You are on page 1of 27

Journal Pre-proof

IL-6: relevance for immunopathology of SARS-CoV-2

E.O. Gubernatorova, E.A. Gorshkova, A.I. Polinova, M.D. Drutskaya

PII: S1359-6101(20)30108-8
DOI: https://doi.org/10.1016/j.cytogfr.2020.05.009
Reference: CGFR 1153

To appear in: Cytokine and Growth Factor Reviews

Received Date: 16 May 2020


Accepted Date: 17 May 2020

Please cite this article as: Gubernatorova EO, Gorshkova EA, Polinova AI, Drutskaya MD,
IL-6: relevance for immunopathology of SARS-CoV-2, Cytokine and Growth Factor Reviews
(2020), doi: https://doi.org/10.1016/j.cytogfr.2020.05.009

This is a PDF file of an article that has undergone enhancements after acceptance, such as
the addition of a cover page and metadata, and formatting for readability, but it is not yet the
definitive version of record. This version will undergo additional copyediting, typesetting and
review before it is published in its final form, but we are providing this version to give early
visibility of the article. Please note that, during the production process, errors may be
discovered which could affect the content, and all legal disclaimers that apply to the journal
pertain.

© 2020 Published by Elsevier.


IL-6: relevance for immunopathology of SARS-CoV-2
Gubernatorova E.O.a,b,*, Gorshkova E.A.a,b, Polinova A.I.a,b, Drutskaya M.D.a,**
a
Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt
Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
b
Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
Corresponding authors at: Center for Precision Genome Editing and Genetic Technologies for
Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow
119991, Vavilova str. 32, Russia
* ekaterina.gubernatorova412@gmail.com
** marinadru@gmail.com

Highlights

 COVID-19 severe outcomes is associated with the acute respiratory distress syndrome
and cytokine release syndrome with elevated IL-6 as a relevant predicting parameter.
 Chronic inflammation, associated with elevated inflammatory serum cytokines, including
IL-6, may exacerbate SARS-CoV-2 induced pathology.

of
 IL-6 is a promising therapeutic target for the anti-cytokine therapy of SARS-CoV-2
severe cases.

ro
Abstract
COVID-19 mortality is strongly associated with the development of severe pneumonia and acute
respiratory distress syndrome with the worst outcome resulting in cytokine release syndrome
-p
and multiorgan failure. It is becoming critically important to identify at the early stage of the
infection those patients who are prone to develop the most adverse effects. Elevated systemic
interleukin-6 levels in patients with COVID-19 are considered as a relevant parameter in
re
predicting most severe course of disease and the need for intensive care. This review discusses
the mechanisms by which IL-6 may possibly contribute to disease exacerbation and the
potential of therapeutic approaches based on anti-IL-6 biologics.
lP

Keywords: IL-6, SARS-CoV-2, cytokine release syndrome, anti-cytokine therapeutics.

Introduction
na

At the end of 2019 and the beginning of 2020, first, China and then the rest of the world have
been confronted with an outbreak of a new airborne transmitted coronavirus disease 2019
(COVID-19), a viral infection caused by severe acute respiratory syndrome coronavirus 2
(SARS-CoV-2). Just a few months later, there are already millions infected worldwide and with
ur

borders closed and strict travel bans imposed local transmission patterns are in place in most
countries [1]. COVID-19 mortality rate appears to be in the range of 3,4-5,5% [2], which is
significantly higher than that for seasonal flu caused by the influenza virus (1%), but lower than
Jo

for both severe acute respiratory syndrome (SARS) (15%) [3] and Middle East respiratory
syndrome (MERS) (34%) [4]. At the same time, the COVID-19 transmission rate is much greater
than those of MERS or SARS, and poses a real threat to life for a large number of people
worldwide, especially, in older patients with other co-morbidities [5]. Thus, in the absence of
effective vaccines and treatments, early detection and the ability to define prognostic
parameters predicting the course of the disease appear to be of particular interest.

COVID-19 pathogenesis and IL-6


Coronaviruses represent a large family of zoonotic respiratory viruses [6] that can cause both
seasonal cold symptoms and fatal respiratory failure associated with severe inflammation of
lower respiratory tract [7]. The major clinical symptoms of COVID-19 are similar to those
experienced during a seasonal flu infection and include fever, dry cough, headache, myalgia
and others. Co-morbidities such as cardiovascular diseases, diabetes, respiratory diseases,
hypertension and age may exacerbate the disease manifestations [8, 9]. Both moderate and
severe COVID-19 infections cases may result in pneumonia with fluffy opacities on the chest
computer tomographic scans, lung edema and accumulation of pleural fluid in the lungs [10],
[11]. Severe cases require invasive oxygen supply.
The exact molecular mechanisms of COVID-19-mediated pathogenesis are still under
investigation. However, the lessons learned from SARS-CoV and MERS-CoV infections may
uncover some key features of COVID-19-related pathologies, as well as molecular mediators
and signaling pathways involved. The genetic sequence of SARS-CoV-2 [12], which was
released almost simultaneously with the first reported cases [10] revealed that it belongs to the
β‐coronavirus genus, with 79.0% nucleotide identity to SARS‐CoV and 51.8% identity to MERS-
CoV. Inoculation with SARS-CoV-2 of human airway epithelial cells in vitro causes cytopathic
effects [10] and cessation of the cilium beating of epithelial cells, similar to the cytopathic effect
observed for SARS-CoV infection [13]. The initial step in viral infection was revealed by the
crystal structure of SARS-CoV-2 spike receptor binding domain [14], which, just as SARS-CoV

