ArticlePDF AvailableLiterature Review

Immunopathological changes, complications, sequelae and immunological memory in COVID-19 patients

Authors:

Abstract and Figures

Confirmed SARS-CoV-2-caused disease (COVID-19) cases have reached 275.65 million worldwide. Although the majority of COVID-19 patients present mild to moderate symptoms, some have severe complications including death. We first reviewed the pathogenesis on ACE2, a binding receptor of SARS-CoV-2 expressed in multiple organs, and prevalent multinucleate syncytia in the lung tissues of COVID-19 patients. Then, we evaluated the pathological, immunological changes and sequelae in the major organs. Finally, we reviewed the immunological memory after SARS-CoV-2 infection and vaccination. The binding of SARS-Cov-2 to ACE2 receptor results in reduced ACE2 protein levels, which may lead to elevated susceptibility to inflammation, cell death, organ failure, and potentially severe illness. These damages increase the risk of health problems over a long period, which result in many complications. The complications in multiple organs lead to the increased risk of long-term health problems that require additional attention. A multidisciplinary care team is necessary for further management and recovery of the COVID-19 survivors. Many COVID-19 patients will probably make antibodies against SARS-CoV-2 virus for most of their lives, and the immunity against reinfection would last for 3–61 months.
Content may be subject to copyright.
Review article
Immunopathological changes, complications, sequelae and immunological
memory in COVID-19 patients
Liqin Yao
a
, Lingeng Lu
b
,
c
, Wenxue Ma
d
,
*
a
Department of Thyroid and Breast Surgery, The First Afliated Hospital, Huzhou University School of Medicine, Huzhou, Zhejiang, 313000, China
b
Department of Chronic Disease Epidemiology, Yale School of Public Health, School of Medicine, New Haven, CT, 06520, USA
c
Center for Biomedical Data Science and Yale Cancer Center, Yale University, 60 College Street, New Haven, CT, 06520, USA
d
Department of Medicine, Moores Cancer Center and Sanford Stem Cell Clinical Center, University of California San Diego, La Jolla, CA, 92093, USA
ARTICLE INFO
Keywords:
SARS-CoV-2
COVID-19
Immunopathology
Complication
Sequelae
ABSTRACT
Conrmed SARS-CoV-2-caused disease (COVID-19) cases have reached 275.65 million worldwide. Although the
majority of COVID-19 patients present mild to moderate symptoms, some have severe complications including
death. We rst reviewed the pathogenesis on ACE2, a binding receptor of SARS-CoV-2 expressed in multiple
organs, and prevalent multinucleate syncytia in the lung tissues of COVID-19 patients. Then, we evaluated the
pathological, immunological changes and sequelae in the major organs. Finally, we reviewed the immunological
memory after SARS-CoV-2 infection and vaccination. The binding of SARS-Cov-2 to ACE2 receptor results in
reduced ACE2 protein levels, which may lead to elevated susceptibility to inammation, cell death, organ failure,
and potentially severe illness. These damages increase the risk of health problems over a long period, which result
in many complications. The complications in multiple organs lead to the increased risk of long-term health
problems that require additional attention. A multidisciplinary care team is necessary for further management and
recovery of the COVID-19 survivors. Many COVID-19 patients will probably make antibodies against SARS-CoV-2
virus for most of their lives, and the immunity against reinfection would last for 361 months.
1. Introduction
Highly contagious severe acute respiratory syndrome coronavirus 2
(SARS-CoV-2) is a novel coronavirus, which has spread worldwide and
caused over 275.65 million infections (Figure 1) since its outbreak in
Wuhan, China in Dec. 2019 [1]. The SARS-Cov-2-caused infection is
named as COVID-19. Although different measurements, such as lock-
down, social distance, masking, and recent roll-out of vaccines, have
been executed and decreased the initial spread of infection, low vacci-
nation rates, the emergence of COVID-19 virus variants, and the release
of control measurements such as re-opening and unmasking has led to the
resurgence of COVID-19, which was driven predominantly by the Omi-
cron (B.1.1.529) variant of SARS-CoV-2 [2].
By binding to angiotensin-converting enzyme 2 (ACE2), SARS-CoV-2
virus particles are fused with membrane of human epithelial cells mainly
in lung and then enter the cells for u-like systemic replication [3]. ACE2
is ubiquitously expressed in different tissues and organs (e.g., lung, heart,
kidney), which catalyzes hepatocytes-generated angiotensin, playing an
important role in regulating blood pressure. Approximately one third of
patients with COVID-19 are asymptomatic [4]. COVID-19 typically pre-
sents with u-like systemic and/or respiratory symptoms, which include
cough, fever higher than 38 C (100.4 F), myalgia, headache, dyspnea,
sore throat, diarrhea, nausea/vomiting, anosmia, ageusia, dysgeusia,
abdominal pain, runny nose, loss of smell and taste. Severe cases
demonstrate acute respiratory distress syndrome (ARDS) complications
with multiple organ injury and failure, and even death [5,6]. Isolation of
symptomatic and asymptomatic cases and tracing of contacts have been
used in many countries, with additional physical distancing measures
[7]. Specic and effective treatments have not been developed before
COVID-19 vaccines are available, and the standard of care for COVID-19
is focused on the alleviation of symptoms (e.g., oxygen supply, preven-
tion of dehydration, etc.). Although monoclonal antibodies (e.g., Tocili-
zumab, a monoclonal antibody binding to human interleukin-6
receptors) have been used to the patients who are at the high risk of
progressing to severe COVID-19 or being hospitalized [8]. According to
The Centers for Disease Control and Prevention (CDC) and Mayo Clinic,
most of patients with mild symptoms recover from COVID-19 with a
minimal standard of care at home and are back to normal life. Some,
* Corresponding author.
E-mail address: wma@ucsd.edu (W. Ma).
Contents lists available at ScienceDirect
Heliyon
journal homepage: www.cell.com/heliyon
https://doi.org/10.1016/j.heliyon.2022.e09302
Received 6 September 2021; Received in revised form 25 November 2021; Accepted 14 April 2022
2405-8440/Published by Elsevier Ltd. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
Heliyon 8 (2022) e09302
however, have post-viral symptoms (termed as sequelae), such as fatigue,
loss of sense of smell or taste, and brain fog, after recovery [9]. These
sequelae can sometimes last for months or longer and are becoming other
critical health issues of post COVID-19 [10]. These symptoms are the
barriers preventing individuals from returning to work [11].
Over the past 2 years, accumulating knowledge in COVID-19 is very
helpful in beating COVID-19 and containing the pandemic. But we still
need to be on alert for this virus to come back at any time. This review
aims to comprehensively summarize the complications and sequelae of
COVID-19 from the perspectives of its pathogenesis, pathological and
immunological characteristics.
2. Pathogenesis
COVID-19 is a systemic disease with the major involvement of the
lungs, showing diffuse alveolar damage (DAD) and pulmonary throm-
botic microangiopathy [12,13]. Inammation and vascular damage were
also observed in other organs such as heart, liver, kidney, and brain [4,
14]. Multi-organ failure is the major cause of COVID-19-related death
[15]. However, the exact pathogenesis of COVID-19 related deaths re-
mains poorly understood [16,17].
ACE2 is a pivotal cell membrane receptor for the entry of SARS-CoV-
2. Under normal circumstances, ACE2 protein plays an important role in
regulating physiological and biological processes including wound
healing, blood pressure, and inammation, via the renin-angiotensin
system (RAS) pathway [18]. ACE2 enzyme converts angiotensin I into
angiotensin II (ANG II) [19]. ANG II, causing vasoconstriction and an
increase in blood pressure, inammation, increasing damage to the lining
of blood vessels and various types of tissue injury [20]. ANG II can be
broken down by ACE2 into the molecules that counteract the harmful
effects of ANG II, whereas its role is blunted if the virus occupies the
ACE2 receptor on the surface of cells [21]. ACE2 is found in a variety of
human organs [3,6,7], which is highly abundant on type 2 pneumocytes
in the respiratory system [22]. Once SARS-CoV-2 spike protein binds to
ACE2 receptor, the complex is cleaved by transmembrane serine protease
2 (TMPRSS2), leading to membrane fusion, and the virus consequently
enters the cells [23]. Because patients with hypertension, diabetes and
coronary heart disease have higher levels of ACE2 protein, it is not sur-
prising that these health conditions are at risk for COVID-19 [9]. A study
has shown that COVID-19 patients with these conditions have higher
viral load and relatively more loss of epithelial cells with ACE2 expres-
sion [10]. On the other hand, the binding of SARS-CoV-2 spike proteins
to ACE2 receptor can block the breakdown of ANG II proteins by ACE2,
consequently leading to cell damage and inammation [21]. Kuba et al.
demonstrated that reduction of ACE2 expression by the injection of
SARS-CoV spike protein into mice worsened acute lung failure in vivo,
which could be attenuated by interrupting the renin-angiotensin
pathway [24]. However, it is unclear whether the prevalent phenome-
non exists in SARS-CoV-2 infection, although both SARS-CoV and
SARS-CoV-2 are the members of coronavirus family.
In addition to ACE2, another potential molecular mechanism was
recently reported. Zhang et al. demonstrated that generation of multi-
nucleate syncytia was prevalently in the lung tissues of COVID-19 pa-
tients [25]. The authors further demonstrated that a few of human
CD45
þ
cells were enclosed in the special structure of multinucleate
syncytia. They also found a negative correlation between the number of
both syncytia and syncytia containing CD45
þ
cells and the number of
peripheral blood lymphocytes. Their ndings conrmed that the multi-
nucleate syncytia internalize human CD45
þ
cells [25]. Consequently,
internalization of human CD45
þ
cells in the multinucleate syncytia in
COVID-19 patients may be responsible for lymphocytopenia [25].
Microthrombosis is an exclusive clinical feature of COVID-19, and it
was found in 91.3% of dead patients [26]. Microthrombi formation oc-
curs mainly in the pulmonary vasculature but can also occur in other
organs [27]. The disruption of the local reninangiotensin system, endo-
thelial injury (mostly DAD), the complement cascade activation and
powerful thromboinammatory reactions can profoundly result from
entry of the SARS-CoV-2 into microvessels [28]. Particularly, microvas-
cular plugging, ischemia and ultimately organ failure might be led by the
induction of von Willebrand factor [28]. The production of membrane
attack complex (MAC) and culminating in acquired ARDS attribute to the
activation of three complement pathways [29]. Through either direct
Figure 1. Reported COVID-19 cases and death daily worldwide and in the US in the past two weeks. These data are from the Center for Systems Science and En-
gineering (CSSE) at Johns Hopkins University. A. New reported COVID-19 cases globally by day, this number is decreased by 10%. B. New reported death globally by
day, this number is decreased by 8%. C. Cumulative conrmed COVID- 19 death worldwide. D. The conrmed new cases increased by 20% daily in USA. E. New
hospitalized cases daily increased by 14% in USA. F. New death increased by 3% daily in USA.
L. Yao et al. Heliyon 8 (2022) e09302
2
action on platelets and the endothelium or indirect effects of inamma-
tion and coagulopathy, COVID-19 can lead to increased thrombus for-
mation, emboli and hemorrhage [11]. Zhang et al. demonstrated that
platelets can be directly activated by the binding of SARS-CoV-2 spike
protein to ACE2 receptor [30]. Thus, thrombus formation and inam-
matory responses can be further promoted in COVID-19 patients [30]. In
addition, neutrophil extracellular traps trigger thromboinammation in
patients with COVID-19 [12], which leads to vascular thrombosis and
then death.
Aggressive inammatory response and its related hypercoagulability
was found in association with disease severity in patients with COVID-19
and poor outcomes [31]. The exact relationship between the levels of
ACE2, infectivity of SARS-CoV-2, and severity of infection, however, are
not well illustrated [12]. Burki et al. also pointed out that when
COVID-19 cases progress to severe disease and death, men are at a sub-
stantial disadvantage [32]. Evidence from a large study shows that men
have higher concentrations of ACE2 in the blood than women [33,34].
Other factors may include gender behavior (i.e., higher rates of drinking
and smoking among males compared to females), and attitude (i.e., more
females are responsible and more cautious regarding the pandemic than
males) [35]. In addition, sex chromosome genes and hormones (i.e., es-
trogens, progesterone and androgens) contribute to the differential
regulation of immune responses between the sexes [36]. Females have 2
copies of X chromosomes that contain genes related to immune response
and gene silencing, while males only have 1 copy [37]. These genes can
inuence the immune system through regulating many other proteins
(e.g., TLR8, CD40L and CXCR3) that are overexpressed in females, as well
as inuence the response to viral infections and vaccinations [37].
In summary, the characteristics of COVID-19 pathogenesis include
damage of endothelial cells, activation of platelet, thrombogenesis, blood
coagulation disorder, and failure of multiple organs [38,39]. Besides the
respiratory system, cardiovascular, gastrointestinal and renal abnormal-
ities have also been injured [40]. These changes are summarized in
Figure 2.
3. Immunological features of COVID-19
The role of innate and adaptive immunities has not been fully
explored in COVID-19. To date, scientists have mainly focused on
adaptive immunity that are carried out by lymphocytes [41]. Among
patients who died of COVID-19-related respiratory failure, it was found
that DAD with perivascular T-cell inltration was the typical histological
pattern in the peripheral lung [42]. T cell inltration is characterized by
CD4
þ
and CD8
þ
T lymphocytes, which are predominantly distributed in
interstitial spaces, larger bronchioles, and perivascular areas [14]. In
addition to these T cells, CD61
þ
megakaryocytes were also notably
found, which were located within alveolar capillaries, actively producing
platelets [43].
A common feature in severe COVID-19 patients is lymphopenia, but
not in mild cases, which are characterized by drastic reduction of im-
mune cells including CD4
þ
T cells, CD8
þ
T cells, B cells, natural killer
(NK) cells, and a decreased proportion of monocytes, eosinophils, and
basophils [44,45]. In contrast, a gain in neutrophil cell count and the
ratio of neutrophil to lymphocyte are found in association with more
severe disease and poorer clinical outcome [46]. These abnormal
changes are restored in the recovered patients [47].
Cytokine release syndrome (CRS) is an acute systemic inammatory
syndrome caused by a sudden and acute increase of different pro-
inammatory cytokines including IL-6, IL-1, TNF
α
, and interferon in
circulation [48]. The increased levels of cytokines result in converging of
immune cells from circulation to the infection sites, lead to destructive
effects on human tissues, which include instability of the interactions
among endothelial cells, damages of diffuse alveoli, vascular barrier,
capillaries, and multi-organ failure [49]. Lung injury is one of the con-
sequences of cytokine storm, which can progress to acute lung injury or
more severe ARDS [50]. In general, severely CRS is life threatening,
while mild patients with CRS have the symptoms of fever, nausea,
headache, rash, rapid heartbeat, low blood pressure, and difculty in
breathing [51]. Cytokine storm induced-vascular endothelial injury
mediates hypercoagulability in blood vessels and disseminated intra-
vascular coagulation (DIC) [52].
ARDS is a major complication of severe COVID-19 patients [53].
Some studies have showed that the overall mortality rate from ARDS in
COVID-19 patients was 39% (95% CI: 2356%) [54], it was 69% (95%
CI: 6772%) in China, 73% in Poland (95% CI: 5886%), while it was
13% (95% CI: 229%) in Germany [6,54]. Risk factors such as older age,
increased neutrophils, elevated lactate dehydrogenase and D-dimer
levels increase the risk of ARDS and death in those patients hospitalized
with COVID-19 [6,55].
ARDS is the prominent immunopathological feature [6,46]. Among
the patients with SARS-CoV-2 infection, severe inammation in lung may
lead to dysregulation of the renin-angiotensin pathway, which then
progressed to ARDS [56]. Cytokine storm is an essential mechanism of
ARDS along with unregulated systemic inammatory stimulus [57].
Nearly 20% of COVID-19 patients experiencing acute kidney injury (AKI)
and ARDS are related to the cytokine storm [52].
A distinct difference of cytokines, chemokines, and additional im-
mune markers has been found between healthy adults and COVID-19
patients at levels from moderate to severe [58]. Several studies have
indicated that the increased levels of serum proinammatory cytokines
were associated with pulmonary inammation, and lung and organ
failure in COVID-19 disease. It was reported that serum levels of IL-1Ra,
IL-1β, IL-7, IL-8, IL-9, IL-10, FGFβ, G-CSF, GM-CSF, IFNγ, IP10, MCP1,
MIP1A, MIP1B, PDGF, TNF
α
, and VEGF concentration were higher in the
COVID-19 patients at both intensive care unit (ICU) and non-ICU than it
in healthy adults [59,60,61]. In contrast, the serum levels of IL-5,
IL-12p70, IL15, Eotaxin, and RANTES (a chemokine, also known as
CCL5) are similar between COVID-19 patients and healthy adults [59,
62]. Further study showed that plasma concentrations of IL-2, IL-7, IL-10,
G-CSF, IP10, MCP1, MIP1A, and TNF
α
in the ICU patients with COVID-19
are higher than those in non-ICU COVID-19 patients [57]. To investigate
the difference in cytokines, chemokines, and other immune markers
between the COVID-19 patients with moderate and severe disease, Lucas
et al. conducted a longitudinal study to determine the correlation of these
soluble proteins in the patients [63]. The results showed that patients in
the groups of both moderate and severe COVID-19 shared a core
COVID-19 signaturedened by the inammatory cytokines, which
positively correlated with each other, including IL-1
α
, IL-1β, IL-17A,
IL-12 p70, and IFN
α
. In the severe patients with COVID-19, other in-
ammatory factors were observed and dened, which are thrombo-
poietin (TPO), IL-33, IL-16, IL-21, IL-23, IFNλ, eotaxin and eotaxin 3.
