-
Fortunately, investigation into the pathophysiological gests a convergent mechanism for clinical manifestation
mechanism of SARS-CoV-2-induced OD is already of long-term OD beyond the apparent anatomical
underway.55–57 As indicated earlier, viral tropism changes. We therefore postulate that the fundamental
330 American Journal of Rhinology & Allergy 35(3)

the preponderance of female patients affected by PVOD,


especially in the elderly.50 However, it should be noted
that this novel theory is speculative and further investi-
gation in warranted.

Novel Therapeutic Strategy


Based on the natural clinical history of PVOD (Figure
3), treatment strategy can be categorized into those that
specifically target the acute phase, and those that target
the chronic phase. Arguably, the best form of interven-
tion is to prevent the initial viral entry, which may be
Figure 3. Natural progression of PVOD. Each phase consists of
distinctive sets of viral insults. Persistence of OD into a long-term, relevant for COVID-19. SARS-CoV utilizes host ACE2
or permanent, basis requires impairment of synaptic plasticity in receptors for cell entry.57 Prevention of SARS-CoV-2
addition to cellular damage. viral entry has been demonstrated using specific protease
inhibitors in vivo.56 Given the current COVID-19 pan-
demic, a push towards viral-entry specific research and
process of olfactory coding is altered following the initial
intervention may be important in the absence of effective
viral insult. This is further supported by fMRI studies in
vaccination. For substrains of viruses that induce inflam-
humans that have demonstrated aberrant functional
mation, topical or systemic corticosteroids may prove
olfactory connectivity in PVOD patients compared to
useful in the acute phase of PVOD. Viral mediated
that of control subjects.64 The question naturally
induction of cytokines may persist for many months.74
arises, then, as to how viral infection alters olfactory
Elevated cytokines such as IFN-c and TNF-a have been
coding. Here, we suggest that synaptic plasticity, or the
well demonstrated to cause OD in vivo.75,76 Therefore,
ability to strengthen or weaken communications
selectively targeting these cytokines may provide yet
between neurons, is a potential target of viral disruption.
another avenue for therapy. Cytokine targeted treatment
may however be complicated by their critical function in
Two-Hit Hypothesis
viral clearance in the early phase of the infection.74 Stem
We propose a “Two-Hit Hypothesis” which posits that cell or gene therapy targeting basal proliferating cells in
secondary impairment of synaptic plasticity in combina- the OE or neuroblast precursors in the subventricular
tion with initial viral insults is necessary for the clinical zone to specifically improve regenerative potential of
development of persistent PVOD (Figure 3). Consistent the OE or OB, respectively, may also prove to be
with our hypothesis, influenza A has been shown to useful in the future.
induce long lasting changes in synaptic plasticity in the However, patients presenting to otolaryngologists are
hippocampus even after viral clearance, presumably more likely to fall in the chronic phase category; anos-
through the action of cytokines, activated microglia, as mia alone may not lead one to seek treatment immedi-
well as altered gene expression.65 Similarly, RSV infec- ately. According to our novel “Two-Hit Hypothesis”,
tion has been shown to impair synaptic plasticity even therapies specifically targeting synaptic plasticity
after viral clearance, leading to cognitive impairment.66 should be explored. Nevertheless, determining the pre-
The ability of neurotropic viruses to hijack synaptic plas- cise mechanism by which different viruses detrimentally
ticity has also been demonstrated in Borna disease virus, affect synaptic plasticity within the olfactory pathway
which disrupts synaptic vesicle recycling.67 It is conceiv- may open a new frontier in therapeutic options.
able that most, if not all, neurotropic viruses have the
ability to alter synaptic plasticity through unknown
mechanisms.
Conclusion
The hallmark of synaptic plasticity is activity-induced The underlying pathophysiological mechanism of PVOD
or learning-induced modification,68 which may, in part, is complex and virus dependent. A combination of direct
explain why olfactory training is an effective therapy for damage to ORNs, impairment in olfactory neuron
PVOD.69,70 Interestingly, older individuals appear to be regeneration, inflammatory response, cytokine action,
more susceptible to develop PVOD.71 Age-dependent as well as higher cortical damage may account for the
changes in synaptic plasticity are also well observed clinical manifestation of PVOD. Permanent or long-term
and may, in part, account for age-related susceptibility OD beyond the period of viral clearance and cellular
to PVOD.72 Similarly, synaptic plasticity is closely reg- regeneration likely involves additional alteration in the
ulated by hormonal changes,73 which may partly explain olfactory system.
Lee et al. 331