of
[15], binds to the host cell receptor angiotensin-converting enzyme 2 (ACE2). The presence of
ACE2 on the cell membrane is crucial for virus virulence, as HeLa cells, lacking ACE2, are
resistant to SARS-CoV-2 infection [16, 17]. Structural analysis identified residues in the SARS-

ro
CoV-2 spike receptor binding domain that are critical for ACE2 binding, the majority of which
either are highly conserved or share similar side chain properties with those of SARS-CoV.
These similarities in virus entry between SARS-CoV-2 and SARS-CoV correlate with detectable
-p
cross-neutralizing activity of serum from SARS-CoV-recovered patients [18]. However, no
available monoclonal antibodies targeting SARS-CoV receptor binding domain were able to
prevent SARS-CoV-2 infecting the cells, highlighting unique intrinsic structure features of SARS-
re
CoV-2 S-protein binding domain, including a much higher binding affinity than that of SARS-
CoV S-protein [19]. In line with this, more recent study reports that human recombinant soluble
ACE2 effectively, but not completely prevents SARS-CoV-2 infection, suggesting alternative
lP

mechanisms for viral entry [20].


Surface molecule CD147 (EMMPRIN or Basigin) is being considered as one alternative
pathway for the virus to enter the host cells, since SARS-CoV-2 S-protein can bind CD147 as
suggested by preliminary data [21]. CD147 is expressed on hematopoietic cells, including red
na

blood cells, epithelial and neuronal cells [22]. Basigin is viewed as the molecular gate for
plasmodium P.malariae [23]. Not surprisingly, CD147 is also associated with the HIV1 [24] and
SARS-CoV [25] infections. In both cases, viral protein binds to the CD147 in complex with
cyclophilin A (CyPA). SARS-CoV N-protein conjugates with CyPA inside ACE2-expressing
ur

infected host cells [25]. With this modification, newly assembled viral particles can infect CD147-
positive cells. However, further studies are needed to evaluate possible impact of SARS-CoV-2
interaction with Basigin in the context of COVID-19.
Jo

ACE2, crucial for SARS-CoV-2 virulence, is expressed in the upper and the lower
respiratory tract, most remarkably on lung alveolar epithelial cells, arterial and venous
endothelial cells, as well as enterocytes of the small intestine, epithelial cells in the kidney.
ACE2 expression is also detected in heart, pancreas, testis and brain [26]. Interestingly, ACE2
expression is not the highest in the upper respiratory tract [27, 26], once again, supporting the
hypothesis that increased transmissibility of SARS-CoV-2 as compared to SARS-CoV may be
attributed to yet to be identified co-receptors or auxiliary factors adopted by SARS-CoV-2 [28].
Also, the fact that ACE2 is widely expressed in other tissues and organs explains a broad
spectrum of adverse effects not limited just to the lungs. In addition, it was shown that SARS-
CoV-2 directly infects ACE2-expressing tissue-resident CD169+ macrophages in the spleens
and LNs, causing lymph follicle depletion, splenic nodule atrophy, histiocyte hyperplasia and
lymphocyte reduction [29]. Thus, virus infection of macrophages, firstly, enhances viral spread
and, secondly, triggers destructive events in the immune organs such as spleen and LNs.
Recent data from the experiments using SARS-CoV-2 capsid or live virus infection of cultured T
cell lines (MT-2 and A3.01) provides first evidence that SARS-CoV-2 could also infects T cells.
However, expression level of ACE2 on T cells is low and, thus, CD147 is considered to mediate
SARS-CoV-2 entry into T cells [17].
After virus binds to ACE2 (Fig.1), serine protease, TMPRSS2, cleaves the viral spike
protein. Then another protease, Furin, subsequently releases spike fusion peptide, and the virus
enters the host cell through an endosomal pathway [18, 30, 31]. Following viral replication,
assembly and release, the infected cells may undergo apoptosis or necrosis, triggering the
inflammatory response with production of pro-inflammatory cytokines and activation of
macrophages and Th1 cells, as well as production of IFNγ, IL-17A, IL-21, and IL-22 by
neutrophils, Th17 and CD8+ cells. In turn, SARS-CoV-2 infection of recruited immune cells may
increase their apoptosis and exacerbate lymphocytosis [32, 33], and, finally, may lead in some
patients to life-threatening conditions, such as respiratory distress syndrome, cytokine storm,
and secondary hemophagocytic lymphohistiocytosis.