These CRS-associated cytokines (e.g., IL-1
α
, IL-1β, IL-6, IL-10, IL-18, and
TNF) are positively correlated with severe disease in patients [63].
Overall, cytokine storm contributes to a poor prognosis [64].
4. Pathological changes and damages
Although COVID-19 mainly affects the respiratory systems, other
systems are also affected, which include cardiovascular, urinary, nervous
system and gastrointestinal tract, etc. [65]. Pathological analyses play a
crucial role in elucidating the pathogenesis of COVID-19, and autopsies
have provided many valuable direct insights of pathological changes
[66].
4.1. Lung
The lung is the main site of injury for SARS-CoV-2 infection [67].
Bilaterally infected lungs show edema and maroon in color, weighing 23
times as much as normal lungs (1939 vs. 6851050 g [68,69,70]. All the
severely infected lungs showed varying degrees of consolidation, pre-
dominantly located at the borders of lobes [70,71]. Many viscous se-
cretions are seen in the section of lung in response to SARS-CoV-2 [43].
L. Yao et al. Heliyon 8 (2022) e09302
3
Pulmonary pathological examination revealed thrombus and micro-
angiopathy in pulmonary small vessels and capillaries, accompanied by
hemorrhage [72]. Other studies also showed severe endothelial injury,
disrupted cell membranes and microthrombi, as well as lymphocytic
interstitial inammation and reactive pneumocyte hyperplasia in the
lung [73]. Thrombosis in pulmonary arteries is associated with hemor-
rhagic lung infarction, and it is found in 2030% of lethal courses [74].
However, venous thromboembolism is more common than arterial
thromboembolism in hospitalized COVID-19 patients [75].
Acute pulmonary infection caused by COVID-19 has distinct char-
acteristics of acute interstitial pneumonia with DAD components,
microvascular involvement of brin deposition and intravascular cap-
ture of neutrophils, as well as microthrombus formation in arterioles
[4,13]. Severe patients presented with ventilation/perfusion imbalance
and respiratory failure emerge [76]. Some patients with respiratory
insufciency present with exudative DAD, accompanied by hyaline
membrane formation [13] and pneumocyte type 2 hyperplasia, which
can progress to DAD at the stage of organization/brosis [72,77]. Firm
thrombi in the peripheral pulmonary vessels along with DAD, have
been the most consistent feature of COVID-19-related lung pathology
[78].
Pulmonary brosis, a symptom of ARDS, is the most severe change in
the lung [79]. Some patients develop brosis [74], which is usually
associated with severe lung injury, permanent pulmonary structural
distortion, and irreversible pulmonary dysfunction [80]. The potential
mechanism underlying SARS-CoV-2-induced pulmonary brosis include
(i) pulmonary brosis is a known sequela to ARDS, 40% of COVID-19
patients develop ARDS, 20% of ARDS cases are severe, (ii) the levels of
TGF-β, TNF-
α
, IL-6, etc. in peripheral blood of severe patients were
increased [81].
Figure 2. Organs affected by COVID-19. When SARS-COV-2 particles enter human body through the respiratory tract, they bind to the ACE2 receptors that are
expressed in multiple organs. With the activation of the immune system, inammation occurs leading to endothelial cell injury, especially the damage of DAD,
resulting in the formation of microthrombosis, ARDS and multiple organ failure via virus, cytokine storm, hypoxia and microthrombosis. IL-6 causes subacute
thyroiditis. SARS-CoV-2 can also cause brain lesions and eye conjunctivitis. In addition to ACE2, individual factors including genetics, immune response, healthy
status, and age also need to be considered in the disparity of COVID-19, and the heterogeneity of COVID-19 manifestations.
L. Yao et al. Heliyon 8 (2022) e09302
4
4.2. Kidney damage
Acute kidney injury (AKI) is one of the most common and most severe
organ complications of COVID-19, which seriously affects the mortality
of patients [82]. Approximately 30% of COVID-19-related deaths had
AKI [83]. Another study showed that 27% of the COVID-19 patients
exhibited AKI [84]. COVID-19 patients with AKI had a three-fold higher
death risk than those without AKI [85]. The ICU/severe patients had
about 30 times higher risk of AKI compared to the non-ICU/severe cases
[86]. Chronic diseases, including hypertension and diabetes, are another
risk factors for kidney damage associated with COVID-19 [87].
SARS-CoV-2 particles has been identied in renal tissue [88,89,90], this
discovery suggested that human kidneys were directly infected by
SARS-CoV-2, leading to pathogenesis of renal tubules and AKI [84].
Nearly 20% of patients experiencing AKI are caused by cytokine storm
[52]. In most cases, the kidneys display acute tubular injury, with the
characteristics of lymphocyte depletion in the lymph nodes spleen, and
hyperplastic adrenal glands [74], while CD68
þ
macrophages inltration
into the tubule-interstitium are observed [84]. The patients present
abnormal renal functions such as increased serum creatinine (sCr), blood
urea nitrogen (BUN), D-dimer, proteinuria, and hematuria [86].
Renal changes include ne granular kidneys and focal cortical scars
[91]. These changes were caused by arteriolosclerosis, mesangial scle-
rosis, hypercellularity, and focal global glomerulosclerosis [91]. How-
ever, no inammatory or ischemic changes were found in the medulla
section [91]. An occasional adrenal cortical nodule and a papillary thy-
roid adenocarcinoma were also detected [91]. COVID-19 patients
develop extensive glomerular and tubular diseases [92]. Acute renal
tubular injury is the most common type of injury in both live kidney
biopsies and autopsy [93].
In summary, current evidences suggest that the causes of renal injury
in patients with COVID-19 include hypovolemia, ARDS associated
cytokine storm, and direct viral invasion as seen on renal autopsy
ndings [94]. COVID-19 may leave people with lasting damage to their
kidneys, among many other organs. The possibilities are the most likely
due to: (1) presence of ACE2 receptors in kidney cells allowing
SARS-CoV-2 to bind them, invade, potentially damaging kidney tissues;
(2) too little oxygen in blood causes kidneys to malfunction, (3) cyto-
kine storms destroy kidney tissue; and (4) tiny clots form in the
bloodstream, blocking the smallest blood vessels in the kidney and
impairing renal function.
4.3. Neurologic disorder
Some COVID-19 patients may develop cerebral edema, neuronal
degeneration, encephalitis, meningoencephalitis, acute disseminated
encephalomyelitis, Guillain-Barr
e Syndrome, Bickerstaff's brainstem en-
cephalitis, Miller Fisher syndrome, polyneuritis, myositis/rhabdomyol-
ysis, toxic encephalopathy, and stroke [95].
An autopsy report revealed the presence of SARS-CoV2 particles in
the brain tissues of a COVID-19 patient [96]. SARS-CoV-2 has been
detected in both the cerebrospinal uid (CSF) and brain parenchyma of
many patients [97]. Brain tissue oedema and partial neurodegeneration
have also been observed [98], showing increased weight (~1221 g) and
hydrocephalus ex vacuo [91]. The pathological changes in brain prob-
ably show the consequence of both direct cytopathic effects due to
SARS-CoV-2 replication and indirect effects such as respiratory failure,
injurious cytokine reaction, reduced immune response and cerebrovas-
cular accidents caused by viral infection [99].
The brainstem is another part infected by SARS-CoV2 in CNS. Au-
topsy studies provide evidence for the presence of SARS-CoV-2 RNA and
proteins in the brainstem [100]. ACE2 receptor expression is relatively
higher in the brainstem than in other brain regions [100]. In addition,
another receptor of SARS-CoV-2 named neuropilin-1 is also expressed in
the brainstem [101]. Moreover, the brainstem is susceptible to damages
derived from either pathological immune or vascular activation [100].
Therefore, patients with encephalitis, encephalomyelitis, and brainstem
encephalitis recover slowly and have a high mortality rate [102].
4.4. Heart damage
The heart is one of the most common organs affected by COVID-19;
however, the nature and scope of cardiac pathology has been contro-
versial [103]. A study reported that cardiac histopathological ndings
associated with COVID-19 are very common, but myocarditis is rare
(<2%) [104]. Another study reported that the existence of SARS-CoV-2
in the myocardium was found in 47% of 316 deceased with COVID-19
[105], while cardiac pathological changes contributed to the death of
4.7% (15/316) cases [105]. Furthermore, postmortem examination
demonstrated other pathological changes including cardiac dilatation
(20%), acute ischemia (8%), intracardiac thrombi (2.5%), pericardial
effusion (2.5%), and myocarditis (1.5%) [105]. Myocardial pathological
examination revealed hypertrophy of myocardial cells with interstitial,
and perivascular brous tissue, but no acute ischemic changes or in-
ammatory inltration were observed [106]. Moreover, Zhou et al. re-
ported that 48 % of the patients (91/191) with COVID-19 had
comorbidities. Hypertension is the most common comorbidity (58/191,
30%), followed by diabetes (36/191, 19%) and coronary artery disease
(15/191, 8%) [107].
Troponin is a marker of cardiomyocyte damage or injury. The ma-
jority of patients with an acute myocardial infarction have an elevated
troponins within 23h[108]. In the case of COVID-19, myocardial injury
can occur, which is dened as elevated troponin level [109]. Among the
patients in ICU, the levels of cardiac troponin are signicantly higher in
those with more severe infections [110]. Overall, the potential mecha-
nisms of myocardial tissue damage include direct myocardial injury
caused by SARS-CoV2 (i.e., viral myocarditis), systemic hyper-
inammatory response (i.e., CRS), hypoxemia, downregulation of ACE2,
endothelialitis induced by systemic virus, and myocardial infarction
[111].
4.5. Liver damage
More than 1/3 patients with COVID-19 develop liver damage that is
manifested by increased liver enzymes [112]. SARS-CoV-2 is mostly
considered unlikely to cause liver infection because ACE2 expression is
very low in liver cells. In the individuals with increased aminotransfer-
ases, autopsy liver biopsies were randomly obtained. Surprisingly, typical
coronavirus particles with spike structures were found in the cytoplasm
of liver cells in the patients with COVID-19 [113].
The distribution of ACE2 receptors is generally considered to be
consistent with the distribution of infected organs [114]. However, the
presence of ACE2 expression was signicantly inconsistent with multiple
organs targeted by SARS-CoV-2. Therefore, it is speculated that another
Extra-ACE2 receptor or co-receptor may exists. Another possibility is that
ACE2 expression in hepatocytes may be upregulated in response to viral
invasion [115]. Analysis of ACE2 expression in
post-SARS-CoV-2-infection hepatocytes will be interesting and helpful to
untangle this inconsistence.
A meta-analysis of 18 studies from 7 countries showed that the
combined prevalence of liver histopathological ndings was: hepatic
steatosis 55.1%, hepatic sinus congestion 34.7%, vascular thrombosis
29.4%, brosis 20.5%, Kupffer cell hyperplasia 13.5%, portal vein
inammation 13.2%, and lobular inammation 11.6% [116]. In addi-
tion, venous outow obstruction, portal vein sclerosis, portal vein her-
niation, abnormal periportal vessels, hemophagocytosis, and necrosis
were also found [116]. In summary, the high prevalence of hepatic
steatosis and vascular thrombosis are the major histological features of
the liver in COVID-19 patients [116]. The liver steatosis, liver cell ne-
crosis, portal inammation, and proliferation of Kupffer cells are also
very common [74].
L. Yao et al. Heliyon 8 (2022) e09302
5
During the clinic evaluation of COVID-19, liver injury has been
observed in a large number of patients, especially in those who are in
severe or critical conditions. The pathologic changes reported were a
slight increase in sinusoidal lymphocytic inltration, sinusoidal dilata-
tion, steatosis and multifocal hepatic necrosis [117].
4.6. Thyroid damage
SARS-CoV-2 infects host cells with ACE2-binding TMPRSS2 as the key
molecular complex. Interestingly, ACE2 and TMPRSS2 were expressed at
higher levels in the thyroid glands than in the lungs [118]. Subacute
thyroiditis is a thyroid disease of viral or post-viral origin. A study re-
ported that about two-thirds (66.7%) of COVID-19 patients developed
subacute thyroiditis among 27 cases, including 11.1% of the COVID-19
patients required hospitalization, and 83.3% of the cases had subacute
thyroiditis after COVID-19 [119]. Accordingly, patients who develop
thyroid inammation during acute COVID-19 may develop subacute
thyroiditis months later, even after thyroid function has returned to
normal.
COVID-19 may be associated with a high risk of hyperthyroidism
associated with systemic immune activation caused by the SARS-CoV-2
infection, and thus plays a pivotal role in inducing hyperthyroidism of
Gravesdisease [120]. On this point, Lania et al. demonstrated that hy-
perthyroidism was signicantly associated with higher IL-6 levels [121].
Similarly, another study showed that 75% of COVID-19 patients devel-
oped thyroid abnormalities and higher IL-6 levels (P<0.01) [122]. Thus,
there are several potential thyroid outcomes in patients with COVID-19,
such as thyrotoxicosis, low-T3 syndrome and subacute thyroiditis [123].
Taken together, COVID-19 are negatively impacting the thyroid. How-
ever, further investigations are required to validate the causal relation-
ship between subacute thyroiditis and COVID-19.
4.7. Gastrointestinal tract
Considering that gastrointestinal epithelial cells express ACE2, SARS-
CoV-2 may also affect gastrointestinal tract. Mao and colleagues reported
that 15% of the patients with COVID-19 had gastrointestinal symptoms,
and the typical gastrointestinal symptoms were nausea, vomiting, diar-
rhea, and anorexia [124]. In addition, intestinal involvement of
COVID-19 can be associated with intestinal ischemia, caused by shock or
local thrombosis [74].
4.8. Eyes
Conjunctivitis is the most common type of eye infection among
COVID-19 patients [125]. SARS-CoV-2 particles were found from
conjunctival swabs and tears of COVID-19 patients, additionally,
SARS-CoV-2 RNA was detected in conjunctival, anterior corneal, poste-
rior corneal, and vitreous from the deceased with COVID-19 [126]. In a
case series report from the patients with COVID-19, some of them had
ocular manifestations, including epiphora, conjunctival congestion, or
chemosis [127]. This positive SARS-CoV-2 nding is common in severe
COVID-19 patients. RT-PCR results of nasopharyngeal swabs and
conjunctival swabs were positive for SARS-CoV-2, blood test results
showed signicant value changes among the COVID-19 patients with
ocular abnormalities [128]. In addition, the presence of SARS-CoV-2
RNA on ocular surface suggests that the eye may be a site of viral repli-
cation [129]. SARS-CoV-2 proteins (e.g., spike and envelope proteins)
were identied in the corneal epithelium undisinfected with
povidone-iodine (PVPI) [126].
5. Complications
Most COVID-19 patients can recover within 26 weeks. But some of
them still have symptoms after recovery. COVID-19 patients can expe-
rience multiple complications during infection. About 1 in 6 people with
COVID-19 will develop complications, which can be life-threatening. The
result of a cohort demonstrated that COVID-19 survivors mainly had
fatigue, muscle weakness, sleep difculties, anxiety, depression, adjust-
ment disorders, and tic disorders. During hospitalization, patients with
severe COVID-19 had severely impaired pulmonary dispersion and
abnormal chest imaging ndings [130]. Another report indicated that
80% of the COVID-19 patients developed one or more long-term symp-
toms. The top ve most common symptoms were fatigue (58%), head-
ache (44%), attention disorder (27%), hair loss (25%), and dyspnea
(24%) [131].
According to the pathogenesis, pathological and immunological
changes, these complications may include acute respiratory failure,
pneumonia, ARDS, acute liver injury, acute myocardial injury, acute
kidney injury, secondary infection, septic shock, DIC, blood clots, multi-
system inammatory syndrome in children, chronic fatigue, and rhab-
domyolysis may adversely affect the prognosis. Some complications may
be unanticipated. In severe cases of COVID-19, cytokine storm-induced
thrombotic complications including DIC are a prominent feature [78].
5.1. Pulmonary complications
Reported complications of COVID-19 include long period damage to
tiny air alveoli in the lungs and the resulting scar tissue, which can lead to
long-term breathing problems. About 3040% of COVID-19 hospitaliza-
tion and nearly 70% of fatal cases develop ARDS [132]. ARDS occurs in
42% of COVID-19 patients, and 6181% of the patients required inten-
sive care [55].