Acknowledgment natural primary CMV infections. Virus Res. 2016;


We would like thank Teresa Lee for kindly providing the illus- 211:96–102.
trations included in this article. 12. Zhang S, Xiang J, Van Doorsselaere J, et al. Comparison
of the pathogenesis of the highly passaged MCMV smith
strain with that of the low passaged MCMV HaNa1 isolate
Declaration of Conflicting Interests in BALB/c mice upon oronasal inoculation. Vet Res.
The author(s) declared no potential conflicts of interest with 2015;46:94–99.
respect to the research, authorship, and/or publication of this 13. Armien AG, Hu S, Little MR, et al. Chronic cortical and
article. subcortical pathology with associated neurological deficits
ensuing experimental herpes encephalitis. Brain Pathol.
2010;20(4):738–750.
Funding 14. Jennische E, Eriksson CE, Lange S, et al. The anterior
The author(s) received no financial support for the research, commissure is a pathway for contralateral spread of
authorship, and/or publication of this article. herpes simplex virus type 1 after olfactory tract infection.
J Neurovirol. 2015;21(2):129–147.
15. Beers DR, Henkel JS, Schaefer DC, et al. Neuropathology
ORCID iDs of herpes simplex virus encephalitis in a rat seizure model.
Jason C. Lee https://orcid.org/0000-0001-9117-4492 J Neuropathol Exp Neurol. 1993;52(3):241–252.
Rohit Nallani https://orcid.org/0000-0001-6313-205X 16. Tomlinson AH, Esiri MM. Herpes simplex encephalitis.
Immunohistological demonstration of spread of virus
References via olfactory pathways in mice. J Neurol Sci.
1983;60(3):473–484.
1. Carfi A, Bernabei R, Landi F, for the Gemelli Against
17. Anderson JR, Field HJ. The distribution of herpes simplex
COVID-19 Post-Acute Care Study Group. Persistent
type 1 antigen in mouse Central nervous system after dif-
symptoms in patients after acute COVID-19. JAMA.
ferent routes of inoculation. J Neurol Sci.
2020;324(6):603.
1983;60(2):181–195.
2. Vaira LA, Salzano G, Deiana G, Riu GD. Anosmia and
18. Mori I, Goshima F, Watanabe D, et al. Herpes simplex
ageusia: common findings in COVID-19 patients.
virus US3 protein kinase regulates virus-induced apoptosis
Laryngoscope. 2020;130(7):1787.
in olfactory and vomeronasal chemosensory neurons in
3. Lechien JR, Chiesa-Estomba CM, De Siati DR, et al.
vivo. Microbes Infect. 2006;8(7):1806–1812.
Olfactory and gustatory dysfunctions as a clinical presen-
19. Chaves AJ, Busquets N, Valle R, et al. Neuropathogenesis
tation of mild-to-moderate forms of the coronavirus dis- of a highly pathogenic avian influenza virus (H7N1) in
ease (COVID-19): a multicenter European study. Eur Arch experimentally infected chickens. Vet Res. 2011;
Otorhinolaryngol. 2020;277(8):2251–2261. 42:106–111.
4. Moein ST, Hashemian SMR, Mansourafshar B, et al. 20. Bodewes R, Kreijtz JH, van Amerongen G, et al.
Smell dysfunction: a biomarker for COVID-19. Int Pathogenesis of influenza a/H5N1 virus infection in ferrets
Forum Allergy Rhinol. 2020;10(8):944–950. differs between intranasal and intratracheal routes of inoc-
5. Henkin RI, Levy LM, Fordyce A. Taste and smell function ulation. Am J Pathol. 2011;179(1):30–36.
in chronic disease: a review of clinical and biochemical 21. van den Brand JM, Stittelaar KJ, van Amerongen G, et al.
evaluations of taste and smell dysfunction in over 5000 Comparison of temporal and spatial dynamics of seasonal
patients at the taste and smell clinic in Washington, DC. H3N2, pandemic H1N1 and highly pathogenic avian influ-
Am J Otolaryngol. 2013;34(5):477–489. enza H5N1 virus infections in ferrets. PLoS One.
6. Seiden AM. Postviral olfactory loss. Otolaryngol Clin 2012;7(8):e42343.
North Am. 2004;37(6):1159–1166. 22. Yamada M, Bingham J, Payne J, et al. Multiple routes of
7. van Riel D, Verdijk R, Kuiken T. The olfactory nerve: a invasion of wild-type clade 1 highly pathogenic avian influ-
shortcut for influenza and other viral diseases into the cen- enza H5N1 virus into the central nervous system (CNS)
tral nervous system. J Pathol. 2015;235(2):277–287. after intranasal exposure in ferrets. Acta Neuropathol.
8. Ueha R, Mukherjee S, Ueha S, et al. Viral disruption of 2012;124(4):505–516.
olfactory progenitors is exacerbated in allergic mice. Int 23. Schrauwen EJ, Herfst S, Leijten LM, et al. The multibasic
Immunopharmacol. 2014;22(1):242–247. cleavage site in H5N1 virus is critical for systemic spread
9. Bryche B, Fretaud M, Saint-Albin Deliot A, et al. along the olfactory and hematogenous routes in ferrets.
Respiratory syncytial virus tropism for olfactory sensory J Virol. 2012;86(7):3975–3984.
neurons in mice. J Neurochem. 2019;e14936. 24. Jang H, Boltz D, Sturm-Ramirez K, et al. Highly patho-
10. Farrell HE, Lawler C, Tan CS, et al. Murine cytomegalo- genic H5N1 influenza virus can enter the Central
virus exploits olfaction to enter new hosts. mBio. 2016;7(2): nervous system and induce neuroinflammation and neuro-
e00251–e00216. degeneration. Proc Natl Acad Sci USA.
11. Xiang J, Zhang S, Nauwynck H. Infections of neonatal 2009;106(33):14063–14068.
and adult mice with murine CMV HaNa1 strain upon 25. Plourde JR, Pyles JA, Layton RC, et al. Neurovirulence of
oronasal inoculation: new insights in the pathogenesis of H5N1 infection in ferrets is mediated by multifocal
332 American Journal of Rhinology & Allergy 35(3)