of
COVID-19 mortality is strongly associated with the development of severe respiratory
distress syndrome, which requires invasive ventilation. It is important to identify those patients
who are most likely to develop a severe form of the disease as early as possible. Based on the

ro
existing statistics, a number of clinical predictors of the disease deterioration and mortality,
including the presence of secondary infections, lymphopenia [34], comorbidities such as
cardiovascular and chronic respiratory diseases, diabetes, hypertension, obesity and cancer
-p
[35], increased cytokine production and serum ferritin [36] have been reported. The other study
showed that the elevated serum creatinine, D-dimers, lactate dehydrogenase, C-reactive
protein, procalcitonin, as well as increased white blood cell counts may indicate impending
re
respiratory failure and the need for invasive oxygen supply [37]. Interestingly, in recently
published reports elevated interleukin-6 (IL-6) level was proposed as a relevant parameter
predicting the unwanted course of the disease and the need for mechanical ventilation [38-40].
lP

These assumptions are consistent with data from a clinical trial in China that demonstrated on a
small number of patients the efficacy of IL-6 neutralization during exacerbation of COVID-19
associated pneumonia [41] (Supplementary Table 1).
IL-6 is a cytokine with pleotropic functions ranging from hematopoiesis and metabolic
na

regulation to inflammation, autoimmunity and acute phase response. IL-6 modulates host
defense through a number of immune stimulating mechanisms: control of monocytes and their
differentiation into macrophages [42], modulation of antigen-dependent B cell differentiation
[43], increased IgG production by B cells [44], and promotion of Th2 response by inhibiting Th1
ur

polarization [45]. Several studies revealed strong correlation between IL-6 levels in the serum
and the upcoming respiratory failure [46-48]. It was shown, that even moderately elevated IL-6
levels above 80 pg/ml were sufficient to identify COVID-19 infected patients with a high risk of
Jo

respiratory failure [37]. Moreover, serum SARS-CoV-2 nucleic acid (RNAaemia), which is
strongly associated with cytokine storm, is closely correlated with extremely high IL-6 serum
levels [47]. It was also suggested that serial measurement of circulating IL-6 might be important
in identifying disease progression or, when evaluated immediately after COVID-19 diagnosis
confirmation, may predict the upcoming respiratory failure or, alternatively, asymptomatic
disease among SARS-CoV-2 infected patients [49].
Therefore, it would be valuable to explore the potential contribution of IL-6 in the events
occurring during SARS-CoV-2 infection. The next section will discuss the mechanisms by which
IL-6 may possibly affect the disease exacerbation and the potential of therapeutic approaches
based on anti-IL-6 biologics.
Controversy concerning IL-6 functions during viral lung infections
IL-6 is one of the key proinflammatory cytokines during infection onset, especially, at the
mucosal sites [50]. However, the impact of IL-6 on the disease outcome may vary significantly
(Fig. 2). On one hand, IL-6-dependent Th17 activation and differentiation are important for
effective neutrophil migration, IL-6 together with IL-15 modulate cytolytic capacity of CD8+ T
cells [51, 52], and, finally, IL-6, as a pyrogenic cytokine, contributes to thermostatic regulation
which is important for effective anti-viral response [53, 54]. Studies, using IL-6-deficient mice,
showed that IL-6 ameliorates acute lung injury in influenza A virus (IAV) infection [55]. IL-6-
deficient mice infected with influenza virus exhibited higher lethality, as well as increased weight
loss and fibroblast accumulation in the lungs associated with virus-induced apoptosis of lung
epithelial cells and neutrophils [56] indicating that IL-6 may promote repair following virus-
induced lung injury [57]. Finally, early IL-6 signaling promotes IL-27-dependent maturation of
Treg cells in the lungs and resolution of viral immunopathology in mouse model of respiratory
syncytial and influenza virus infection [58].
On the other hand, IL-6 has been implicated in the progression of viral infections. In this
regard, IL-6 synergizes with IL-1b and TNF to upregulate trypsin expression, which activates

of
matrix metalloproteinases and causes the breakdown of basal membrane and extracellular
matrix, which, in turn, results in increased tissue permeability and edema [59]. Th1 cell-derived
IFNγ is essential for effective antiviral immune response. However, IL-6 may inhibit Th1

ro
polarization by stimulating CD4+ cells to differentiate into Th2 cells or by suppressing IFNγ
expression [60, 61]. Moreover, IL-6 promotes Th17 cell differentiation and IL-17A secretion,
which, in turn, activates the expression of anti-apoptotic molecules, such as Bcl-XL, favoring
-p
survival of virus-infected cells in the model of persistent viral infection [62]. At the same time, IL-
17 promotes the migration [63], and survival [64] of neutrophils which, in turn, contribute to the
pathogenesis of COVID-19-driven ARDS [65]. Finally, in contrast to influenza virus infection, IL-
re
6-deficient mice infected with murine pneumonia virus are characterized by better survival than
WT mice with decreased lung edema and diminished neutrophil recruitment [66].
Taking into account that in several experimental models of viral lung infections IL-6
lP

demonstrates either pathogenic [66] or protective [55] effects in vivo, the role of this cytokine in
SARS-CoV-2 infection should be carefully evaluated. Moreover, the consequences of IL-6
induction in COVID-19 may vary depending on the infection stage and on the host immune
status.
na