5.2. Cardiac concerns
Imaging tests taken months after recovery from COVID-19 showed
that even people with only mild symptoms of COVID-19 suffered lasting
damage to the heart muscle. This damage can be caused by the formation
of blood clots in both large and tiny vessels. This could increase the risk of
heart failure or other cardiac complications in the future. Data on long-
term cardiovascular complications of COVID-19 are scarce. However, it
can be learned from the experience of dealing with other types of
myocardial injury. In a follow-up study of patients with acute myocarditis
(mean age 40.2 years), the authors noted that the rate of hospitalization
associated with heart failure ranged from 6% to 8% [133]. Patients with
viral myocarditis may be associated with COVID-19 related myocarditis
and/or brosis due to inammation (regional or local) associated with
acute disease.
5.3. Brain
While SARS-CoV-2 virus mainly targets the respiratory system, pa-
tients and survivors of COVID-19 can also experience neurological
changes and develop neurological and psychiatric symptoms. SARS-CoV-
2 bind directly to the cells in neural tissue [95]. 73% of hospitalized
COVID-19 patients had neurological symptoms, mainly headache, my-
algias and impaired consciousness [134]. These symptoms including
anosmia, hypogeusia, headache, nausea and altered consciousness are
very common, but more severe and specic conditions have also been
clinically reported, such as acute cerebrovascular accident, encephalitis,
and demyelinating disease. Even in young people, COVID-19 can also
cause strokes, seizures, and Guillain-Barre syndrome according to Mayo
Clinic. Additionally, COVID-19 may also raise the risk of suffering Par-
kinson's disease and Alzheimer's disease.
The manifestation in central nervous system (CNS) is caused by direct
viral invasion of CNS or by indirect mechanisms [99]. Studies have
shown that SARS-CoV-2 can get access into CNS through olfactory nerves
and even stretch to the medulla. The virus impairs CNS through either
direct viral damage or immunopathological damage to nerve cells.
Neurological symptoms involving the CNS can lead to acute or longer
period consequences [135]. Wyss-Coray and colleagues compared
L. Yao et al. Heliyon 8 (2022) e09302
6
samples of the frontal cortex and choroid plexus from control and
COVID-19 patients, they observed extensive cellular perturbations, with
T cell inltrating into the parenchyma. They identied subpopulations of
microglia and astrocyte associated with COVID-19 disease that share
genetic signatures with pathological cell states in human neurodegen-
erative disease, such as cognitive disorders, schizophrenia, and depres-
sion [136]. These ndings may help to interpret the symptoms of brain
fog, fatigue, and other neurological and psychiatric symptoms.
5.4. Kidney
Kidney complications are relatively common. As above mentioned,
AKI is a lethal complication in COVID-19 patients [137]. In addition,
rhabdomyolysis is also a fatal syndrome caused by the breakdown of
skeletal muscle bers and leakage of muscle contents into the systemic
circulation. However, this can lead to serious complications of renal
failure. A study has reported rhabdomyolysis in 10 COVID-19 patients
[138].
6. Sequelae or long-standing effects of COVID-19
Much remain unknown about the long period effects of COVID-19 on
human health. Sequelae of SARS-CoV-2 infection include fatigue, dys-
pnea, chest pain, cognitive impairment, joint pain, and reduced quality of
life [139]. Daugherty et al. reported that 14% of the adult patients (65
years) with COVID-19 had at least one clinical sequelae. These sequelae
including chronic respiratory failure, cardiac arrythmia, hypercoagula-
bility, encephalopathy, peripheral neuropathy, amnesia, diabetes,
abnormal liver function tests, myocarditis, anxiety, and fatigue need
medical care after passing through the acute phase of COVID-19 [140].
SARS-CoV-2 infection-induced cellular damage, a strong innate immune
response that produces inammatory cytokines, and a pro-coagulant
state may contribute to these sequelae [141,142]. COVID-19 infection
can cause lasting damage to kidneys.
Close monitoring of the COVID-19 patients is recommended to un-
derstand the function of their organs after recovery. Many medical cen-
ters are going to set up and open specialist clinics to provide better cares
for the individuals who have persistent symptoms or related illnesses
after recovering from COVID-19. In general, most COVID-19 patients
recover quickly; however, the potential long standing problems make it
more important in addition to reduce the spread of COVID-19. Extreme
fatigue with sleep changes, post-exercise nerve failure, multiregional
cognitive dysfunction, persistent headache, demyelinating syndromes,
peripheral neuropathy, and autonomic nervous instability are notable
features of post-viral syndromes; similar concerns exist for people with
persistent symptoms of COVID-19 [9]. Currently, there is no curative
treatment for post-viral syndromes. The aim of treatment is symptomatic
treatment to relieve symptoms.
6.1. Problems with mood, fatigue, and chronic brain disorders
Patients with severe COVID-19 who are admitted to an ICU might
experience mechanical assistance for breathing. The patients who had
ventilator-aid experience are more likely to experience post-traumatic
stress syndrome, depression, and anxiety. Early intervention should
begin to reduce the risk for posttraumatic stress syndrome (PTSS) after
hospitalization [143]. In addition, an extensive cellular perturbation was
discovered in the brains of COVID-19 patients, may be due to inam-
mation caused by the infection. T cell inltration was observed in the
parenchyma, and these disturbances overlap with chronic cognitive
impairment, schizophrenia, and depression [87].
6.2. Clots in blood vessel
SARS-CoV-2 infection makes blood cells at high risk of clotting and
forming blood clots in many organs (e.g., lungs, heart, legs, liver, and
kidneys, etc.). While large clots can cause heart attacks and strokes,
smaller clots can damage heart by blocking capillaries in cardiac muscle.
COVID-19 can also damage the lining of blood vessels, resulting in vessel
leaking, and potentially long-term issues with both the liver and kidneys.
A previous study reported that blood clots in the veins occurred in 20% of
the COVID-19 patients, and in 31% patients in the ICU. Clots in vein or
thrombosis in deep vein can circulate to the lungs and form pulmonary
embolism, which consequently result in higher risk of death. An ampu-
tation may be needed if the clots are not treated timely with either a
surgical or interventional treatment [144].
7. Prevention and immunological memory
Isolation and quarantine of infected individuals are effective ap-
proaches in controlling respiratory infectious diseases aside from vacci-
nation of susceptible persons. Nevertheless, a key blind spot in the
pandemic is asymptomatic infection in the community and pre-
symptomatic viral transmission, which posed a major barrier in halting
the spread of the virus. Although large-scale screening with RNA detec-
tion or rapid antibody detection are helpful in identifying these potential
infection sources; however, society and economic costs are huge.
The publication of SARS-CoV-2's genome sequence has accelerated
the development of vaccines, particularly novel RNA vaccines [145]. As
of Aug. 31, 2021, WHO has authorized the following COVID-19 vaccines
for emergency use (EUA). All the vaccines under EUA (17) together with
2 more with rolling data submission (89) were summarized in Table 1.
As of Nov. 23, 2021, 132 vaccines are still in clinical development and
194 vaccines are in pre-clinical development according to WHO (https://
www.who.int/publications/m/item/draft-landscape-of-covid-19-candi
date-vaccines). Novel delivery systems are under developing, for
example, exosomes-mediated mRNA delivery [146], and exosomes car-
rying immunogenic viral peptides from COVID-19 patients (www
.covidx.eu/exocovac).
mRNA vaccine (e.g., BNT162b/Pzer, or mRNA-1273/Moderna) uses
genetically engineered mRNA derived from S-protein of SARS-CoV-2 and
it is delivered through lipid nanoparticles. Once the mRNA vaccine is
administered, the lipid nanoparticles encapsulated mRNA are taken up
by cells. The mRNA is then translated into the S-protein within the
recipient cell cytoplasm. S-protein are next transported to the cytoplasm
and cell membrane of the host cells. The S-protein can be recognized by
the innate immune cells including NK cells, eosinophils, etc. and
phagocytic cells (macrophages, neutrophils, and dendritic cells) for an-
tigen presentation; adaptive immune cells (T- and B cells) are then trig-
gered to recognize and memorize how to attack the virus that causes
COVID-19 if infected again in the future.
Protein vaccines (e.g., inactivated cronavac/Sinovac and Sinopharm)
include harmless protein pieces, which are derived from SARS-CoV-2.
After vaccination, the immune system recognizes the proteins. B cells
produce antibodies and T-lymphocytes recognizes and memorize the
proteins, and therefore attack the virus once invaded in the future.
Vector-based vaccines (e.g., Ad26.COV2. S/Johnson &Johnson) use a
modied version of a different virus as a vector to deliver the nucleic acid
coding S-protein to the recipient's host cells. Once administrated, the host
will build immune responses including T- and B-lymphocytes.
Duration of immunological memory (memory B cells, memory T cells
including CD4þT cells, and/or memory CD8þT cells, as well as anti-
bodies) after SARS-CoV-2 infection is unclear. Many COVID-19 patients
will probably make life time antibodies against SARS-CoV-2 [147].
Because Turner et al. recently reported that long-lived anti-
body-producing cells (plasma cells) were identied in the bone marrow
from the patients who recovered from COVID-19 [148,149]. These
people infected with SARS-CoV-2 induce robust antigen-specic, long--
lived humoral immune memory [148]. Another study investigated
immunological memory by measuring the titer of antibodies against
SARS-CoV-2 specic antigens, and immune cells in the blood. The results
demonstrated that 95% of subjects remained immunological memory for
L. Yao et al. Heliyon 8 (2022) e09302
7
8 months since infection [150]. Another similar study showed specic
immunological memory for 5 months after recovery [151]. In addition, a
very recent study reported that unvaccinated people should have im-
munity against reinfection for 361 months after getting infected with
COVID-19 [152]. A further study has demonstrated that SARS-CoV-2
antibodies remain stable for at least 7 months after an infection with
the virus [153]. The relatively short immunological memory suggests
that COVID-19 infection-induced immune memory will not last too long.
Similarly, COVID-19 vaccine-induced immune memory may have a
similar situation, suggesting that people may need the fourth dose as a
booster. Before the rollout of a large-scale of booster vaccination, it is a
priority to understand who need the booster vaccination, how the
booster vaccination affects long period health, and how long the
immunological memory lasts for the booster vaccination.
Breakthrough and second infection of COVID-19 indicate that it is
unknown what antibody levels are able to prevent reinfection and how
Table 1. Status of COVID-19 vaccines within WHO EUA.
Vaccine Brand Vaccine Name Vaccine Types/Platform EOI
Accepted
EUA Date Age to get
vaccines
Doses Interval
1Pzer-BioNTech BNT162b2 Nucleoside modied
mRNA
Yes Dec. 31, 2020 12 years 3 shots 3 weeks for
the rst 2 doses,
6 months for
the booster
2 Moderna mRNA-1273 mRNA encapsulated in
lipid nanoparticle (LNP)
Yes Apr. 30, 2021 18 years 3 shots 4 weeks for
the rst 2 doses,
6 months for
the booster
3 Janssen Ad26.COV2. S Vectored vaccine
encoding the (SARS-CoV-
2) Spike (S) protein
Yes Mar. 12, 2021 18 years 2 shot 2 months after
completing the
primary
vaccination
4 Astrazeneca-Oxford
University
AZD1222 Adenoviral vector
encoding the Spike
protein antigen of the
SARS-CoV-2
Yes Apr. 16, 2021 18 years 2 shots 4 weeks
5 Serum Institute
of India
Covishield
(ChAdOx1_nCoV- 19)
Adenoviral vector
encoding the Spike
protein antigen of the
SARS-CoV-2
Yes Feb. 15, 2021 18 years 2 shots 812 weeks
6 Sinopharm SARS-CoV-2 Vaccine
(Vero Cell), Inactivated
(lnCoV)
Inactivated, produced in
Vero cells
Yes May 7, 2021 18 years 2 shots 24 weeks
7 Sinovac COVID-19 Vaccine (Vero
Cell), Inactivated/
Coronavac
Inactivated, produced in
Vero cells
Yes Jun. 1, 2021 18 years 2 shots 24 weeks
8 The Gamaleya
National Center
Sputnik V
Russian MRNA, Human
Adenovirus Vector- based
Covid-19 vaccine
Additional
information
submitted
Rolling submission
of clinical and CMC
data has started.
Anticipated date will be
set once all data is submitted,
and follow-up of inspection
observations completed.
18 years 2 shots 3 weeks
9 Bharat Biotech,
India
SARS-CoV-2 Vaccine,
Inactivated (Vero Cell)/
COVAXIN
DCGI, Whole-Virion
Inactivated Vero Cell
Yes Rolling data started
06 July 2021. Decision
date: To be conrmed.
12 years 2 shots 48 weeks
Abbreviation: WHO: The World Health Organization; EUA: Emergency Use Authorization; EOI: Expression of Interest; DCGI: the Drugs Controller General of India.
Table 2. SARS-CoV-2 variants of concern (VOC).
Bango Lineage Name WHO Label First Identied Spike Protein Substitutions
B.1.1.7 Alpha United Kingdom 69del, 70del, 144del, (E484K*), (S494P*), N501Y,
A570D, D614G, P681H, T716I, S982A, D1118H (K1191N*)
B.1.351
B.1.351.2, B.1.351.3
Beta South Africa D80A, D215G, 241del, 242del, 243del, K417N, E484K,
N501Y, D614G, A701V
B.1.617.2
AY.1, AY.2, AY.3, AY.4,
AY.5, AY.6, AY.7, AY.8,
AY.9, AY.10, AY.11, AY.12
Delta India T19R, (V70F*), T95I, G142D, E156-, F157-, R158G, (A222V*),
(W258L*), (K417N*), L452R, T478K, D614G, P681R, D950N
P.1
P.1.1,
P.1.2
Gamma Japan/Brazil L18F, T20N, P26S, D138Y, R190S, K417T, E484K, N501Y,
D614G, H655Y, T1027I
B.1.1.529 Omicron South Africa and
Botswana
A67V, Δ6970, T95I, G142D, Δ143145, N211I, Δ212, ins215EPE,
G339D, S371L, S373P, S375F, K417N, N440K, G446S, S477N, T478K,
E484A, Q493R, G496S, Q498R, N501Y, Y505H, T547K, D614G, H655Y,
N679K, P681H, N764K, D796Y, N856K, Q954H, N969K, L981F
L. Yao et al. Heliyon 8 (2022) e09302
8
long it can last against the virus, even if the neutralizing antibodies
produced by the COVID-19 vaccines are protective, protecting vulnerable
populations from severe disease, and limiting the spread of the virus.
8. Conclusions
COVID-19 is a systemic disease that mainly affects the lungs, but often
other organs as well. COVID-19 is a new virus infection disease, and
nobody has adaptive immunity against it. Infected individuals were re-
ported to have a broad range of symptoms ranging from mild to severe
sicknesses. Minor symptoms include fever or chills, cough, shortness of
breath, fatigue muscle or body aches, headache, loss of taste or smell,
sore throat, congestion or runny nose, nausea or vomiting, diarrhea, joint
pain, and chest pain. Other concerns include cognitive disorders, dif-
culty in focusing, depression, myalgia, headache, fast heartbeat, and
sporadic fever. Severe illness include difculty in breathing, constant
pain or compression in the chest, and cyanosis caused by hypoxemia, etc.
Organ damage caused by SARS-CoV-2 increase the risk of long period
health problems, leading to complications and sequelae that require
special attention to pay. 95% of the COVID-19 patients retained immu-
nological memory for up to 8 months, a booster of COVID-19 vaccine may
need eight months after being fully vaccinated.
9. Outlook
The complete eradication of SARS-CoV-2 remains challenging at least
in a short term for a variety of reasons. SARS-CoV-2 is going to be present
for a while, especially the emergence of the mutant viruses such as Delta
and Omicron variants, etc. Even if we eventually eliminate SARS-CoV-2,
there might be other novel coronavirus strains in nature waiting their
turn. Given the large number of incident cases are diagnosed and a
certain considerable number of COVID-19-related death occurring every
day globally, the intellectual collaboration on SARS-CoV-2 and the var-
iants must continue, requiring multidisciplinary knowledge collabora-
tion not only between academia but also between industry.
Generally, herd immunity can be reached if over 80% of individuals
are vaccinated. Vaccination coverage is still far below this level, partic-
ularly in undeveloped countries. Severe inequality exists in vaccine re-
sources and supplies around the world. Even in the developed countries,
although they have enriched vaccine resources with strong economic and
treasure support, the goal of herd immunity has not been all reached. One
of the potential reasons for those who are reluctant to get vaccinated is
the concern for long-term effects of vaccines since all vaccines are EUA
except recently FDA-approved Pzer/BNT and Moderna vaccines. It is
still debatable whether mRNA from the VOVID-19 vaccines can be in-
tegrated into human genome DNA or not [154,155]. A study demon-
strated that chimeric transcription of SARS-CoV-2 and infected host cell
RNA were observed, suggesting the ability of SARS-CoV-2 RNA to be
reverse-transcribed and integrated into the genome of the infected host
cells [156]. Another study showed no evidence that SARS-CoV-2 mRNA
was able to be reverse transcribed and integrated into host cells [157].