replication in distinct permissive neuronal cell regions. (OBLV) of mouse hepatitis strain JHM. Chem Senses.
PLoS One. 2012;7(10):e46605. 2001;26(8):953–963.
26. Shinya K, Makino A, Hatta M, et al. Subclinical brain 40. Bihun CG, Percy DH. Morphologic changes in the nasal
injury caused by H5N1 influenza virus infection. J Virol. cavity associated with sialodacryoadenitis virus infection in
2011;85(10):5202–5207. the wistar rat. Vet Pathol. 1995;32(1):1–10.
27. Park CH, Ishinaka M, Takada A, et al. The 41. Tian J, Pinto JM, Cui X, et al. Sendai virus induces per-
invasion routes of neurovirulent a/Hong Kong/483/97 sistent olfactory dysfunction in a murine model of PVOD
(H5N1) influenza virus into the Central nervous system via effects on apoptosis, cell proliferation, and response to
after respiratory infection in mice. Arch Virol. odorants. PLoS One. 2016;11(7):e0159033.
2002;147(7):1425–1436. 42. Mori I, Nakakuki K, Kimura Y. Temperature-sensitive
28. Iwasaki T, Itamura S, Nishimura H, et al. Productive parainfluenza type 1 vaccine virus directly accesses the
infection in the murine central nervous system with avian Central nervous system by infecting olfactory neurons.
influenza virus A (H5N1) after intranasal inoculation. Acta J Gen Virol. 1996;77(9):2121–2124.
Neuropathol. 2004;108(6):485–492. 43. Myers LJ, Nusbaum KE, Swango LJ, et al. Dysfunction of
29. de Wit E, Siegers JY, Cronin JM, et al. 1918 H1N1 influ- sense of smell caused by canine parainfluenza virus infec-
enza virus replicates and induces proinflammatory cyto- tion in dogs. Am J Vet Res. 1988;49(2):188–190.
kine responses in extrarespiratory tissues of ferrets. 44. Lundh B, Kristensson K, Norrby E. Selective infections of
J Infect Dis. 2018;217(8):1237–1246. olfactory and respiratory epithelium by vesicular stomatitis
30. Leyva-Grado VH, Churchill L, Wu M, et al. Influenza and sendai viruses. Neuropathol Appl Neurobiol.
virus- and cytokine-immunoreactive cells in the murine 1987;13(2):111–122.
olfactory and central autonomic nervous systems before 45. Mori I, Komatsu T, Takeuchi K, et al. Parainfluenza virus
and after illness onset. J Neuroimmunol. 2009;211(1– type 1 infects olfactory neurons and establishes long-term
2):73–83. persistence in the nerve tissue. J Gen Virol.
31. Majde JA, Bohnet SG, Ellis GA, et al. Detection of mouse- 1995;76(5):1251–1254.
adapted human influenza virus in the olfactory bulbs of 46. Altundag A, Temirbekov D, Haci C, et al. Olfactory cleft
mice within hours after intranasal infection. J Neurovirol. width and volume: possible risk factors for postinfectious
2007;13(5):399–409. olfactory dysfunction [published online ahead of print
32. Mori I, Goshima F, Imai Y, et al. Olfactory receptor neu- February 6, 2020]. Laryngoscope. doi:10.1002/lary.28524
rons prevent dissemination of neurovirulent influenza a 47. Wolf A, Liesinger L, Spoerk S, et al. Olfactory cleft pro-