SARS-CoV-2-associated cytokine release syndrome


Patients with the most severe outcome of SARS-CoV-2 displayed high serum levels of
proinflammatory cytokines responsible for cytokine storm induction [36, 67]. Interestingly, the
ur

majority of severe COVID-19 cases with respiratory distress syndrome were associated with
high systemic IL-1b, TNF and IL-6 levels, suggesting a direct connection between
proinflammatory cytokine induction and adverse effects of COVID-19 [34]. As SARS-CoV-2
Jo

shows 79% nucleotide identity to SARS-CoV, major pathogenic mechanisms, including cytokine
storm and extensive lung damage during SARS and COVID-19 infection may be similar.
Cytokine storm is a condition caused by extensive activation of the immune system, and, as a
result, extremely high production of cytokines and chemokines. Since cytokine storm can lead to
multiple organ failure, it is important to understand the mechanisms driving this condition.
In lower respiratory tract SARS-CoV-2 infects predominantly type 2 alveolar
epitheliocytes, the cell type which does not participate in active gas exchange. Following
replication and disengagement of the lung epithelial layer, virus enters the underlying tissues
and infects or is being captured by macrophages, dendritic cells and neutrophils, resulting in
further viral spread [29] (Fig. 3). At the same time, damaged epitheliocytes release danger
molecules which drive the activation of lung epithelium and resident immune cells. Thus, even
at the early onset of viral infection, its penetration of the mucosa to the epithelial layer activates
the innate immune response. Adaptive immunity is also activated during COVID-19, mainly by
antigen presenting dendritic cells, which produce large amounts of cytokines, including IL-6, IL-
1b and TNF and migrate to the regional lymph nodes to present viral antigen to naïve T cells,
pushing their differentiation and migration into the affected tissue [68]. Specific humoral
response represented by viral-specific antibody-producing B cells is also in place [69].
Replication of the virus results in further damage of affected organs together with self-boosting
immune response manifesting to uncontrolled overexpression of inflammatory mediators in
severe cases. Another mechanism driving cytokine storm during viral infections is increased
blood vessel permeability which enables infiltration of effector cells, producing additional
amounts of inflammatory molecules and exacerbating hyper cytokine production [70]. In
addition, leakiness of blood vessels allows virus to spread to other tissues and organs,
compromising their functions. Finally, under persistence of inflammatory factors, an increased
number of inflammatory exudates and erythrocytes enter the alveoli, resulting in lung damage
and respiratory failure syndrome.
As mentioned above, severe COVID-19 induced pneumonia is marked by

of
hyperactivation of the immune system and excessive production of IL-6 [36, 67]. The main
source of pathogenic IL-6 appears to be myeloid cells as their number is significantly increased
among PBMC [69] and in BALF of severe COVID-19 patients [71]. Myeloid cell recruitment and

ro
activation in the lungs is associated with increased mortality and morbidity in SARS-CoV mouse
model [72]. Human primary macrophages and dendritic cells can be infected, but they fail to
support productive replication of SARS-CoV [73, 74], unlike alveocytes, in which virus can
-p
replicate very quickly. At the same time, SARS-CoV and, apparently, SARS-CoV-2-infected
macrophages demonstrate up-regulation of IL-6 production and low expression of interferons
[29, 73]. Interestingly, patients requiring intensive care and invasive lung ventilation display
re
negative correlation between IL-6, TNF and IL-1b concentrations and CD4+ and CD8+ T cell
counts [75], confirming previous studies in animal models, which suggested that cytokine storm
actually dampens adaptive immunity against SARS-CoV infection [76]. Moreover, production of
lP

IFNγ by CD4+ T cells tended to be lower in severe as compared to moderate cases, suggesting
that SARS-CoV-2 infection induces lymphopenia with the suppression of antiviral IFNγ
production [36]. Furthermore, it has been suggested that this pathogenic IL-6 is likely to be
produced by highly inflammatory macrophages [71], but not by alveolar macrophages, which
na

are almost absent in the lung aspirates of severely affected patients [77]. At the same time, it
should be noted that complete ablation of IL-6 at the very early stages of viral infection may
result in depressed Tfh cell maturation and delayed antiviral antibody response [78].
It is known that acute inflammation and cytokine storm, as its extreme manifestation, can
ur

have a significant effect on hemodynamics and also on platelets and erythrocytes. The
dysregulated coagulation cascade and the subsequent formation of intra-alveolar or systemic
fibrin clots are prominent findings in previous zoonotic coronavirus infections (SARS and MERS)
Jo