Additional possibility is that some individuals doubt the transparency of
vaccines. Injected lipid nanoparticles encapsulated mRNA vaccines
invoke immune response when they are engulfed by antigen-presenting
cells (e.g., macrophage and dendritic cells). However, lipid nano-
particles are non-specic vesicles, which can be engulfed by other types
of cells (e.g., cardiocytes, endothelial cells, etc.) even after the release of
packaged mRNAs from macrophages and dendritic cells. Spike-protein
expressed on cardiocytes and endothelial cells may lead to myocarditis
and thrombosis. Thus, it is necessary to develop a relatively more specic
vaccine. For example, by modifying proteins in lipid membrane, which
can be engulfed by a certain type of cells through ligand-receptor
mechanisms or targeting with certain antibodies. In addition, adjuvants
are usually used in vaccine manufacture. Whereas AS03 (Adjuvant Sys-
tem 03), and MF59 (an oil-in-water emulsion of squalene oil) are
squalene-based immunologic adjuvants that have been used in licensed
vaccine products by GSK (GlaxoSmithKline) and Novartis, respectively
[158], it is unknown what adjuvants are used in mRNA vaccines or other
types of vaccines. Breakthroughs and the emergence of new variants
remind the scientic communities that we still have much uncover and
improve regarding future vaccines.
Lockdown is an effective measurement to suppress the spreading.
However, lengthy period lockdown brings a heavy society and economic
burden. Re-opening is necessary and on the way around the world. With
new variants of SARS-CoV-2 emerging, e.g., Omicron (B.1.1.529) and
Delta (B.1.617.2) variants, which can double hospitalization risk
compared to the Alpha variant (B.1.1.7), however, new strategies are
needed to re-open.
As a major target in controlling SARS-CoV-2, mutations in the re-
ceptor binding domain (RBD) are being of particular concern, which may
substantially weaken RBD-binding antibodies. It is a gap to ll how much
cross-protection exists against variant strains following the original
vaccination or infection [159]. The Delta variant has 3 RBD mutations,
417, 452 and 478, respectively, which change the conformation of S
protein, and may aid immune escape. Novel vaccines may be necessary,
and mRNA vaccines may have some advantages by including the muta-
tion containing ORFs. The SARS-CoV-2 variants of concern have been
listed in Table 2.
SARS-CoV-2 may also infect other species besides human. Certain
strains of wild type (i.e., non-hACE2 bearing) mice are vulnerable to the
SARS-CoV-2 variants, e.g., B.1.1.7, B.1.351, and P.1 can infect mice via
the endogenous mouse ACE2 receptor (https://www.jax.org/jax-mice
-and-services/sarscov2-test-kit).
We still have some key unknown questions to be addressed: how soon
after infection do T cells become activated to stop the spread of the virus?
How long do T cells retain SARS-CoV-2 memory? These would be of
concern to us because traditional vaccines focus on producing neutral-
izing antibodies. As our understanding of the interaction between the
immune system and SARS-CoV-2 continues to grow, it is important to go
beyond neutralizing antibodies to pursue T cell immunity.
Each newly emerging variant raises concerns: Will the disease course
be changed? How will the immune system respond to new variants? Can
the variant evade a pre-existing immune response from previous infec-
tion or vaccination, or pre-existing vaccines are still valid or not?
Apart from the above SARS-CoV-2 variants of concern, the variants of
interest (VOI) have also caught the attention of scientists (Table 3).
Among the VOI, Mu was just designated by the WHO on August 30, 2021.
Outbreaks of the Mu variant has been reported in South America and
Table 3. SARS-CoV-2 variants of interest (VOI).
Pango Lineage Name WHO Label First Identied Spike Protein Substitution
B.1.525 Eta UK/Nigeria A67V, 69del, 70del, 144del, E484K, D614G, Q677H, F888L
B.1.526 Iota USA/New York L5F, (D80G*), T95I, (Y144-*), (F157S*), D253G, (L452R*), (S477N*),
E484K, D614G, A701V, (T859N*), (D950H*), (Q957R*)
B.1.617.1 Kappa India (T95I), G142D, E154K, L452R, E484Q, D614G, P681R, Q1071H
B.1.617.3 None India T19R, G142D, L452R, E484Q, D614G, P681R, D950N
B.1.621
B.1.621.1
Mu Columbia N/A
L. Yao et al. Heliyon 8 (2022) e09302
9
Europe, a descendent (B.1.621.1) of Mu has already been detected in 43
countries. Although this Mu belongs to VOI, but it has mutations that
indicate a risk of resistance to the current COVID-19 vaccines. Table 3
summarized the VOI according to the previous information from the
CDC. Currently, no SARS-CoV-2 variants are designated as VOI.
Scientic source-tracking of SARS-CoV-2 is another important issue in
prevention and control of SARS-CoV-2 infection. With accumulating
knowledges in this novel emerging virus, in addition to the rst wave of
COVID-19 cases reported in Wuhan, China in Dec. 2019 [57], COVID-19
was also spreading in other countries. Alteri, etc. described that the
highest numbers of SARS-CoV-2 cases was found in Lombardy, Italy from
February to April 2020. Over 16,000 deaths were reported in Lombardy
in a couple of weeks during the time frame [160]. After analyzing 346
whole SARS-CoV-2 genomes, seven viral lineages were found; at least
two were likely originated in Lombardy, Italy [160].
Declarations
Author contribution statement
All authors listed have signicantly contributed to the development
and the writing of this article.
Funding statement
This research did not receive any specic grant from funding agencies
in the public, commercial, or not-for-prot sectors.
Data availability statement
No data was used for the research described in the article.
Declaration of interests statement
The authors declare no conict of interest.
Additional information
No additional information is available for this paper.
Acknowledgements
The authors would like to thank Ms. Amy Yiming Ma for her critical
reading and editing.
References
[1] A.G. Harrison, T. Lin, P. Wang, Mechanisms of SARS-CoV-2 transmission and
pathogenesis, Trends Immunol. 41 (12) (2020) 11001115.
[2] A. Maxmen, Omicron blindspots: why it's hard to track coronavirus variants,
Nature (2021).
[3] P. V'Kovski, A. Kratzel, S. Steiner, H. Stalder, V. Thiel, Coronavirus biology and
replication: implications for SARS-CoV-2, Nat. Rev. Microbiol. 19 (3) (2021)
155170.
[4] D.P. Oran, E.J. Topol, Prevalence of asymptomatic SARS-CoV-2 infection : a
narrative review, Ann. Intern. Med. 173 (5) (2020) 362367.
[5] G. Mansueto, M. Niola, C. Napoli, Can COVID 2019 induce a specic
cardiovascular damage or it exacerbates pre-existing cardiovascular diseases?
Pathol. Res. Pract. 216 (9) (2020) 153086.
[6] S. D'Errico, M. Zanon, M. Montanaro, D. Radaelli, F. Sessa, G. Di Mizio,
A. Montana, S. Corrao, M. Salerno, C. Pomara, More than pneumonia: distinctive
features of SARS-cov-2 infection. From autopsy ndings to clinical implications: a
systematic review, Microorganisms 8 (11) (2020).
[7] A.J. Kucharski, P. Klepac, A.J.K. Conlan, S.M. Kissler, M.L. Tang, H. Fry, J.R. Gog,
W.J. Edmunds, C.C.-w. group, Effectiveness of isolation, testing, contact tracing,
and physical distancing on reducing transmission of SARS-CoV-2 in different
settings: a mathematical modelling study, Lancet Infect. Dis. 20 (10) (2020)
11511160.
[8] J.H. Stone, M.J. Frigault, N.J. Serling-Boyd, A.D. Fernandes, L. Harvey,
A.S. Foulkes, N.K. Horick, B.C. Healy, R. Shah, A.M. Bensaci, A.E. Woolley,
S. Nikiforow, N. Lin, M. Sagar, H. Schrager, D.S. Huckins, M. Axelrod,
M.D. Pincus, J. Fleisher, C.A. Sacks, M. Dougan, C.M. North, Y.D. Halvorsen,
T.K. Thurber, Z. Dagher, A. Scherer, R.S. Wallwork, A.Y. Kim, S. Schoenfeld,
P. Sen, T.G. Neilan, C.A. Perugino, S.H. Unizony, D.S. Collier, M.A. Matza,
J.M. Yinh, K.A. Bowman, E. Meyerowitz, A. Zafar, Z.D. Drobni, M.B. Bolster,
M. Kohler, K.M. D'Silva, J. Dau, M.M. Lockwood, C. Cubbison, B.N. Weber,
M.K. Mansour, B.B.T.T. Investigators, Efcacy of Tocilizumab in patients
hospitalized with covid-19, N. Engl. J. Med. 383 (24) (2020) 23332344.
[9] N. Praschan, S. Josephy-Hernandez, D.D. Kim, M.D. Kritzer, S. Mukerji,
A. Newhouse, M. Pasinski, Z. Chemali, Implications of COVID-19 sequelae for
health-care personnel, Lancet Respir. Med. 9 (3) (2021) 230231.
[10] A.D. Proal, M.B. VanElzakker, Long COVID or post-acute sequelae of COVID-19
(PASC): an overview of biological factors that may contribute to persistent
symptoms, Front. Microbiol. 12 (2021) 698169.
[11] J.K. Logue, N.M. Franko, D.J. McCulloch, D. McDonald, A. Magedson, C.R. Wolf,
H.Y. Chu, Sequelae in adults at 6 Months after COVID-19 infection, JAMA Netw.
Open 4 (2) (2021), e210830.
[12] S.K. Mohanty, A. Satapathy, M.M. Naidu, S. Mukhopadhyay, S. Sharma,
L.M. Barton, E. Stroberg, E.J. Duval, D. Pradhan, A. Tzankov, A.V. Parwani, Severe
acute respiratory syndrome coronavirus-2 (SARS-CoV-2) and coronavirus disease
19 (COVID-19) - anatomic pathology perspective on current knowledge, Diagn.
Pathol. 15 (1) (2020) 103.
[13] A. Maiese, A.C. Manetti, R. La Russa, M. Di Paolo, E. Turillazzi, P. Frati,
V. Fineschi, Autopsy ndings in COVID-19-related deaths: a literature review,
Forensic Sci. Med. Pathol. 17 (2) (2021) 279296.
[14] C. Mondello, S. Roccuzzo, O. Malfa, D. Sapienza, P. Gualniera, E. Ventura
Spagnolo, N. Di Nunno, M. Salerno, C. Pomara, A. Asmundo, Pathological ndings
in COVID-19 as a tool to dene SARS-CoV-2 pathogenesis. A systematic review,
Front. Pharmacol. 12 (2021) 614586.
[15] S. Lucas, Pandemics and pathology: a reection on inuenza, HIV/AIDS and SARS
(COVID-19) pandemic infections, Diagn. Histopathol. 27 (3) (2021) 128133.
[16] S. Satturwar, M. Fowkes, C. Farver, A.M. Wilson, A. Eccher, I. Girolami, E. Pujadas,
C. Bryce, F. Salem, S.M. El Jamal, A. Paniz-Mondol, B. Petersen, R.E. Gordon,
J. Reidy, F. Fraggetta, D.A. Marshall, L. Pantanowitz, Postmortem ndings
associated with SARS-CoV-2: systematic review and meta-analysis, Am. J. Surg.
Pathol. 45 (5) (2021) 587603.
[17] N.K. Jha, S. Ojha, S.K. Jha, H. Dureja, S.K. Singh, S.D. Shukla, D.K. Chellappan,
G. Gupta, S. Bhardwaj, N. Kumar, M. Jeyaraman, R. Jain, S. Muthu, R. Kar,
D. Kumar, V.K. Goswami, J. Ruokolainen, K.K. Kesari, S.K. Singh, K. Dua, Evidence
of coronavirus (CoV) pathogenesis and emerging pathogen SARS-CoV-2 in the
nervous system: a review on neurological impairments and manifestations, J. Mol.
Neurosci. (2021).
[18] M. Gheblawi, K. Wang, A. Viveiros, Q. Nguyen, J.C. Zhong, A.J. Turner,
M.K. Raizada, M.B. Grant, G.Y. Oudit, Angiotensin-converting enzyme 2: SARS-
CoV-2 receptor and regulator of the renin-angiotensin system: celebrating the 20th
anniversary of the discovery of ACE2, Circ. Res. 126 (10) (2020) 14561474.
[19] N.M. Hooper, D.W. Lambert, A.J. Turner, Discovery and characterization of ACE2 -
a 20-year journey of surprises from vasopeptidase to COVID-19, Clin. Sci. (Lond.)
134 (18) (2020) 24892501.
[20] M. Ekholm, T. Kahan, The impact of the renin-angiotensin-aldosterone system on
inammation, coagulation, and atherothrombotic complications, and to
aggravated COVID-19, Front. Pharmacol. 12 (2021) 640185.
[21] S. Beyerstedt, E.B. Casaro, E.B. Rangel, COVID-19: angiotensin-converting enzyme
2 (ACE2) expression and tissue susceptibility to SARS-CoV-2 infection, Eur. J. Clin.
Microbiol. Infect. Dis. 40 (5) (2021) 905919.
[22] I. Hamming, W. Timens, M.L. Bulthuis, A.T. Lely, G. Navis, H. van Goor, Tissue
distribution of ACE2 protein, the functional receptor for SARS coronavirus. A rst
step in understanding SARS pathogenesis, J. Pathol. 203 (2) (2004) 631637.
[23] J. Zhao, H. Zhou, W. Huang, J. Zhou, M. Qiu, Z. Deng, L. Chen, Y. Weng, L. Cai,
Y. Gu, Q. Zheng, Q. Chen, X. Hou, L. Wang, L. Shen, Z. Yang, Cell morphological
analysis of SARS-CoV-2 infection by transmission electron microscopy, J. Thorac.
Dis. 12 (8) (2020) 43684373.
[24] K. Kuba, Y. Imai, S. Rao, H. Gao, F. Guo, B. Guan, Y. Huan, P. Yang, Y. Zhang,
W. Deng, L. Bao, B. Zhang, G. Liu, Z. Wang, M. Chappell, Y. Liu, D. Zheng,
A. Leibbrandt, T. Wada, A.S. Slutsky, D. Liu, C. Qin, C. Jiang, J.M. Penninger,
A crucial role of angiotensin converting enzyme 2 (ACE2) in SARS coronavirus-
induced lung injury, Nat. Med. 11 (8) (2005) 875879.
[25] Z. Zhang, Y. Zheng, Z. Niu, B. Zhang, C. Wang, X. Yao, H. Peng, D.N. Franca,
Y. Wang, Y. Zhu, Y. Su, M. Tang, X. Jiang, H. Ren, M. He, Y. Wang, L. Gao, P. Zhao,
H. Shi, Z. Chen, X. Wang, M. Piacentini, X. Bian, G. Melino, L. Liu, H. Huang,
Q. Sun, SARS-CoV-2 spike protein dictates syncytium-mediated lymphocyte
elimination, Cell Death Differ. 28 (9) (2021) 27652777.
[26] W. Chen, J.Y. Pan, Anatomical and pathological observation and analysis of SARS
and COVID-19: microthrombosis is the main cause of death, Biol. Proced. Online
23 (1) (2021) 4.
[27] K.G. Kyriakoulis, D.G. Kokkinidis, I.A. Kyprianou, C.A. Papanastasiou,
P. Archontakis-Barakakis, I. Doundoulakis, C. Bakoyiannis, G. Giannakoulas,
L. Palaiodimos, Venous thromboembolism in the era of COVID-19, Phlebology 36
(2) (2021) 9199.
[28] R. Vinci, D. Pedicino, F. Andreotti, G. Russo, A. D'Aiello, R. De Cristofaro, F. Crea,
G. Liuzzo, From angiotensin-converting enzyme 2 disruption to
thromboinammatory microvascular disease: a paradigm drawn from COVID-19,
Int. J. Cardiol. 326 (2021) 243247.
[29] S. Ram Kumar Pandian, S. Arunachalam, V. Deepak, S. Kunjiappan, K. Sundar,
Targeting complement cascade: an alternative strategy for COVID-19, 3 Biotech 10
(11) (2020) 479.
[30] S. Zhang, Y. Liu, X. Wang, L. Yang, H. Li, Y. Wang, M. Liu, X. Zhao, Y. Xie, Y. Yang,
S. Zhang, Z. Fan, J. Dong, Z. Yuan, Z. Ding, Y. Zhang, L. Hu, SARS-CoV-2 binds
L. Yao et al. Heliyon 8 (2022) e09302
10
platelet ACE2 to enhance thrombosis in COVID-19, J. Hematol. Oncol. 13 (1)
(2020) 120.
[31] T. Sengupta, R. Majumder, S. Majumder, Role of vitamin D in treating COVID-19-
associated coagulopathy: problems and perspectives, Mol. Cell. Biochem. 476 (6)
(2021) 24212427.
[32] T. Burki, The indirect impact of COVID-19 on women, Lancet Infect. Dis. 20 (8)
(2020) 904905.
[33] I.E. Sama, A. Ravera, B.T. Santema, H. van Goor, J.M. Ter Maaten, J.G.F. Cleland,
M. Rienstra, A.W. Friedrich, N.J. Samani, L.L. Ng, K. Dickstein, C.C. Lang,
G. Filippatos, S.D. Anker, P. Ponikowski, M. Metra, D.J. van Veldhuisen,
A.A. Voors, Circulating plasma concentrations of angiotensin-converting enzyme 2
in men and women with heart failure and effects of renin-angiotensin-aldosterone
inhibitors, Eur. Heart J. 41 (19) (2020) 18101817.