virus into the brain by undergoing virus-induced apopto- teome does not reflect olfactory performance in patients
sis. J Gen Virol. 2002;83(Pt 9):2109–2116. with idiopathic and postinfectious olfactory disorder: a
33. Aronsson F, Robertson B, Ljunggren HG, et al. Invasion pilot study. Sci Rep. 2018;8(1):17554–17512.
and persistence of the neuroadapted influenza virus a/ 48. Yao L, Yi X, Pinto JM, et al. Olfactory cortex and olfac-
WSN/33 in the mouse olfactory system. Viral Immunol. tory bulb volume alterations in patients with post-
2003;16(3):415–423. infectious olfactory loss. Brain Imag Behav.
34. Wheeler DL, Sariol A, Meyerholz DK, et al. Microglia are 2018;12(5):1355–1362.
required for protection against lethal coronavirus enceph- 49. Kim YK, Hong SL, Yoon EJ, et al. Central presentation of
alitis in mice. J Clin Invest. 2018;128(3):931–943. postviral olfactory loss evaluated by positron emission
35. Tsai JC, de Groot L, Pinon JD, et al. Amino acid tomography scan: a pilot study. Am J Rhinol Allergy.
substitutions within the heptad repeat domain 1 of 2012;26(3):204–208.
murine coronavirus spike protein restrict viral antigen 50. Rombaux P, Martinage S, Huart C, et al. Post-infectious
spread in the central nervous system. Virology. olfactory loss: a cohort study and update. B-ENT. 2009;
2003;312(2):369–380. 5(Suppl 13):89–95.
36. Netland J, Meyerholz DK, Moore S, et al. Severe acute 51. Rombaux P, Mouraux A, Bertrand B, et al.
respiratory syndrome coronavirus infection causes neuro- Olfactory function and olfactory bulb volume in patients
nal death in the absence of encephalitis in mice transgenic with postinfectious olfactory loss. Laryngoscope.
for human ACE2. J Virol. 2008;82(15):7264–7275. 2006;116(3):436–439.
37. Schwartz T, Fu L, Lavi E. Differential induction of apo- 52. Yamagishi M, Fujiwara M, Nakamura H. Olfactory
ptosis in demyelinating and nondemyelinating infection by mucosal findings and clinical course in patients with olfac-
mouse hepatitis virus. J Neurovirol. 2002;8(5):392–399. tory disorders following upper respiratory viral infection.
38. Schwob JE, Saha S, Youngentob SL, et al. Intranasal inoc- Rhinology. 1994;32(3):113–118.
ulation with the olfactory bulb line variant of mouse hep- 53. Youngentob SL, Schwob JE, Sheehe PR, et al. Odorant
atitis virus causes extensive destruction of the olfactory threshold following methyl bromide-induced lesions of the
bulb and accelerated turnover of neurons in the olfactory olfactory epithelium. Physiol Behav. 1997;62(6):1241–1252.
epithelium of mice. Chem Senses. 2001;26(8):937–952. 54. Wang JH, Kwon HJ, Jang YJ. Detection of
39. Youngentob SL, Schwob JE, Saha S, et al. Functional parainfluenza virus 3 in turbinate epithelial cells of post-
consequences following infection of the olfactory system viral olfactory dysfunction patients. Laryngoscope.
by intranasal infusion of the olfactory bulb line variant 2007;117(8):1445–1449.
Lee et al. 333