orchestrated by CRS [79-81]. COVID-19 patients also show significant changes in coagulative
status (i.e. low platelet count, increased D-dimer, fibrinogen levels) and dysfunction of micro-
vessels in pulmonary circulation [82, 83], thus, thrombosis of the pulmonary capillaries is
believed to contribute to rapidly developing hypoxia. The role of IL-6 in inflammatory
hypercoagulation is controversial: IL-6 can increase the rate of fibrotic clot formation in whole
blood, but the effect is less pronounced than it is shown for IL-1 or IL-8 [84]. On the other hand,
IL-6 is important for thrombus formation being a regulator of MMPs and other thrombolysis-
associated genes in macrophages [85].
Other characteristic feature of cytokine release syndrome is change in iron homeostasis
and hyperferritinemia as indicated by high levels of serum ferritin [86]. In COVID-19 patients
ferritin elevation correlated with poor prognosis [36], as it was an indication of hemolytic events
caused by hypercoagulation and hemophagocytosis. In general, inadequate macrophage
activation leads to pathologic phagocytosis of erythrocytes and reutilization of heme iron using
ferritin [87]. In line with this, high systemic ferritin may serve as an indicator of developing
macrophage activation syndrome, rather than a factor driving COVID-19 pathogenesis.
Another key proinflammatory cytokine, TNF, is also released abundantly during cytokine
storm, caused by SARS-CoV-2 infection. Serum TNF levels were found to be elevated in
severely affected COVID-19 patients [34]. Previously, it was shown for SARS-CoV that its spike
protein is able to modulate TNF converting enzyme (TACE)-dependent shedding of the ACE2
ectodomain [88]. TACE expression is coupled to TNF production. Therefore, it is hypothesized
that the use of TNF inhibitors may also show some efficacy in preventing viral entry [89], as well
as in decreasing immunopathology associated mortality, as it was shown in preclinical studies in
mice infected with severe respiratory syncytial virus [90]. Moreover, anti-TNF therapy was
effective in decreasing the IL-6 and IL-1b serum levels in lethal bacteremia, suggesting that
such treatment may provide complex anti-inflammatory effect [91]. Data obtained from COVID-
19 infected patients with inflammatory bowel diseases undergoing anti-TNF therapy support
safety of such intervention in the context of SARS-CoV-2 treatment [92, 93]. These findings

of
argue for a potentially protective effect of TNF inhibition in COVID-19 and further studies are
needed to address this possibility.

ro
ACE2 downregulation may induce IL-6 production in Angiotensin II-dependent manner
Hypertension has been suggested as a risk factor for severe COVID-19 outcomes [94]. Early
data from the literature already assumed an association between blood pressure and circulating
-p
IL-6 levels in otherwise healthy individuals [95]. Moreover, several studies reported an
increased IL-6 expression in patients with hypertension [96, 97]. IL-6-transgenic mice,
characterized by elevated plasma IL-6 levels, displayed pulmonary hypertension [98, 99]. Taken
re
together, this data suggest that IL-6 might be connected with increased risk of respiratory failure
in hypertensive patients with COVID-19.
One of the mechanisms regulating blood pressure and electrolyte balance is
lP

angiotensin-renin system. It is comprised of two pathways, which are normally functioning in the
state of dynamic equilibrium: ACE2-Angiotensin II–Angiotensin 1 Receptor axis, activating
vasoconstriction and inflammatory response, and ACE2-Angiotensin 1–7-Mas receptor axis,
supporting vasodilation and suppression of inflammation (Fig. 4, A) [100]. Changes in renin-
na

angiotensin system activity are related to the pathogenesis of hypertension and of inflammatory
lung diseases, in particular, acute lung injury caused by viral infections [15, 101].
SARS-CoV2 uses ACE2 as an entry site, thus, reducing ACE2 availability, promoting an
imbalance between the ACE2-Angiotensin II–Angiotensin 1 receptor axis and the ACE2-
ur

Angiotensin 1–7-Mas receptor axis [15, 102]. In line with this, recent studies have reported that
COVID-19 patients have increased Angiotensin II compared to healthy individuals [103].
Altogether, the abovementioned data suggests that COVID-19 infection leads to imbalanced
Jo

angiotensin conversion and a shift towards pro-inflammatory signaling mediated by Angiotensin


1 receptor axis [104] (Fig. 4, B).
Angiotensin II is known to significantly increase the expression of IL-6 in a dose-
dependent manner [105] through pleiotropic activation of NF-kB transcription factors [106],
[107]. Angiotensin 1 receptor downstream activation of NADPH oxidase results in elevated
oxidative stress, in particularly, superoxide that activates transcriptional mechanisms that
directly promotes IL-6 gene expression. In turn, plasma IL-6 concentrations correlate with blood
pressure, plasma Angiotensin II levels and vascular hypertrophy, suggesting an important role
of IL-6 in the development and maintenance of hypertension, specifically, that mediated by
Angiotensin II [108, 109]. IL-6 promotes Angiotensin 1 receptor expression sensitizing the
vascular wall to angiotensin II-dependent signaling mechanisms. Activated cells can release
cytokines, including IL-6, which promote expression of adhesion molecules and induce
endothelial activation, inflammatory cell infiltration and vascular inflammation [110]. This further
drives oxidative stress and IL-6 expression. Therefore, it is plausible that IL-6 and Angiotensin II
activate each other through a positive feedback mechanism. In support of this hypothesis,
ACE2 protects against lethal avian influenza A H5N1 infection [111] through prevention of
oxidative stress and acute respiratory distress syndrome. Moreover, it was recently reported
that in COVID-19 patients with hypertension Angiotensin receptor blockade attenuated the
inflammatory response, possibly through IL-6 inhibition [102, 112]. Taking together, Angiotensin
II accumulation due to SARS-CoV-2-mediated ACE2 downregulation may cause Angiotensin 1
receptor downstream activation of NADPH oxidase, which, in turn, leads to elevated ROS
production and to induction of transcriptional mechanisms that directly promote IL-6 expression,
implicated in inflammation-induced injury and immunopathology.