[34] M.C. Gagliardi, P. Tieri, E. Ortona, A. Ruggieri, ACE2 expression and sex disparity
in COVID-19, Cell Death Dis. 6 (2020) 37.
[35] G.M. Bwire, Coronavirus: why men are more vulnerable to covid-19 than women?
SN Compr. Clin. Med. (2020) 13.
[36] S.L. Klein, K.L. Flanagan, Sex differences in immune responses, Nat. Rev.
Immunol. 16 (10) (2016) 626638.
[37] P. Conti, A. Younes, Coronavirus COV-19/SARS-CoV-2 affects women less than
men: clinical response to viral infection, J. Biol. Regul. Homeost. Agents 34 (2)
(2020) 339343.
[38] H. Liu, Z. Wang, H. Sun, T. Teng, Y. Li, X. Zhou, Q. Yang, Thrombosis and
coagulopathy in COVID-19: current understanding and implications for
antithrombotic treatment in patients treated with percutaneous coronary
intervention, Front. Cardiovasc. Med. 7 (2020) 599334.
[39] R. Gan, N.P. Rosoman, D.J.E. Henshaw, E.P. Noble, P. Georgius, N. Sommerfeld,
COVID-19 as a viral functional ACE2 deciency disorder with ACE2 related multi-
organ disease, Med. Hypotheses 144 (2020) 110024.
[40] A. Ghosh, R. Colling, An overview of COVID-19 for diagnostic pathologists:
clinicopathological correlation and diagnostic techniques, Diagn. Histopathol. 26
(11) (2020) 529536.
[41] A. Sette, S. Crotty, Adaptive immunity to SARS-CoV-2 and COVID-19, Cell 184 (4)
(2021) 861880.
[42] S. Si-Mohamed, N. Chebib, M. Sigovan, L. Zumbihl, S. Turquier, S. Boccalini,
L. Boussel, J.F. Mornex, V. Cottin, P. Douek, In vivo demonstration of pulmonary
microvascular involvement in COVID-19 using dual-energy computed
tomography, Eur. Respir. J. 56 (4) (2020).
[43] S.E. Fox, A. Akmatbekov, J.L. Harbert, G. Li, J. Quincy Brown, R.S. Vander Heide,
Pulmonary and cardiac pathology in African American patients with COVID-19: an
autopsy series from New Orleans, Lancet Respir. Med. 8 (7) (2020) 681686.
[44] Z. Chen, E. John Wherry, T cell responses in patients with COVID-19, Nat. Rev.
Immunol. 20 (9) (2020) 529536.
[45] E. Zaboli, H. Majidi, R. Alizadeh-Navaei, A. Hedayatizadeh-Omran, H. Asgarian-
Omran, L. Vahedi Larijani, V. Khodaverdi, O. Amjadi, Lymphopenia and lung
complications in patients with coronavirus disease-2019 (COVID-19): a
retrospective study based on clinical data, J. Med. Virol. 93 (9) (2021)
54255431.
[46] X. Cao, COVID-19: immunopathology and its implications for therapy, Nat. Rev.
Immunol. 20 (5) (2020) 269270.
[47] M. Zheng, Y. Gao, G. Wang, G. Song, S. Liu, D. Sun, Y. Xu, Z. Tian, Functional
exhaustion of antiviral lymphocytes in COVID-19 patients, Cell. Mol. Immunol. 17
(5) (2020) 533535.
[48] V.J. Costela-Ruiz, R. Illescas-Montes, J.M. Puerta-Puerta, C. Ruiz, L. Melguizo-
Rodriguez, SARS-CoV-2 infection: the role of cytokines in COVID-19 disease,
Cytokine Growth Factor Rev. 54 (2020) 6275.
[49] S. Bhaskar, A. Sinha, M. Banach, S. Mittoo, R. Weissert, J.S. Kass, S. Rajagopal,
A.R. Pai, S. Kutty, Cytokine storm in COVID-19-immunopathological mechanisms,
clinical considerations, and therapeutic approaches: the REPROGRAM consortium
position paper, Front. Immunol. 11 (2020) 1648.
[50] D. Ragab, H. Salah Eldin, M. Taeimah, R. Khattab, R. Salem, The COVID-19
cytokine storm; what we know so far, Front. Immunol. 11 (2020) 1446.
[51] Y. Que, C. Hu, K. Wan, P. Hu, R. Wang, J. Luo, T. Li, R. Ping, Q. Hu, Y. Sun, X. Wu,
L. Tu, Y. Du, C. Chang, G. Xu, Cytokine release syndrome in COVID-19: a major
mechanism of morbidity and mortality, Int. Rev. Immunol. (2021) 114.
[52] M.A. Haimei, Pathogenesis and treatment strategies of COVID-19-related
hypercoagulant and thrombotic complications, Clin. Appl. Thromb. Hemost. 26
(2020), 1076029620944497.
[53] P.G. Gibson, L. Qin, S.H. Puah, COVID-19 acute respiratory distress syndrome
(ARDS): clinical features and differences from typical pre-COVID-19 ARDS, Med.
J. Aust. 213 (2) (2020) 5456 e1.
[54] S.S. Hasan, T. Capstick, R. Ahmed, C.S. Kow, F. Mazhar, H.A. Merchant,
S.T.R. Zaidi, Mortality in COVID-19 patients with acute respiratory distress
syndrome and corticosteroids use: a systematic review and meta-analysis, Expet
Rev. Respir. Med. 14 (11) (2020) 11491163.
[55] C. Wu, X. Chen, Y. Cai, J. Xia, X. Zhou, S. Xu, H. Huang, L. Zhang, X. Zhou, C. Du,
Y. Zhang, J. Song, S. Wang, Y. Chao, Z. Yang, J. Xu, X. Zhou, D. Chen, W. Xiong,
L. Xu, F. Zhou, J. Jiang, C. Bai, J. Zheng, Y. Song, Risk factors associated with
acute respiratory distress syndrome and death in patients with coronavirus disease
2019 pneumonia in wuhan, China, JAMA Intern. Med. 180 (7) (2020) 934943.
[56] E. Mortaz, P. Tabarsi, M. Varahram, G. Folkerts, I.M. Adcock, The immune
response and immunopathology of COVID-19, Front. Immunol. 11 (2020) 2037.
[57] C. Huang, Y. Wang, X. Li, L. Ren, J. Zhao, Y. Hu, L. Zhang, G. Fan, J. Xu, X. Gu,
Z. Cheng, T. Yu, J. Xia, Y. Wei, W. Wu, X. Xie, W. Yin, H. Li, M. Liu, Y. Xiao,
H. Gao, L. Guo, J. Xie, G. Wang, R. Jiang, Z. Gao, Q. Jin, J. Wang, B. Cao, Clinical
features of patients infected with 2019 novel coronavirus in Wuhan, China, Lancet
395 (10223) (2020) 497506.
[58] D.M. Del Valle, S. Kim-Schulze, H.H. Huang, N.D. Beckmann, S. Nirenberg,
B. Wang, Y. Lavin, T.H. Swartz, D. Madduri, A. Stock, T.U. Marron, H. Xie,
M. Patel, K. Tuballes, O. Van Oekelen, A. Rahman, P. Kovatch, J.A. Aberg,
E. Schadt, S. Jagannath, M. Mazumdar, A.W. Charney, A. Firpo-Betancourt,
D.R. Mendu, J. Jhang, D. Reich, K. Sigel, C. Cordon-Cardo, M. Feldmann,
S. Parekh, M. Merad, S. Gnjatic, An inammatory cytokine signature predicts
COVID-19 severity and survival, Nat. Med. 26 (10) (2020) 16361643.
[59] Y. Chi, Y. Ge, B. Wu, W. Zhang, T. Wu, T. Wen, J. Liu, X. Guo, C. Huang, Y. Jiao,
F. Zhu, B. Zhu, L. Cui, Serum cytokine and chemokine prole in relation to the
severity of coronavirus disease 2019 in China, J. Infect. Dis. 222 (5) (2020)
746754.
[60] E. Mortaz, A. Bassir, N. Dalil Roofchayee, N.K. Dezfuli, H. Jamaati, P. Tabarsi,
A. Moniri, M. Rezaei, P. Mehrian, M. Varahram, M. Marjani, S. Mumby,
I.M. Adcock, Serum cytokine levels of COVID-19 patients after 7 days of treatment
with Favipiravir or Kaletra, Int. Immunopharm. 93 (2021) 107407.
[61] H. Han, Q. Ma, C. Li, R. Liu, L. Zhao, W. Wang, P. Zhang, X. Liu, G. Gao, F. Liu,
Y. Jiang, X. Cheng, C. Zhu, Y. Xia, Proling serum cytokines in COVID-19 patients
reveals IL-6 and IL-10 are disease severity predictors, Emerg. Microb. Infect. 9 (1)
(2020) 11231130.
[62] Z.S. Xu, T. Shu, L. Kang, D. Wu, X. Zhou, B.W. Liao, X.L. Sun, X. Zhou, Y.Y. Wang,
Temporal proling of plasma cytokines, chemokines and growth factors from
mild, severe and fatal COVID-19 patients, Signal Transduct. Targeted Ther. 5 (1)
(2020) 100.
[63] C. Lucas, P. Wong, J. Klein, T.B.R. Castro, J. Silva, M. Sundaram, M.K. Ellingson,
T. Mao, J.E. Oh, B. Israelow, T. Takahashi, M. Tokuyama, P. Lu, A. Venkataraman,
A. Park, S. Mohanty, H. Wang, A.L. Wyllie, C.B.F. Vogels, R. Earnest, S. Lapidus,
I.M. Ott, A.J. Moore, M.C. Muenker, J.B. Fournier, M. Campbell, C.D. Odio,
A. Casanovas-Massana, I.T. Yale, R. Herbst, A.C. Shaw, R. Medzhitov, W.L. Schulz,
N.D. Grubaugh, C. Dela Cruz, S. Farhadian, A.I. Ko, S.B. Omer, A. Iwasaki,
Longitudinal analyses reveal immunological misring in severe COVID-19, Nature
584 (7821) (2020) 463469.
[64] A.K. Azkur, M. Akdis, D. Azkur, M. Sokolowska, W. van de Veen, M.C. Bruggen,
L. O'Mahony, Y. Gao, K. Nadeau, C.A. Akdis, Immune response to SARS-CoV -2 and
mechanisms of immunopathological changes in COVID-19, Allergy 75 (7) (2020)
15641581.
[65] V. Deshmukh, R. Motwani, A. Kumar, C. Kumari, K. Raza, Histopathological
observations in COVID-19: a systematic review, J. Clin. Pathol. 74 (2) (2021)
7683.
[66] S. Iida, T.S.T. Arashiro, Insights into pathology and pathogenesis of coronavirus
disease 2019 from a histopathological and immunological perspective, JMA J 4
(3) (2021).
[67] F. Wang, R.M. Kream, G.B. Stefano, Long-term respiratory and neurological
sequelae of COVID-19, Med. Sci. Mon. Int. Med. J. Exp. Clin. Res. 26 (2020),
e928996.
[68] X. Cui, W. Chen, H. Zhou, Y. Gong, B. Zhu, X. Lv, H. Guo, J. Duan, J. Zhou,
E. Marcon, H. Ma, Pulmonary edema in COVID-19 patients: mechanisms and
treatment potential, Front. Pharmacol. 12 (2021) 664349.
[69] C. Suess, R. Hausmann, Gross and histopathological pulmonary ndings in a
COVID-19 associated death during self-isolation, Int. J. Leg. Med. 134 (4) (2020)
12851290.
[70] L. Szekely, B. Bozoky, M. Bendek, M. Ostad, P. Lavignasse, L. Haag, J. Wu, X. Jing,
S. Gupta, E. Saccon, A. Sonnerborg, Y. Cao, M. Bjornstedt, A. Szakos, Pulmonary
stromal expansion and intra-alveolar coagulation are primary causes of COVID-19
death, Heliyon 7 (5) (2021), e07134.
[71] E. Youd, L. Moore, COVID-19 autopsy in people who died in community settings:
the rst series, J. Clin. Pathol. 73 (12) (2020) 840844.
[72] S. Caramaschi, M.E. Kapp, S.E. Miller, R. Eisenberg, J. Johnson, G. Epperly,
A. Maiorana, G. Silvestri, G.A. Giannico, Histopathological ndings and
clinicopathologic correlation in COVID-19: a systematic review, Mod. Pathol. 34
(2021).
[73] M. Ackermann, S.E. Verleden, M. Kuehnel, A. Haverich, T. Welte, F. Laenger,
A. Vanstapel, C. Werlein, H. Stark, A. Tzankov, W.W. Li, V.W. Li, S.J. Mentzer,
D. Jonigk, Pulmonary vascular endothelialitis, thrombosis, and angiogenesis in
covid-19, N. Engl. J. Med. 383 (2) (2020) 120128.
[74] S.F. Lax, K. Skok, P.M. Zechner, L. Setaffy, H.H. Kessler, N. Kaufmann, K. Vander,
N. Cokic, U. Maierhofer, U. Bargfrieder, M. Trauner, Systemic consequences
and clinical aspects of SARS-CoV-2 infection, Pathologe 42 (2) (2021) 155163.
[75] N. Mackman, S. Antoniak, A.S. Wolberg, R. Kasthuri, N.S. Key, Coagulation
abnormalities and thrombosis in patients infected with SARS-CoV-2 and
other pandemic viruses, Arterioscler. Thromb. Vasc. Biol. 40 (9) (2020)
20332044.
[76] Z. Wu, Q. Zhang, G. Ye, H. Zhang, B.C. Heng, Y. Fei, B. Zhao, J. Zhou, Structural
and physiological changes of the human body upon SARS-CoV-2 infection,
J. Zhejiang Univ. - Sci. B 22 (4) (2021) 310317.
[77] H. Bosmuller, M. Matter, F. Fend, A. Tzankov, The pulmonary pathology of
COVID-19, Virchows Arch. 478 (1) (2021) 137150.
[78] P. Pandey, S. Agarwal, Rajkumar, lung pathology in COVID-19: a systematic
review, Int. J. Appl. Basic Med. Res. 10 (4) (2020) 226233.
[79] D.K. Rai, P. Sharma, R. Kumar, Post covid 19 pulmonary brosis. Is it real threat?
Indian J. Tubercul. 68 (3) (2021) 330333.
[80] A.S. Ojo, S.A. Balogun, O.T. Williams, O.S. Ojo, Pulmonary brosis in COVID-19
survivors: predictive factors and risk reduction strategies, Pulm Med. 2020 (2020)
6175964.
L. Yao et al. Heliyon 8 (2022) e09302
11
[81] C. Zhang, Z. Wu, J.W. Li, K. Tan, W. Yang, H. Zhao, G.Q. Wang, Discharge may not
be the end of treatment: pay attention to pulmonary brosis caused by severe
COVID-19, J. Med. Virol. 93 (3) (2021) 13781386.
[82] P. Gabarre, G. Dumas, T. Dupont, M. Darmon, E. Azoulay, L. Zafrani, Acute kidney
injury in critically ill patients with COVID-19, Intensive Care Med. 46 (7) (2020)
13391348.
[83] M.K. Nadim, L.G. Forni, R.L. Mehta, M.J. Connor Jr., K.D. Liu, M. Ostermann,
T. Rimmele, A. Zarbock, S. Bell, A. Bihorac, V. Cantaluppi, E. Hoste, F. Husain-
Syed, M.J. Germain, S.L. Goldstein, S. Gupta, M. Joannidis, K. Kashani,
J.L. Koyner, M. Legrand, N. Lumlertgul, S. Mohan, N. Pannu, Z. Peng, X.L. Perez-
Fernandez, P. Pickkers, J. Prowle, T. Reis, N. Srisawat, A. Tolwani, A. Vijayan,
G. Villa, L. Yang, C. Ronco, J.A. Kellum, COVID-19-associated acute kidney injury:
consensus report of the 25th Acute Disease Quality Initiative (ADQI) Workgroup,
Nat. Rev. Nephrol. 16 (12) (2020) 747764.
[84] B. Diao, C. Wang, R. Wang, Z. Feng, J. Zhang, H. Yang, Y. Tan, H. Wang, C. Wang,
L. Liu, Y. Liu, Y. Liu, G. Wang, Z. Yuan, X. Hou, L. Ren, Y. Wu, Y. Chen, Human
kidney is a target for novel severe acute respiratory syndrome coronavirus 2
infection, Nat. Commun. 12 (1) (2021) 2506.
[85] N.V. Kolhe, R.J. Fluck, N.M. Selby, M.W. Taal, Acute kidney injury associated with
COVID-19: a retrospective cohort study, PLoS Med. 17 (10) (2020), e1003406.
[86] Q. Yang, X. Yang, Incidence and risk factors of kidney impairment on patients with
COVID-19: a meta-analysis of 10180 patients, PLoS One 15 (11) (2020),
e0241953.
[87] R.T. Gansevoort, L.B. Hilbrands, CKD is a key risk factor for COVID-19 mortality,
Nat. Rev. Nephrol. 16 (12) (2020) 705706.
[88] K. Amann, P. Boor, T. Wiech, J. Singh, E. Vonbrunn, A. Knoll, M. Hermann,
M. Buttner-Herold, C. Daniel, A. Hartmann, COVID-19 effects on the kidney,
Pathologe 42 (2021).