55. Brann D, Tsukahara T, Weinreb C, et al. Non-neural 66. Espinoza JA, Bohmwald K, Cespedes PF, et al. Impaired
expression of SARS-CoV-2 entry genes in the olfactory learning resulting from respiratory syncytial virus infec-
epithelium suggests mechanisms underlying anosmia in tion. Proc Natl Acad Sci USA. 2013;110(22):9112–9117.
COVID-19 patients. bioRxiv. 2020:2020.2003.2025.009084. 67. Volmer R, Prat CM, Le Masson G, et al. Borna disease
56. Hoffmann M, Kleine-Weber H, Schroeder S, et al. SARS- virus infection impairs synaptic plasticity. J Virol.
CoV-2 cell entry depends on ACE2 and TMPRSS2 and is 2007;81(16):8833–8837.
blocked by a clinically proven protease inhibitor. Cell. 68. Martin SJ, Grimwood PD, Morris RG. Synaptic plasticity
2020;181(2):271–207. and memory: an evaluation of the hypothesis. Annu Rev
57. Li W, Moore MJ, Vasilieva N, et al. Angiotensin-convert- Neurosci. 2000;23:649–711.
ing enzyme 2 is a functional receptor for the SARS coro- 69. Damm M, Pikart LK, Reimann H, et al. Olfactory training
navirus. Nature. 2003;426(6965):450–454. is helpful in postinfectious olfactory loss: a randomized,
58. Slotnick B, Cockerham R, Pickett E. Olfaction in olfactory controlled, multicenter study. Laryngoscope. 2014;
bulbectomized rats. J Neurosci. 2004;24(41):9195–9200. 124(4):826–831.
59. Lu XC, Slotnick BM. Olfaction in rats with extensive 70. Hummel T, Rissom K, Reden J, et al. Effects of olfactory
lesions of the olfactory bulbs: implications for odor training in patients with olfactory loss. Laryngoscope.
coding. Neuroscience. 1998;84(3):849–866. 2009;119(3):496–499.
60. Slotnick BM, Gutman LA. Evaluation of intranasal zinc 71. Temmel AF, Quint C, Schickinger-Fischer B, et al.
sulfate treatment on olfactory discrimination in rats. Characteristics of olfactory disorders in relation to major
J Comp Physiol Psychol. 1977;91(4):942–950. causes of olfactory loss. Arch Otolaryngol Head Neck Surg.
61. McGrath N, Anderson NE, Croxson MC, et al. Herpes 2002;128(6):635–641.
simplex encephalitis treated with acyclovir: diagnosis and 72. Lynch G, Rex CS, Gall CM. Synaptic plasticity in early
long term outcome. J Neurol Neurosurg Psychiatry. aging. Ageing Res Rev. 2006;5(3):255–280.
1997;63(3):321–326. 73. Garcia-Segura LM, Chowen JA, Parducz A, et al. Gonadal
62. Rombaux P, Mouraux A, Bertrand B, et al. Can we smell hormones as promoters of structural synaptic plasticity:
without an olfactory bulb? Am J Rhinol. 2007;21(5): cellular mechanisms. Prog Neurobiol. 1994;44(3):279–307.
548–550. 74. Mogensen TH, Paludan SR. Molecular pathways in virus-
63. Weiss T, Soroka T, Gorodisky L, et al. Human olfaction induced cytokine production. Microbiol Mol Biol Rev.
without apparent olfactory bulbs. Neuron. 2020; 2001;65(1):131–150.
105(1):35–45. 75. Sultan B, May LA, Lane AP. The role of TNF-alpha in
64. Kollndorfer K, Jakab A, Mueller CA, et al. Effects of inflammatory olfactory loss. Laryngoscope. 2011;121(11):
chronic peripheral olfactory loss on functional brain net- 2481–2486.
works. Neuroscience. 2015; 310:589–599. 76. Pozharskaya T, Lane AP. Interferon gamma causes olfac-
65. Hosseini S, Wilk E, Michaelsen-Preusse K, et al. Long- tory dysfunction without concomitant neuroepithelial
Term neuroinflammation induced by influenza a virus damage. Int Forum Allergy Rhinol. 2013;3(11):861–865.
infection and the impact on hippocampal neuron morphol-
ogy and function. J Neurosci. 2018;38(12):3060–3080.

You might also like