Risk factors associated with COVID-19


Chronic inflammation is the one common feature of hypertension, diabetes, obesity and age –
the risk factors associated with COVID-19 severe outcomes. Elderly, obese and diabetic

of
persons are affected by a low-grade chronic inflammation and are characterized by systemically
increased levels of proinflammatory cytokines, which may contribute to and facilitate the
cytokine storm, the main cause of COVID-19 mortality.

ro
Among conditions, predisposing patients infected with SARS-CoV-2 to severe outcomes,
diabetes is the most prominent. It is estimated that mortality rate of those with COVID-19 and
diabetes is about 16% [8] to 35% [113]. IL-6 is known for its dual role in autoimmune diabetes,
-p
as it is involved in the development of insulin resistance and β-cell dysfunction and, at the same
time, has anti-inflammatory effects and improves glucose metabolism [114]. Recent clinical data
demonstrates significantly elevated systemic IL-6 in COVID-19 patients with diabetes as
re
compared to those without diabetes [115]. COVID-19 may exacerbate the pre-existing diabetes,
since its entry site, ACE2, is expressed in the pancreas, with a potential role in the development
of insulin resistance and impaired insulin secretion [116]. IL-6 may be the link between diabetes
lP

and severe COVID-19 outcomes through its ability to modulate T cell response. Some indirect
evidence to support this comes from mouse studies of closely related MERS-CoV infection
demonstrating that abnormal elevation of IL-17A produced by IL-6-dependent Th17 cells
exacerbates disease severity in diabetic mice [117]. Consistent with that finding, patients with
na

COVID-19 have a higher proportion of Th17 cells in the blood [118], suggesting that diabetics
due to improper activation of Th1 and Th17 cells may have a compromised anti-viral immune
response [31].
Obese patients with COVID-19 are more likely to develop severe respiratory distress
ur

syndrome [119, 120]. Just as diabetics, obese patients have higher concentration of
proinflammatory cytokines both in steady state [121] and, apparently, during COVID-19 infection
[8]. In addition to decreased anti-viral T cell response, enhanced IL-6 production by adipose
Jo

tissue in obese patients [122] may alter innate anti-viral immune response by neutrophils [121]
and cause uncontrolled replication of the virus at the early stages of infection.
From the very beginning of COVID-19 pandemic, age was identified as a risk factor [38],
supporting the idea that ACE2 may be a marker of cell senescence [123]. Myofibroblasts, which
increase in number with age, express high levels of ACE2 [124], thus, myofibroblasts are more
susceptible to SARS-CoV-2, and elderly people, bearing higher proportion of myofibroblasts,
might be more susceptible to COVID-19 infection (reviewed in [125]). IL-6 is a critical
inflammatory regulator of senescence-associated secretory phenotype (SASP) [126]. Therefore,
IL-6 overexpression may correlate both with senescent status of cells and tissues, as well as
with increased expression of ACE2. In line with this idea, hydroxychloroquine, an anti-malaria
drug, which is now approved in some countries for COVID-19 treatment, effectively reduced the
salivary and serum levels of IL-6 in patients with Sjorgen’s syndrome [127]. Moreover,
azithromycin, prescribed together with hydroxychloroquine, can target and selectively remove
senescent cells [128] and, thus, provide anti-fibrotic activity by protecting the lungs from
inflammation-induced injury [129, 130].
Another issue facing elderly patients with COVID-19 is a compromised activation status
of their effector T cells. These cells may display limited function during prolonged infection as a
result of their exhaustion, associated with the surface expression of such immune-inhibitory
factors as PD-1 [75]. Since aging itself is a process known to promote reorganization of the T
cell landscape [131] and IL-6 is a SASP predictor and regulator, T cell exhaustion in the elderly
patients with COVID-19 is much more evident due to higher basal concentration of IL-6, which
can enhance the exhaustion process [132, 133] .
Smoking is another potential risk factor for SARS-CoV-2 infection. Smokers are more
vulnerable to influenza virus infections [134, 135] and are more likely to develop chronic
obstructive pulmonary disease (COPD) [136]. Enhanced ACE2 expression had been reported in
the lungs of healthy smokers [137, 138]. In line with that, studies in mice have established that
cigarette smoke can trigger the expansion of a subpopulation of respiratory epithelial cells that

of
express ACE2 in a dose-dependent manner [139]. Patients with COPD, as well as active
smokers, have increased ACE2 expression in the lungs [140], which may potentially explain a
fourfold increased risk of adverse development and severe outcome of COVID-19 [141, 142].

ro
Taken together, these recent reports suggest that smokers may be at higher risk of infection.
Examination of factors, predisposing to severe COVID-19 in Wuhan, China, revealed, that
19.2% of patients with severe disease were smokers [143]. On the other hand, preliminary
-p
meta-analysis based on Chinese patients suggest that active smoking was not associated with
the severity of COVID-19 [144], indicating that impact of smoking on COVID-19 progression is
controversial and requires further assessment.
re
Intuitively, patients with pre-existing asthma should display higher proportion of COVID-
19 severe cases. However, the prevalence of asthma in COVID-19 patients in Wuhan study was
markedly lower (0,9%) than that reported in the adult population of Wuhan (6,4%) [143]. One
lP

possible explanation is a shift toward Th2-type immune response in asthmatics as opposed to