[89] M.B. Le Stang, J. Desenclos, M. Flamant, B.G. Chousterman, N. Tabibzadeh, The
good treatment, the bad virus, and the ugly inammation: pathophysiology of
kidney involvement during COVID-19, Front. Physiol. 12 (2021) 613019.
[90] M. Wang, H. Xiong, H. Chen, Q. Li, X.Z. Ruan, Renal injury by SARS-CoV-2
infection: a systematic review, Kidney Dis. 7 (2) (2021) 100110.
[91] J.M. Lacy, E.G. Brooks, J. Akers, D. Armstrong, L. Decker, A. Gonzalez,
W. Humphrey, R. Mayer, M. Miller, C. Perez, J.A.R. Arango, L. Sathyavagiswaran,
W. Stroh, S. Utley, COVID-19: postmortem diagnostic and biosafety
considerations, Am. J. Forensic Med. Pathol 41 (3) (2020) 143151.
[92] I. Gagliardi, G. Patella, A. Michael, R. Serra, M. Provenzano, M. Andreucci,
COVID-19 and the kidney: from epidemiology to clinical practice, J. Clin. Med. 9
(8) (2020).
[93] P. Sharma, J.H. Ng, V. Bijol, K.D. Jhaveri, R. Wanchoo, Pathology of COVID-19-
associated acute kidney injury, Clin. Kidney J. 14 (Suppl 1) (2021) i30i39.
[94] A.R. Ahmed, C.A. Ebad, S. Stoneman, M.M. Satti, P.J. Conlon, Kidney injury in
COVID-19, World J. Nephrol. 9 (2) (2020) 1832.
[95] V.K. Paliwal, R.K. Garg, A. Gupta, N. Tejan, Neuromuscular presentations in
patients with COVID-19, Neurol. Sci. 41 (11) (2020) 30393056.
[96] A. Gasmi, T. Tippairote, P.K. Mujawdiya, A. Gasmi Benahmed, A. Menzel,
M. Dadar, G. Bjorklund, Neurological involvements of SARS-CoV2 infection, Mol.
Neurobiol. 58 (3) (2021) 944949.
[97] Y.C. Li, Y. Zhang, B.H. Tan, What can cerebrospinal uid testing and brain
autopsies tell us about viral neuroinvasion of SARS-CoV-2, J. Med. Virol. 93 (7)
(2021) 42474257.
[98] S. Kumar, A. Veldhuis, T. Malhotra, Neuropsychiatric and cognitive sequelae of
COVID-19, Front. Psychol. 12 (2021) 577529.
[99] S. Al-Sarraj, C. Troakes, B. Hanley, M. Osborn, M.P. Richardson, M. Hotopf,
E. Bullmore, I.P. Everall, Invited Review: the spectrum of neuropathology in
COVID-19, Neuropathol. Appl. Neurobiol. 47 (1) (2021) 316.
[100] S.J. Yong, Persistent brainstem dysfunction in long-COVID: a hypothesis, ACS
Chem. Neurosci. 12 (4) (2021) 573580.
[101] J. Davies, H.S. Randeva, K. Chatha, M. Hall, D.A. Spandidos, E. Karteris, I. Kyrou,
Neuropilin1 as a new potential SARSCoV2 infection mediator implicated in the
neurologic features and central nervous system involvement of COVID19, Mol.
Med. Rep. 22 (5) (2020) 42214226.
[102] T. Umapathi, W.M.J. Quek, J.M. Yen, H.S.W. Khin, Y.Y. Mah, C.Y.J. Chan,
L.M. Ling, W.Y. Yu, Encephalopathy in COVID-19 patients; viral, parainfectious, or
both? eNeurologicalSci 21 (2020) 100275.
[103] S.E. Fox, R.S.V. Heide, COVID-19: the heart of the matter-pathological changes
and a proposed mechanism, J. Cardiovasc. Pharmacol. Therapeut. 26 (3) (2021)
217224.
[104] M.K. Halushka, R.S. Vander Heide, Myocarditis is rare in COVID-19 autopsies:
cardiovascular ndings across 277 postmortem examinations, Cardiovasc. Pathol.
50 (2021) 107300.
[105] A. Roshdy, S. Zaher, H. Fayed, J.G. Coghlan, COVID-19 and the heart: a systematic
review of cardiac autopsies, Front. Cardiovasc. Med. 7 (2020) 626975.
[106] B.T. Bradley, H. Maioli, R. Johnston, I. Chaudhry, S.L. Fink, H. Xu, B. Najaan,
G. Deutsch, J.M. Lacy, T. Williams, N. Yarid, D.A. Marshall, Histopathology and
ultrastructural ndings of fatal COVID-19 infections in Washington State: a case
series, Lancet 396 (10247) (2020) 320332.
[107] F. Zhou, T. Yu, R. Du, G. Fan, Y. Liu, Z. Liu, J. Xiang, Y. Wang, B. Song, X. Gu,
L. Guan, Y. Wei, H. Li, X. Wu, J. Xu, S. Tu, Y. Zhang, H. Chen, B. Cao, Clinical
course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan,
China: a retrospective cohort study, Lancet 395 (10229) (2020) 10541062.
[108] S. Patibandla, K. Gupta, K. Alsayouri, Cardiac Enzymes, StatPearls, Treasure Island
(FL), 2021.
[109] B. Alexander, A. Baranchuk, COVID-19 and myocardial injury, CMAJ (Can. Med.
Assoc. J.) 192 (28) (2020) E812E813.
[110] M. Imazio, K. Klingel, I. Kindermann, A. Brucato, F.G. De Rosa, Y. Adler, G.M. De
Ferrari, COVID-19 pandemic and troponin: indirect myocardial injury, myocardial
inammation or myocarditis? Heart 106 (15) (2020) 11271131.
[111] O.A. Centurion, K.E. Scavenius, L.B. Garcia, J.M. Torales, L.M. Mino, Potential
mechanisms of cardiac injury and common pathways of inammation in patients
with COVID-19, Crit. Pathw. Cardiol. 20 (1) (2021) 4452.
[112] M.J. Lizardo-Thiebaud, E. Cervantes-Alvarez, N. Limon-de la Rosa, F. Tejeda-
Dominguez, M. Palacios-Jimenez, O. Mendez-Guerrero, M. Delaye-Martinez,
F. Rodriguez-Alvarez, B. Romero-Morales, W.H. Liu, C.A. Huang, D. Kershenobich,
N. Navarro-Alvarez, Direct or collateral liver damage in SARS-CoV-2-infected
patients, Semin. Liver Dis. 40 (3) (2020) 321330.
[113] Y. Wang, S. Liu, H. Liu, W. Li, F. Lin, L. Jiang, X. Li, P. Xu, L. Zhang, L. Zhao,
Y. Cao, J. Kang, J. Yang, L. Li, X. Liu, Y. Li, R. Nie, J. Mu, F. Lu, S. Zhao, J. Lu,
J. Zhao, SARS-CoV-2 infection of the liver directly contributes to hepatic
impairment in patients with COVID-19, J. Hepatol. 73 (4) (2020) 807816.
[114] F. Salamanna, M. Maglio, M.P. Landini, M. Fini, Body localization of ACE-2: on the
trail of the keyhole of SARS-CoV-2, Front. Med. 7 (2020) 594495.
[115] T. Marjot, G.J. Webb, A.S.t. Barritt, A.M. Moon, Z. Stamataki, V.W. Wong,
E. Barnes, COVID-19 and liver disease: mechanistic and clinical perspectives, Nat.
Rev. Gastroenterol. Hepatol. 18 (5) (2021) 348364.
[116] L.A. Diaz, F. Idalsoaga, M. Cannistra, R. Candia, D. Cabrera, F. Barrera, A. Soza,
R. Graham, A. Riquelme, M. Arrese, M.D. Leise, J.P. Arab, High prevalence of
hepatic steatosis and vascular thrombosis in COVID-19: a systematic review and
meta-analysis of autopsy data, World J. Gastroenterol. 26 (48) (2020) 76937706.
[117] Y. Li, S.Y. Xiao, Hepatic involvement in COVID-19 patients: pathology,
pathogenesis, and clinical implications, J. Med. Virol. 92 (9) (2020) 14911494.
[118] L. Scappaticcio, F. Pitoia, K. Esposito, A. Piccardo, P. Trimboli, Impact of COVID-
19 on the thyroid gland: an update, Rev. Endocr. Metab. Disord. 22 (2020).
[119] P. Trimboli, C. Cappelli, L. Croce, L. Scappaticcio, L. Chiovato, M. Rotondi, COVID-
19-Associated subacute thyroiditis: evidence-based data from a systematic review,
Front. Endocrinol. 12 (2021) 707726.
[120] A.K. Murugan, A.S. Alzahrani, SARS-CoV-2 plays a pivotal role in inducing
hyperthyroidism of Graves' disease, Endocrine 73 (2) (2021) 243254.
[121] A. Lania, M.T. Sandri, M. Cellini, M. Mirani, E. Lavezzi, G. Mazziotti,
Thyrotoxicosis in patients with COVID-19: the THYRCOV study, Eur. J.
Endocrinol. 183 (4) (2020) 381387.
[122] J. Malik, A. Malik, M. Javaid, T. Zahid, U. Ishaq, M. Shoaib, Thyroid function
analysis in COVID-19: a retrospective study from a single center, PLoS One 16 (3)
(2021), e0249421.
[123] P. Trimboli, C. Camponovo, L. Scappaticcio, G. Bellastella, A. Piccardo,
M. Rotondi, Thyroid sequelae of COVID-19: a systematic review of reviews, Rev.
Endocr. Metab. Disord. 22 (2) (2021) 485491.
[124] R. Mao, Y. Qiu, J.S. He, J.Y. Tan, X.H. Li, J. Liang, J. Shen, L.R. Zhu, Y. Chen,
M. Iacucci, S.C. Ng, S. Ghosh, M.H. Chen, Manifestations and prognosis of
gastrointestinal and liver involvement in patients with COVID-19: a systematic
review and meta-analysis, Lancet Gastroenterol. Hepatol. 5 (7) (2020) 667678.
[125] M. Sen, S.G. Honavar, N. Sharma, M.S. Sachdev, COVID-19 and eye: a review of
ophthalmic manifestations of COVID-19, Indian J. Ophthalmol. 69 (3) (2021)
488509.
[126] O.B. Sawant, S. Singh, R.E. Wright 3rd, K.M. Jones, M.S. Titus, E. Dennis, E. Hicks,
P.A. Majmudar, A. Kumar, S.I. Mian, Prevalence of SARS-CoV-2 in human post-
mortem ocular tissues, Ocul. Surf. 19 (2021) 322329.
[127] K. Hu, J. Patel, C. Swiston, B.C. Patel, Ophthalmic Manifestations of Coronavirus
(COVID-19), StatPearls, Treasure Island (FL), 2021.
[128] P. Wu, F. Duan, C. Luo, Q. Liu, X. Qu, L. Liang, K. Wu, Characteristics of ocular
ndings of patients with coronavirus disease 2019 (COVID-19) in hubei province,
China, JAMA Ophthalmol. 138 (5) (2020) 575578.
[129] S.T. Armenti, D. Ballouz, S.I. Mian, Updated safety precautions and guidance on
eye banking procedures during the coronavirus disease 2019 pandemic, Curr.
Opin. Ophthalmol. 32 (4) (2021) 338342.
[130] C. Huang, L. Huang, Y. Wang, X. Li, L. Ren, X. Gu, L. Kang, L. Guo, M. Liu, X. Zhou,
J. Luo, Z. Huang, S. Tu, Y. Zhao, L. Chen, D. Xu, Y. Li, C. Li, L. Peng, Y. Li, W. Xie,
D. Cui, L. Shang, G. Fan, J. Xu, G. Wang, Y. Wang, J. Zhong, C. Wang, J. Wang,
D. Zhang, B. Cao, 6-month consequences of COVID-19 in patients discharged from
hospital: a cohort study, Lancet 397 (10270) (2021) 220232.
[131] S. Lopez-Leon, T. Wegman-Ostrosky, C. Perelman, R. Sepulveda, P.A. Rebolledo,
A. Cuapio, S. Villapol, More than 50 long-term effects of COVID-19: a systematic
review and meta-analysis, Sci. Rep. 11 (1) (2021) 16144.
[132] M. Dres, D. Hajage, S. Lebbah, A. Kimmoun, T. Pham, G. Beduneau, A. Combes,
A. Mercat, B. Guidet, A. Demoule, M. Schmidt, C.-I. investigators, Characteristics,
management, and prognosis of elderly patients with COVID-19 admitted in the
ICU during the rst wave: insights from the COVID-ICU study : prognosis of
COVID-19 elderly critically ill patients in the ICU, Ann. Intensive Care 11 (1)
(2021) 77.
[133] J.J. Chang, M.S. Lin, T.H. Chen, D.Y. Chen, S.W. Chen, J.T. Hsu, P.C. Wang,
Y.S. Lin, Heart failure and mortality of adult survivors from acute myocarditis
requiring intensive care treatment - a nationwide cohort study, Int. J. Med. Sci. 14
(12) (2017) 12411250.
[134] A. Maury, A. Lyoubi, N. Peiffer-Smadja, T. de Broucker, E. Meppiel, Neurological
manifestations associated with SARS-CoV-2 and other coronaviruses: a narrative
review for clinicians, Rev. Neurol. (Paris) 177 (1-2) (2021) 5164.
[135] N. Ritschel, H. Radbruch, C. Herden, N. Schneider, C. Dittmayer, J. Franz,
C. Thomas, G. Silva Boos, A. Pagenstecher, W. Schulz-Schaeffer, C. Stadelmann,
M. Glatzel, F.L. Heppner, J. Weis, K. Sohrabi, A. Schanzer, A. Nemeth, T. Acker,
D.G.-T. CNS-COVID, D.P.-N.R.b. COVID, COVID-19 and the central and peripheral
nervous system, Pathologe 42 (2) (2021) 172182.
L. Yao et al. Heliyon 8 (2022) e09302
12
[136] A.C. Yang, F. Kern, P.M. Losada, M.R. Agam, C.A. Maat, G.P. Schmartz,
T. Fehlmann, J.A. Stein, N. Schaum, D.P. Lee, K. Calcuttawala, R.T. Vest,
D. Berdnik, N. Lu, O. Hahn, D. Gate, M.W. McNerney, D. Channappa, I. Cobos,
N. Ludwig, W.J. Schulz-Schaeffer, A. Keller, T. Wyss-Coray, Dysregulation of brain
and choroid plexus cell types in severe COVID-19, Nature (2021).
[137] T. Minami, Y. Iwata, T. Wada, Renal complications in coronavirus disease 2019: a
systematic review, Inamm. Regen. 40 (1) (2020) 31.
[138] B. Singh, P. Kaur, R.R. Reid, Case reports: rhabdomyolysis associated with COVID-
19, Am. Fam. Phys. 102 (11) (2020) 645648.
[139] A. Nalbandian, K. Sehgal, A. Gupta, M.V. Madhavan, C. McGroder, J.S. Stevens,
J.R. Cook, A.S. Nordvig, D. Shalev, T.S. Sehrawat, N. Ahluwalia, B. Bikdeli,
D. Dietz, C. Der-Nigoghossian, N. Liyanage-Don, G.F. Rosner, E.J. Bernstein,
S. Mohan, A.A. Beckley, D.S. Seres, T.K. Choueiri, N. Uriel, J.C. Ausiello, D. Accili,
D.E. Freedberg, M. Baldwin, A. Schwartz, D. Brodie, C.K. Garcia, M.S.V. Elkind,
J.M. Connors, J.P. Bilezikian, D.W. Landry, E.Y. Wan, Post-acute COVID-19
syndrome, Nat. Med. 27 (4) (2021) 601615.
[140] S.E. Daugherty, Y. Guo, K. Heath, M.C. Dasmarinas, K.G. Jubilo, J. Samranvedhya,
M. Lipsitch, K. Cohen, Risk of clinical sequelae after the acute phase of SARS-CoV-
2 infection: retrospective cohort study, BMJ 373 (2021) n1098.
[141] W. Sungnak, N. Huang, C. Becavin, M. Berg, R. Queen, M. Litvinukova,
C. Talavera-Lopez, H. Maatz, D. Reichart, F. Sampaziotis, K.B. Worlock,
M. Yoshida, J.L. Barnes, H.C.A.L.B. Network, SARS-CoV-2 entry factors are highly
expressed in nasal epithelial cells together with innate immune genes, Nat. Med.
26 (5) (2020) 681687.
[142] O.J. McElvaney, N.L. McEvoy, O.F. McElvaney, T.P. Carroll, M.P. Murphy,
D.M. Dunlea, O. Ni Choileain, J. Clarke, E. O'Connor, G. Hogan, D. Ryan,
I. Sulaiman, C. Gunaratnam, P. Branagan, M.E. O'Brien, R.K. Morgan,
R.W. Costello, K. Hurley, S. Walsh, E. de Barra, C. McNally, S. McConkey,
F. Boland, S. Galvin, F. Kiernan, J. O'Rourke, R. Dwyer, M. Power, P. Geoghegan,
C. Larkin, R.A. O'Leary, J. Freeman, A. Gaffney, B. Marsh, G.F. Curley,
N.G. McElvaney, Characterization of the inammatory response to severe COVID-
19 illness, Am. J. Respir. Crit. Care Med. 202 (6) (2020) 812821.