Th1-associated inflammatory response induced by SARS-CoV-2 infection [118]. Recent study
reveals that TMRSS2 expression in upper respiratory tract correlates positively with type 2
response genes, especially with IL-13, whereas ACE2 expression is suppressed by IL-13.
na

Considering low numbers of cells co-expressing these proteases in the upper respiratory tract, it
has been proposed that in the context of IL-13-dependent reduction in ACE2 expression allergy
should promote resistance to infection [145]. In line with this, it was shown that ACE2
expression is decreased in nasal and bronchial epithelium of allergic individuals following
ur

allergen challenge, suggesting, that asthma-associated immune response actually protects from
COVID-19-induced pathology [146]. Nevertheless, further studies are required to reveal the
impact of COVID-19 severity in patients with preexisting asthma.
Jo

Meta-analysis studies of COVID-19 cases in China from December 2019 to February


2020 report lower incidence of the disease [147] and risk of death [35] in women than in men.
Similar data on association of gender with poorer prognosis for males were obtained earlier for
SARS-CoV [148], [149] and MERS-CoV [150, 151]. In connection to SARS-CoV-2, the X-linked
ACE2 gene is of a particular interest. Significant fraction of genes encoded on the X
chromosome undergo dosage compensation, however, ACE2 is located at Xp22, i.e. the area in
which genes may escape from X chromosome inactivation [152], possibly contributing to gender
disparity in disease susceptibility. Gender differences in immune responses are known to
depend not only on genetic, but also on endocrine factors [153, 154]. High doses of estradiol
acting through estrogen receptors decrease production of proinflammatory cytokines via
suppressing NF-kB signaling pathway [155]. In relation to COVID-19 the effects of estrogens on
IL-6 and on disease progression is of particular interest. Estrogen may suppress LPS-mediated
IL-6 expression in mouse macrophages by both blocking of NF-kB activation [155, 156] and
inhibiting p38 MAPK phosphorylation [156]. Furthermore, downregulation of IL-6 expression in
Kupffer cells appears to be ERα-dependent [157]. Overall, immunomodulatory effects of
estrogens may contribute to decreased susceptibility to SARS-CoV-2 infection in women and
this question requires further study.

The aftermath of COVID-19 infection


One of the common complications from the respiratory viral infections, including SARS [158]
and COVID-19 [159], is lung fibrosis. This is characterized by alveolar epithelial cell injury,
recruitment and activation of fibroblasts and subsequent production of extracellular matrix
(ECM) in the presence of specifically activated immune cells. Fibrosis develops as an
exaggerated repair process following injury associated with abnormal immune response leading
to chronic progressive disease [160]. Histological examination of lung biopsies from COVID-19
patients show massive apoptosis of pneumocytes, hyaline membrane formation, fibromyxoid
exudates [161] which are manifestations that are characteristic for ARDS and represent key

of
events preceding development of fibrosis (reviewed in [162]). Significant pro-fibrotic M2-like
SPP1+macrophage infiltration was also described for COVID-19 correlating with the disease
severity [71]. Damage to the epithelial layer caused by the virus further attracts various immune

ro
cells participating in inflammation-repair balance (monocytes, macrophages, granulocytes and T
cells). In addition, some viral components, such as N-protein and papain-like protease, can
directly stimulate production of TGF-β, one of the pro-fibrotic hallmarks in respiratory epithelium,
-p
similar to what has previously been reported for SARS-CoV [163, 164].
The role of IL-6 had been extensively studied in bleomycin model of lung fibrosis in mice.
Bleomycin is a chemotherapeutic antibiotic which causes damage of tissues lacking bleomycin
re
hydrolase, predominantly, in the lungs [165]. Mice with genetic ablation of IL-6 were
characterized by impaired fibrosis [166]. However, in the same model mice with
pharmacological rather than genetic inhibition of IL-6 demonstrated opposing phenotypes
lP

depending on the phase of inflammation-repair cycle. Specifically, IL-6 neutralization at the


early inflammatory phase (day 2 following bleomycin-induced lung injury) resulted in apoptosis
of pneumocytes, which further contributed to pulmonary fibrosis, whereas IL-6 blockade at the
early fibrotic stage (day 8 following bleomycin-induced lung injury) significantly ameliorated lung
na

fibrosis [167]. Interestingly, treatment of mice with recombinant soluble gp130Fc at the late
stage of chronic bleomycin model attenuated lung fibrosis, emphasizing the importance of sIL-
6Ra and IL-6 trans-signaling in mediating this condition [168]. It was further suggested that M2-
like infiltrating macrophages are the major source of soluble IL-6Ra in fibrotic foci, whereas
ur

pulmonary fibroblasts are the recipient cells, which proliferate and produce extracellular matrix
under trans-IL-6 activation. Finally, overexpression of IL-6 in respiratory tract mediated by
adenoviral vectors also resulted in the exacerbation of fibrosis and was associated with
Jo