[143] N. Matalon, S. Dorman-Ilan, I. Hasson-Ohayon, N. Hertz-Palmor, S. Shani, D. Basel,
R. Gross, W. Chen, A. Abramovich, A. Afek, A. Ziv, Y. Kreiss, I.M. Pessach,
D. Gothelf, Trajectories of post-traumatic stress symptoms, anxiety, and depression
in hospitalized COVID-19 patients: a one-month follow-up, J. Psychosom. Res. 143
(2021) 110399.
[144] M.B. Malas, I.N. Naazie, N. Elsayed, A. Mathlouthi, R. Marmor, B. Clary,
Thromboembolism risk of COVID-19 is high and associated with a higher risk of
mortality: a systematic review and meta-analysis, EClinicalMedicine 29 (2020)
100639.
[145] F. Wu, S. Zhao, B. Yu, Y.M. Chen, W. Wang, Z.G. Song, Y. Hu, Z.W. Tao, J.H. Tian,
Y.Y. Pei, M.L. Yuan, Y.L. Zhang, F.H. Dai, Y. Liu, Q.M. Wang, J.J. Zheng, L. Xu,
E.C. Holmes, Y.Z. Zhang, A new coronavirus associated with human respiratory
disease in China, Nature 579 (7798) (2020) 265269.
[146] C. Aslan, S.H. Kiaie, N.M. Zolbanin, P. Lotnejad, R. Ramezani, F. Kashanchi,
R. Jafari, Exosomes for mRNA delivery: a novel biotherapeutic strategy with
hurdles and hope, BMC Biotechnol. 21 (1) (2021) 20.
[147] E. Callaway, Had COVID? You'll probably make antibodies for a lifetime, Nature
(2021).
[148] J.S. Turner, W. Kim, E. Kalaidina, C.W. Goss, A.M. Rauseo, A.J. Schmitz,
L. Hansen, A. Haile, M.K. Klebert, I. Pusic, J.A. O'Halloran, R.M. Presti,
A.H. Ellebedy, SARS-CoV-2 infection induces long-lived bone marrow plasma cells
in humans, Nature 595 (7867) (2021) 421425.
[149] J.S. Turner, W. Kim, E. Kalaidina, C.W. Goss, A.M. Rauseo, A.J. Schmitz,
L. Hansen, A. Haile, M.K. Klebert, I. Pusic, J.A. O'Halloran, R.M. Presti,
A.H. Ellebedy, SARS-CoV-2 infection induces long-lived bone marrow plasma cells
in humans, Res. Sq. (2020).
[150] J.M. Dan, J. Mateus, Y. Kato, K.M. Hastie, E.D. Yu, C.E. Faliti, A. Grifoni,
S.I. Ramirez, S. Haupt, A. Frazier, C. Nakao, V. Rayaprolu, S.A. Rawlings, B. Peters,
F. Krammer, V. Simon, E.O. Saphire, D.M. Smith, D. Weiskopf, A. Sette, S. Crotty,
Immunological memory to SARS-CoV-2 assessed for up to 8 months after infection,
Science 371 (6529) (2021).
[151] A. Ansari, R. Arya, S. Sachan, S.N. Jha, A. Kalia, A. Lall, A. Sette, A. Grifoni,
D. Weiskopf, P. Coshic, A. Sharma, N. Gupta, Immune memory in mild COVID-19
patients and unexposed donors reveals persistent T cell responses after SARS-CoV-
2 infection, Front. Immunol. 12 (2021) 636768.
[152] J.P. Townsend, H.B. Hassler, Z. Wang, S. Miura, J. Singh, S. Kumar, N.H. Ruddle,
A.P. Galvani, A. Dornburg, The durability of immunity against reinfection by
SARS-CoV-2: a comparative evolutionary study, Lancet Microbe. (2021).
[153] N. Ortega, M. Ribes, M. Vidal, R. Rubio, R. Aguilar, S. Williams, D. Barrios,
S. Alonso, P. Hernandez-Luis, R.A. Mitchell, C. Jairoce, A. Cruz, A. Jimenez,
R. Santano, S. Mendez, M. Lamoglia, N. Rosell, A. Llupia, L. Puyol, J. Chi,
N.R. Melero, D. Parras, P. Serra, E. Pradenas, B. Trinite, J. Blanco, A. Mayor,
S. Barroso, P. Varela, A. Vilella, A. Trilla, P. Santamaria, C. Carolis, M. Tortajada,
L. Izquierdo, A. Angulo, P. Engel, A.L. Garcia-Basteiro, G. Moncunill, C. Dobano,
Seven-month kinetics of SARS-CoV-2 antibodies and role of pre-existing antibodies
to human coronaviruses, Nat. Commun. 12 (1) (2021) 4740.
[154] W. Doerer, Adenoviral vector DNA- and SARS-CoV-2 mRNA-based covid-19
vaccines: possible integration into the human genome - are adenoviral genes
expressed in vector-based vaccines? Virus Res. 302 (2021) 198466.
[155] B. Yan, S. Chakravorty, C. Mirabelli, L. Wang, J.L. Trujillo-Ochoa, D. Chauss,
D. Kumar, M.S. Lionakis, M.R. Olson, C.E. Wobus, B. Afzali, M. Kazemian, Host-
virus chimeric events in SARS-CoV-2-infected cells are infrequent and artifactual,
J. Virol. 95 (15) (2021), e0029421.
[156] L. Zhang, A. Richards, M.I. Barrasa, S.H. Hughes, R.A. Young, R. Jaenisch, Reverse-
transcribed SARS-CoV-2 RNA can integrate into the genome of cultured human
cells and can be expressed in patient-derived tissues, Proc. Natl. Acad. Sci. U. S. A.
118 (21) (2021).
[157] R. Parry, R.J. Gifford, S. Lytras, S.C. Ray, L.J.M. Coin, No evidence of SARS-CoV-2
reverse transcription and integration as the origin of chimeric transcripts in
patient tissues, Proc. Natl. Acad. Sci. U. S. A. 118 (33) (2021).
[158] T. Thanh Le, Z. Andreadakis, A. Kumar, R. Gomez Roman, S. Tollefsen, M. Saville,
S. Mayhew, The COVID-19 vaccine development landscape, Nat. Rev. Drug Discov.
19 (5) (2020) 305306.
[159] M. Becker, A. Dulovic, D. Junker, N. Ruetalo, P.D. Kaiser, Y.T. Pinilla, C. Heinzel,
J. Haering, B. Traenkle, T.R. Wagner, M. Layer, M. Mehrlaender, V. Mirakaj,
J. Held, H. Planatscher, K. Schenke-Layland, G. Krause, M. Strengert, T. Bakchoul,
K. Althaus, R. Fendel, A. Kreidenweiss, M. Koeppen, U. Rothbauer, M. Schindler,
N. Schneiderhan-Marra, Immune response to SARS-CoV-2 variants of concern in
vaccinated individuals, Nat. Commun. 12 (1) (2021) 3109.
[160] C. Alteri, V. Cento, A. Piralla, V. Costabile, M. Tallarita, L. Colagrossi, S. Renica,
F. Giardina, F. Novazzi, S. Gaiarsa, E. Matarazzo, M. Antonello, C. Vismara,
R. Fumagalli, O.M. Epis, M. Puoti, C.F. Perno, F. Baldanti, Genomic
epidemiology of SARS-CoV-2 reveals multiple lineages and early spread of SARS-
CoV-2 infections in Lombardy, Italy, Nat. Commun. 12 (1) (2021) 434.
L. Yao et al. Heliyon 8 (2022) e09302
13
... Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a novel coronavirus, causing highly contagious coronavirus disease (COVID) [1,2]. Since it emerged in December 2019 in Wuhan, China [3,4], there have been over 605 million confirmed cases of COVID-19, and over 6.4 million deaths worldwide as of 12 September 2022. Epidemiological studies show that cases in other parts of the world had direct travel history from affected areas or with exposure or contact to a known case [5,6]. ...
... The COVID-19 pandemic worldwide directly or indirectly affects mental health. Individuals infected by COVID-19 experience post-traumatic stress syndrome, depression, and anxiety [4]. Some may suffer multiregional cognitive dysfunction, persistent headache, and extreme fatigue with sleep disorders [4]. ...
... Individuals infected by COVID-19 experience post-traumatic stress syndrome, depression, and anxiety [4]. Some may suffer multiregional cognitive dysfunction, persistent headache, and extreme fatigue with sleep disorders [4]. Fear to contract highly contagious COVID-19 with severe sequalae and complications afflicts more individuals. ...
Article
Full-text available
Background: To explore the association between the self-reported health status, depressive tendency, psychological capital, and self-acceptance of college students in China during the COVID-19 pandemic. Methods: Using the online survey platform "questionnaire star", a two-phase cross-sectional study was conducted on a total number of 1438 undergraduates with informed consents. The questionnaires of Self-Rated Health Measurement Scale (SRHMS), the Center for Epidemiological Studies-Depression Scale (CES-D), Psychological Capital Questionnaire (PCQ-24), and self-acceptance questionnaire were administered to each participant. Results: Male college students had significantly higher depressive tendency scores than female (17.59 vs. 15.82) (p < 0.01). College students having no siblings had significantly higher psychological capital scores than those having siblings (108.63 vs. 105.60) (p < 0.05). Exercise had significantly positive associations with self-rated health, psychological capital, and self-acceptance scores, while online time per day had significantly negative associations. Multivariate analysis showed that the interaction between depressive tendency, psychological capital, and self-acceptance was statistically significant (β = 0.004, p = 0.013 for phase 1 and β = 0.002, p = 0.025 for phase 2) in health status with depressive tendency ranking the top (β = -0.54 for phase 1 and -0.41 for phase 2, p < 0.001). Mediation analysis showed that psychological capital and self-acceptance modified the association of depressive tendency with health status. Conclusion: Physical exercise is beneficial to both physical and psychological health. Depressive tendency is the main risk factor that associates with self-rated health. Regardless of depressive tendency level, high psychological capital and self-acceptance could improve college students' health.
... Another example of alteration in the NS in patients with SARS-CoV-2 is detailed in the article by Padrón González and Dorta Contreras, where it is stated that the virus not only attacks the CNS but also the peripheral NS, producing manifestations that can be mild and transitory. However, complications of direct cytopathic damage or complications secondary to the effects of the inflammatory response can arise [21]. In line with the study of biomarkers and the pathogenesis of SARS-CoV-2, Royer Rodriguez-Guzmán, in a letter to the editor in the journal Rev Neurol. ...
... Direct and indirect effects on mental health, including not only neurological events but also neuropsychiatric and neuropsychological manifestations, either early on or in the long term, have been associated with COVID-19, and it is believed that in the future these disorders, in particular, neurodegenerative disorders, will increase around the world. Diverse theories, some of which were mentioned previously in this manuscript, point to brain damage as a consequence of direct neuronal infection by SARS-CoV-2 that contributes to neurodegenerative disorders in the long term [21]. However, the exact mechanism has not been found yet. ...
Article
Full-text available
Neuropsychiatric manifestations of viral infections (both per se and secondary to the neuroinflammatory reaction of the host) are mainly attributed to immunological reactions, so many aspects of their pathogenesis are still nuclear. Some novel therapeutic strategies are progressively emerging in which a vaccination may be having a particular impact on recovery and reduction of death. In this context, it is accepted that the SARS-CoV-2 virus is profoundly neurotropic and neuroinvasive, with various effects on the nervous system, although there is no complete understanding of the mechanism of neuroinvasion, brain injury, or short- or long-term neuropsychiatric sequelae. Therefore, it is necessary to understand the post-infectious manifestations of COVID-19 to guide the management of neuropsychiatric diseases. Thus, based on different research groups focused on this field, in this manuscript we summarize papers on COVID-19 and the nervous system (NS) published in a series of articles by Cuban authors. This review focuses on cognitive and affective emotional states, pathogenesis, biomarkers, clinical manifestations, and intervention strategies.
... However, as vaccines have been distributed globally at an unprecedented rate, there have been more and more reports of serious adverse events after vaccination. In particular, cases of vaccine-induced thrombotic and ischemic events such as immune thrombotic thrombocytopenia (VITT) have been recorded since February 2021, primarily after receiving the ChAdOx1 nCoV-19 vaccine (one of many adenovirus vectorbased vaccines) [4,5,[7][8][9][10][11][12][13][14][15][16][17]. VITT is caused by immunoglobulin G molecules that recognize platelet factor 4 (PF4) bound to platelets, which eventually causes platelet activation and the stimulation of the coagulation system; these antibodies are detectable through a PF4 enzyme-linked immunosorbent assay (ELISA) [8,[18][19][20][21]. ...
... The case definition criteria for vaccine-induced immune thrombocytopenia and thrombosis, as decided in a study published by Pavord S. et al. [23], and also discussed in other studies [13,14,16,17], are found in Table 2. For a definite VITT diagnosis, a patient should meet all five of the following criteria: (1) the onset of symptoms 5-30 days after vaccination against SARS-CoV-2 (or less than 42 days in patients with isolated deep-vein thrombosis or pulmonary embolism), (2) the presence of thrombosis, (3) thrombocytope-Vaccines 2022, 10, 1950 6 of 10 nia (PLT < 150,000 per cubic millimeter), (4) D-dimer levels of greater than 4000 FEU, and (5) positive anti-PF4 antibodies from ELISA. ...
Article
Full-text available
COVID-19 has significantly affected public health, social life, and economies worldwide. The only effective way to combat the pandemic is through vaccines. Although the vaccines have been in use for some time, safety concerns have still been raised. The most typical adverse effects of receiving a COVID-19 vaccine are localized reactions near the injection site, followed by general physical symptoms such as headaches, fatigue, muscle pain, and fever. Additionally, some people may experience VITT (vaccine-induced immune thrombotic thrombocytopenia), a rare side effect after vaccination. We present the case of a 60-year-old female patient that developed VITT-like symptoms with spleno-portal thrombosis and intestinal ischemia two weeks after the administration of the Ad26.COV2-S vaccine. Surgical treatment consisted of extensive bowel resection with end jejunostomy and feeding ileostomy. Two weeks after the first operation, a duodenal-ileal anastomosis was performed. The patient was discharged five weeks after the onset of the symptoms. Although some rare adverse effects are associated with the SARS-CoV-2 vaccines, the risk of hospitalization from these harmful effects is lower than the risk of hospitalization from COVID-19. Therefore, recognizing VITT is significant for ensuring the early treatment of clots and proper follow-up.
... There appears to be some correlation between the ability of SARS-CoV-2 to form syncytia and the severity of the illness [13,64], but the underlying mechanism for this is not known. Several potential links have been examined. ...
Article
SARS-CoV-2 has the ability to form large multi-nucleated cells known as syncytia. Little is known about how syncytia affect the dynamics of the infection or severity of the disease. In this manuscript, we extend a mathematical model of cell–cell fusion assays to estimate both the syncytia formation rate and the average duration of the fusion phase for five strains of SARS-CoV-2. We find that the original Wuhan strain has the slowest rate of syncytia formation (6.4 × 10−4 ∕h), but takes only 4.0 h to complete the fusion process, while the Alpha strain has the fastest rate of syncytia formation (0.36 /h), but takes 7.6 h to complete the fusion process. The Beta strain also has a fairly fast syncytia formation rate (9.7 × 10−2 ∕h), and takes the longest to complete fusion (8.4 h). The D614G strain has a fairly slow syncytia formation rate (2.8 × 10−3 ∕h), but completes fusion in 4.0 h. Finally, the Delta strain is in the middle with a syncytia formation rate of 3.2 × 10−2 ∕h and a fusing time of 6.1 h. We note that for these SARS-CoV-2 strains, there appears to be a tradeoff between the ease of forming syncytia and the speed at which they complete the fusion process.
... The heart muscle is damaged by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus and its immunopathological cardiac inflammation consequences (3). It is associated with several direct or indirect cardiovascular complications, such as myocardial damage, myocarditis, heart failure (HF), arrhythmia, and venous thromboembolism (4)(5)(6). ...
Article
Full-text available
Background: Respiratory infections are one of the most common comorbidities identified in hospitalized patients. The coronavirus disease 2019 (COVID-19) pandemic greatly impacted healthcare systems, including acute cardiac services. Aim: This study aimed to describe the echocardiographic findings of patients with COVID-19 infections and their correlations with inflammatory biomarkers, disease severity, and clinical outcomes. Methods: This observational study was conducted between June 2021 and July 2022. The analysis included all patients diagnosed with COVID-19 who had transthoracic echocardiographic (TTE) scans within 72 h of admission. Results: The enrolled patients had a mean age of 55.6 ± 14.7 years, and 66.1% were male. Of the 490 enrolled patients, 203 (41.4%) were admitted to the intensive care unit (ICU). Pre-ICU TTE findings showed significantly higher incidence right ventricular dysfunction (28 [13.8%] vs. 23 [8.0%]; P = 0.04) and left ventricular (LV) regional wall motion abnormalities (55 [27.1%] vs. 29 [10.1%]; p < 0.001) in ICU patients compared to non-ICU patients. In-hospital mortality was 11 (2.2%), all deaths of ICU patients. The most sensitive predictors of ICU admission (p < 0.05): cardiac troponin I level (area under the curve [AUC] = 0.733), followed by hs-CRP (AUC = 0.620), creatine kinase-MB (AUC = 0.617), D-dimer (AUC = 0.599), and lactate dehydrogenase (AUC = 0.567). Binary logistic regression showed that reduced LV ejection fraction (LVEF), elevated pulmonary artery systolic pressure, and dilated right ventricle were echocardiographic predictors of poor outcomes (p < 0.05). Conclusion: Echocardiography is a valuable tool in assessing admitted patients with COVID-19. Lower LVEF, pulmonary hypertension, higher D-dimer, C-reactive protein, and B-type natriuretic peptide levels were predictors of poor outcomes.
... [41][42][43] Typically, individuals who are more symptomatic with a particular illness develop a stronger immune response to fight the illness and recover. 38,44 This is well established and these individuals have long been targeted as a source of CP. 45,46 Thus, older donors who had perhaps experienced enhanced immune responses (both in initial titers and in longevity of response) to SARS-CoV-2 than their younger counterparts may have had significantly higher levels anti-SARS-CoV-2. Additionally, immune response is known to wane over time; however, the anti-SARS-CoV-2 antibody response is particularly fast to wane. ...
Article
Background: COVID-19 convalescent plasma (CCP) was approved under emergency authorization to treat critically ill patients with COVID-19 in the United States in 2020. We explored the demographics of donors contributing plasma for a hyperimmune, plasma-derived therapy to evaluate factors that may be associated with anti-SARS-CoV-2 antibody response variability and, subsequently, antibody titers. Study design: An electronic search of CCP donors was performed across 282 US plasma donation centers. Donations were screened for nucleocapsid protein-binding-IgG using the Abbott SARS-CoV-2 IgG assay. Results: Overall, 52 240 donors donated 418 046 units of CCP. Donors were of various ethnicities: 43% Caucasian, 34% Hispanic, 17% African American, 2% Native American, 1% Asian, and 3% other. Females had higher initial mean anti-SARS-CoV-2 antibody titers but an overall faster rate of decline (P < .0001). Initial antibody titers increased with age: individuals aged 55 to 66 years had elevated anti-SARS-CoV-2 titers for longer periods compared with other ages (P = .0004). African American donors had the lowest initial antibody titers but a slower rate of decline (P < .0001), while Caucasian (P = .0088) and Hispanic (P = .0193) groups had the fastest rates of decline. Most donor antibody levels decreased below the inclusion criteria (≥1.50) within 30 to 100 days of first donation, but donation frequency did not appear to be associated with rate of decline. Conclusion: Several factors may be associated with anti-SARS-CoV-2 antibody response including donor age and sex. Evaluating these factors during development of future hyperimmune globulin products may help generation of therapies with optimal efficacy.
Article
Full-text available
Background Among the most consequential unknowns of the devastating COVID-19 pandemic are the durability of immunity and time to likely reinfection. There are limited direct data on SARS-CoV-2 long-term immune responses and reinfection. The aim of this study is to use data on the durability of immunity among evolutionarily close coronavirus relatives of SARS-CoV-2 to estimate times to reinfection by a comparative evolutionary analysis of related viruses SARS-CoV, MERS-CoV, human coronavirus (HCoV)-229E, HCoV-OC43, and HCoV-NL63. Methods We conducted phylogenetic analyses of the S, M, and ORF1b genes to reconstruct a maximum-likelihood molecular phylogeny of human-infecting coronaviruses. This phylogeny enabled comparative analyses of peak-normalised nucleocapsid protein, spike protein, and whole-virus lysate IgG antibody optical density levels, in conjunction with reinfection data on endemic human-infecting coronaviruses. We performed ancestral and descendent states analyses to estimate the expected declines in antibody levels over time, the probabilities of reinfection based on antibody level, and the anticipated times to reinfection after recovery under conditions of endemic transmission for SARS-CoV-2, as well as the other human-infecting coronaviruses. Findings We obtained antibody optical density data for six human-infecting coronaviruses, extending from 128 days to 28 years after infection between 1984 and 2020. These data provided a means to estimate profiles of the typical antibody decline and probabilities of reinfection over time under endemic conditions. Reinfection by SARS-CoV-2 under endemic conditions would likely occur between 3 months and 5·1 years after peak antibody response, with a median of 16 months. This protection is less than half the duration revealed for the endemic coronaviruses circulating among humans (5–95% quantiles 15 months to 10 years for HCoV-OC43, 31 months to 12 years for HCoV-NL63, and 16 months to 12 years for HCoV-229E). For SARS-CoV, the 5–95% quantiles were 4 months to 6 years, whereas the 95% quantiles for MERS-CoV were inconsistent by dataset. Interpretation The timeframe for reinfection is fundamental to numerous aspects of public health decision making. As the COVID-19 pandemic continues, reinfection is likely to become increasingly common. Maintaining public health measures that curb transmission—including among individuals who were previously infected with SARS-CoV-2—coupled with persistent efforts to accelerate vaccination worldwide is critical to the prevention of COVID-19 morbidity and mortality. Funding US National Science Foundation.
Article
Full-text available
Subacute thyroiditis (SAT) is a thyroid disease of viral or post-viral origin. Whether SAT represents a complication of coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is still unclear. Our aim was to systematically review the literature to 1) explore the size of the literature about SAT in COVID-19 and 2) evaluate the clinical characteristics of SAT. PubMed/MEDLINE, Embase, and Scopus were searched until April 20, 2021. Original papers, case reports, and case series reporting SAT in COVID-19 patients were included. Authors and their country, journal, year of publication, COVID-19 and SAT clinical presentation, thyroid function, therapy, and follow-up data were extracted. Nineteen papers (17 case reports and 2 case series) were included, describing 27 patients, 74.1% females, aged 18 to 69 years. COVID-19 was diagnosed by nasopharyngeal swab in 66.7% cases and required hospitalization in 11.1%. In 83.3% cases, SAT occurred after COVID-19. Neck pain was present in 92.6% cases and fever in 74.1%. Median TSH, fT3, and fT4 were 0.01 mU/l, 10.79 pmol/l, and 27.2 pmol/l, respectively. C-reactive-protein and erythrocyte sedimentation rate were elevated in 96% of cases. Typical ultrasonographic characteristics of SAT were observed in 83.3% of cases. Steroids were the most frequent SAT therapy. Complete remission of SAT was recorded in most cases. In conclusion, the size and quality of published data of SAT in COVID-19 patients are poor, with only case reports and case series being available. SAT clinical presentation in COVID-19 patients seems to be similar to what is generally expected.
Article
Full-text available
Since the first case of COVID-19 was reported in Wuhan, China, in December 2019, the SARS-CoV-2 epidemic has spread all over the world and has become a significant public health issue. The development of treatments for COVID-19 is currently in progress; however, their effects remain limited, and the development of more effective therapeutics is desired. Thus, sufficient understanding of the pathophysiology of COVID-19 is essential to develop effective therapeutics for this disease. Pathological analyses in particular play an important role to demonstrate the causal link between an infectious disease and the pathogen and elucidate the mechanism of pathogenesis. As per pathological analyses to date, respiratory organs are identified as the major affected organs in most COVID-19 cases; also, various lesions were noted in other organs. Further, there have been increasing reports that show that the immune responses of the host contribute to the deterioration of the pathological condition of COVID-19, and a novel concept of MIS-C/MIS-A is also being established. Thus, in this article, we have provided an overview of the pathology of COVID-19 from a histopathological and immunological perspective focusing on the mechanisms of COVID-19 pathogenesis.
Article
Full-text available
COVID-19 can involve persistence, sequelae, and other medical complications that last weeks to months after initial recovery. This systematic review and meta-analysis aims to identify studies assessing the long-term effects of COVID-19. LitCOVID and Embase were searched to identify articles with original data published before the 1st of January 2021, with a minimum of 100 patients. For effects reported in two or more studies, meta-analyses using a random-effects model were performed using the MetaXL software to estimate the pooled prevalence with 95% CI. PRISMA guidelines were followed. A total of 18,251 publications were identified, of which 15 met the inclusion criteria. The prevalence of 55 long-term effects was estimated, 21 meta-analyses were performed, and 47,910 patients were included (age 17–87 years). The included studies defined long-COVID as ranging from 14 to 110 days post-viral infection. It was estimated that 80% of the infected patients with SARS-CoV-2 developed one or more long-term symptoms. The five most common symptoms were fatigue (58%), headache (44%), attention disorder (27%), hair loss (25%), and dyspnea (24%). Multi-disciplinary teams are crucial to developing preventive measures, rehabilitation techniques, and clinical management strategies with whole-patient perspectives designed to address long COVID-19 care.
Article
Full-text available
Unraveling the long-term kinetics of antibodies to SARS-CoV-2 and the individual characteristics influencing it, including the impact of pre-existing antibodies to human coronaviruses causing common cold (HCoVs), is essential to understand protective immunity to COVID-19 and devise effective surveillance strategies. IgM, IgA and IgG levels against six SARS-CoV-2 antigens and the nucleocapsid antigen of the four HCoV (229E, NL63, OC43 and HKU1) were quantified by Luminex, and antibody neutralization capacity was assessed by flow cytometry, in a cohort of health care workers followed up to 7 months ( N = 578). Seroprevalence increases over time from 13.5% (month 0) and 15.6% (month 1) to 16.4% (month 6). Levels of antibodies, including those with neutralizing capacity, are stable over time, except IgG to nucleocapsid antigen and IgM levels that wane. After the peak response, anti-spike antibody levels increase from ~150 days post-symptom onset in all individuals (73% for IgG), in the absence of any evidence of re-exposure. IgG and IgA to HCoV are significantly higher in asymptomatic than symptomatic seropositive individuals. Thus, pre-existing cross-reactive HCoVs antibodies could have a protective effect against SARS-CoV-2 infection and COVID-19 disease.
Article
Full-text available
There is interest in understanding the mechanisms that underlie reports that patients infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) remain PCR positive many weeks after initial infection. The recent paper by Zhang et al. (1) suggests a potential explanation of this phenomenon by claiming that SARS-CoV-2 RNA can integrate into the genome of infected human cells. The authors also reanalyze RNA se-quencing (RNA-seq) data and report that SARS-CoV-2− host chimeric reads are present in cells and patient tissues. Given the potential implications of this research on the long-term impacts of COVID-19, we feel that it's necessary to scrutinize the evidence presented. To determine whether SARS-CoV-2 RNA might be retrotranscribed and integrated into the genome, the authors conducted a proof-of-principle experiment where human lung cells (Calu3) and kidney cells overexpress-ing class I transposable elements and wild type (HEK293T-L1/HEK293T) were infected with SARS-CoV-2 and subjected to high-throughput DNA sequencing (1). The very low frequency of identified chimeric events (Table 1) suggests that SARS-CoV-2 integration into the host genome is unlikely. Given that the HEK293T-L1 model increases detection of "rare integration events" and L1 can retrotranspose any poly-adenylated cellular RNA (2) and "insertions" are found preferentially in protein-coding exons, a bias unknown to L1 endonuclease insertions (3), these findings are likely spurious. Additionally, 2 of the identified 61 chime-ric nanopore genomic DNA (gDNA) reads contain human DNA from separate chromosomes (chr1,chr22 and chr18,chrX, respectively), suggesting a portion of chime-ric gDNA nanopore reads have arisen due to infrequent technical artifacts, such as base-calling software not recognizing an open-pore state between distinct molecules. Further to this, the authors reanalyzed published sequencing data and identified SARS-CoV-2 and human host chimeric reads in vitro and in patient RNA-seq (1). They show that the fraction of human-viral chimeric reads derived from negative-sense SARS-CoV-2 RNAs is higher in patients than that observed in vitro. These data are presented as evidence of SARS-CoV-2 integration and transcription. We question the presented data as evidence for this phenomenon for several key reasons. Firstly, chimeric virus−host reads are often reported in RNA-seq (4), including SARS-CoV-2 (5). This is partly due to complementary DNA fusions introduced during the reverse transcription step of library preparation (6). Secondly, larger pools of negative-sense SARS-CoV-2 chimeric reads may be due to differences in RNA extraction, library preparation, and sequencing platform. Given that negative-sense RNA is formed during the replication of SARS-CoV-2 and a template for messenger RNA production, significant variation of negative-sense reads is expected in patient RNA samples. Finally, there is no evidence of coronaviruses ever having integrated into the germ-line of host species, as might be expected if retro-transcription and integration occurs in nature, as systematic screening of >750 animal species failed to identify any coronavirus-derived endogenous viral elements (7).
Article
Full-text available
The novel virus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused a pandemic of coronavirus disease 2019 (COVID-19). Across the globe, a subset of patients who sustain an acute SARS-CoV-2 infection are developing a wide range of persistent symptoms that do not resolve over the course of many months. These patients are being given the diagnosis Long COVID or Post-acute sequelae of COVID-19 (PASC). It is likely that individual patients with a PASC diagnosis have different underlying biological factors driving their symptoms, none of which are mutually exclusive. This paper details mechanisms by which RNA viruses beyond just SARS-CoV-2 have be connected to long-term health consequences. It also reviews literature on acute COVID-19 and other virus-initiated chronic syndromes such as post-Ebola syndrome or myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) to discuss different scenarios for PASC symptom development. Potential contributors to PASC symptoms include consequences from acute SARS-CoV-2 injury to one or multiple organs, persistent reservoirs of SARS-CoV-2 in certain tissues, re-activation of neurotrophic pathogens such as herpesviruses under conditions of COVID-19 immune dysregulation, SARS-CoV-2 interactions with host microbiome/virome communities, clotting/coagulation issues, dysfunctional brainstem/vagus nerve signaling, ongoing activity of primed immune cells, and autoimmunity due to molecular mimicry between pathogen and host proteins. The individualized nature of PASC symptoms suggests that different therapeutic approaches may be required to best manage care for specific patients with the diagnosis.
Article
Full-text available
Though SARS-CoV-2 primarily targets the respiratory system, patients and survivors can suffer neurological symptoms1–3. Yet, an unbiased understanding of the cellular and molecular processes affected in the brains of COVID-19 patients is still missing. Here, we profile 65,309 single-nucleus transcriptomes from 30 frontal cortex and choroid plexus samples across 14 control (including 1 terminal influenza) and 8 COVID-19 patients. While a systematic analysis yields no molecular traces of SARS-CoV-2 in the brain, we observe broad cellular perturbations which predict that choroid plexus barrier cells sense and relay peripheral inflammation into the brain and show that peripheral T cells infiltrate the parenchyma. We discover COVID-19 disease-associated microglia and astrocyte subpopulations that share features with pathological cell states reported in human neurodegenerative disease4–6. Synaptic signaling of upper-layer excitatory neurons—evolutionarily expanded in humans7 and linked to cognitive function8—are preferentially affected in COVID-19. Across cell types, COVID-19 perturbations overlap with those in chronic brain disorders and reside in genetic variants associated with cognition, schizophrenia, and depression. Our findings and public dataset provide a molecular framework to understand COVID-19 related neurological disease observed now and which may emerge later.
Article
Full-text available
Atherosclerosis is considered a disease caused by a chronic inflammation, associated with endothelial dysfunction, and several mediators of inflammation are up-regulated in subjects with atherosclerotic disease. Healthy, intact endothelium exhibits an antithrombotic, protective surface between the vascular lumen and vascular smooth muscle cells in the vessel wall. Oxidative stress is an imbalance between anti- and prooxidants, with a subsequent increase of reactive oxygen species, leading to tissue damage. The renin-angiotensin-aldosterone system is of vital importance in the pathobiology of vascular disease. Convincing data indicate that angiotensin II accelerates hypertension and augments the production of reactive oxygen species. This leads to the generation of a proinflammatory phenotype in human endothelial and vascular smooth muscle cells by the up-regulation of adhesion molecules, chemokines and cytokines. In addition, angiotensin II also seems to increase thrombin generation, possibly via a direct impact on tissue factor. However, the mechanism of cross-talk between inflammation and haemostasis can also contribute to prothrombotic states in inflammatory environments. Thus, blocking of the renin-angiotensin-aldosterone system might be an approach to reduce both inflammatory and thrombotic complications in high-risk patients. During COVID-19, the renin-angiotensin-aldosterone system may be activated. The levels of angiotensin II could contribute to the ongoing inflammation, which might result in a cytokine storm, a complication that significantly impairs prognosis. At the outbreak of COVID-19 concerns were raised about the use of angiotensin converting enzyme inhibitors and angiotensin receptor blocker drugs in patients with COVID-19 and hypertension or other cardiovascular comorbidities. However, the present evidence is in favor of continuing to use of these drugs. Based on experimental evidence, blocking the renin-angiotensin-aldosterone system might even exert a potentially protective influence in the setting of COVID-19.
Article
Researchers are rapidly sequencing the genomes of virus samples worldwide, but shortcomings in the global surveillance system make the task a challenge. Researchers are rapidly sequencing the genomes of virus samples worldwide, but shortcomings in the global surveillance system make the task a challenge.