increased levels of M2-macrophages with pro-fibrotic features [169]. These pro-fibrotic


macrophages are also characterized by increased IL-6Ra expression [168]. ACE2 expression is
reported for lung, heart, kidney, intestine, liver and brain. Thus, similar fibrosis-associated
complications should be expected, not just for respiratory tract, but also for other tissues and
organs in correlation with viral load and severity of cytokine release syndrome. Summarizing the
data available at the moment from infection-free experimental models of fibrosis, one can
conclude that IL-6 may affect fibroblast proliferation and functions directly or through
potentiation of a distinct macrophage phenotype. Taking into account the importance of this
cytokine at the repair stage, anti-IL-6 therapy may prove beneficial not only in acute phase, but
also in subacute proliferative phase of COVID-19-related ARDS.
SARS-CoV-2 tropism to neural system is of particular interest while neurological
symptoms, such as dizziness, headache, ataxia and anosmia, are often found in mild COVID-19
patients and develop early in the infection [170]. ACE2 expression in CNS is attributed to the
nuclei in brain stem and is associated with cardio‐respiratory control [171, 172]. Recent study
based on scRNA sequencing suggests co-expression of ACE2 and TMPRSS2 in olfactory
epithelial cells, but not in mature olfactory sensory neurons, which were initially considered as
the most obvious target population for the virus in CNS [173]. Even without the understanding of
the exact mechanisms of SARS-CoV-2 penetration and distribution in CNS, it should be noted
that as a result of the disseminated intravascular coagulopathy and cytokine storm caused by
infection, blood-brain-barrier permeability is significantly increased, which can lead to negative
consequences for CNS functioning [174, 175].

Therapeutic potential of IL-6 inhibitors


An effective therapeutic approach should include a combination of specific antiviral drugs, which
inhibit viral dissemination and limit direct virus-induced cytopathic effects coupled with anti-
inflammatory therapeutics, directed against immunopathology induced by a viral infection [176].

of
SARS-CoV-2-induced immunopathology exhibits features of systemic hyper-inflammation
reminiscent both of cytokine storm or macrophage activation syndrome and secondary
haemophagocytic lymphohistocytosis [177]. Since overexpression of IL-6 appears to be

ro
associated with severe COVID-19 outcomes, neutralizing antibodies used to treat a number of
autoimmune diseases by targeting exacerbated inflammatory immune response of the host may
provide a life-saving approach by preventing the cytokine release syndrome. In line with that,
-p
patients responding to CD19 CAR T cell therapy experience cytokine release syndrome [178],
which is well managed by IL-6 neutralizing antibodies [179]. In a non-randomized, open-label
clinical trial, 21 patients with severe or critical COVID-19 were treated with a single dose of
re
Tocilizumab, in addition to routine therapy [180]. 90% of patients recovered, suggesting that
anti-IL-6 might be a powerful potential rescue therapy in managing acute respiratory distress
syndrome of COVID-19 and inhibiting the most adverse outcome. Peripheral mononuclear cell
lP

scRNA-seq profiling data obtained from these patients indicate that treatment with Tocilizumab
does not reduce the number of cytotoxic CD8 + and plasma B cells, thus, does not interfere with
robust adaptive immune response [69]. Preliminary data from 21 patients with COVID-19 who
developed acute respiratory distress syndrome and participated in a compassionate-use
na

program at Papa Giovanni XXIII hospital in Bergamo, Italy, indicate that intravenous introduction
of Siltuximab reduced C-reactive protein level in all patients and significantly improved clinical
conditions with a reduced need for ventilation in 33% of patients. 43% of patients showed
stabilization of their conditions, and 24% of patients experienced a worsening of the disease
ur

and required intubation [181].


Sarilumab is a monoclonal antibody that works by inhibiting the IL-6 pathway through
binding and blocking the IL-6 receptor. Recently, another clinical trial was initiated, which will
Jo

evaluate the effects of Sarilumab in patients with severe COVID-19. This U.S.-based multi-
center, double-blind, phase 2/3 trial will begin at medical centers in New York and is anticipated
to enroll up to 400 patients [182].
The overview of initiated clinical trials of anti-IL-6 therapeutics in COVID-19 patients is
presented in Supplementary Table 1. In summary, strong scientific evidence of the beneficial
impact of IL-6 inhibitors in the modulation of the COVID-19 infection requires further clinical
data.

Conclusion
In the face of a pandemic, affecting millions of people worldwide, the need to develop new
approaches or reassess the existing ones in the treatment of severe viral infections is more
pressing than ever. Despite the fact that humanity experienced viral infections causing deadly
pneumonia twice in the past 20 years, there are still no drugs specific for coronaviruses. The
evidence discussed in this review suggests that for patients with risk factors, especially those
with high systemic levels of IL-6, clinically approved neutralizing antibodies against IL-6 or its
receptor, IL-6R, in combination with standard treatment protocols may provide benefit. Gaining
a deeper understanding of the factors that affect the immune response and mechanistic link of
these factors to disease severity is crucial for effective clinical management of COVID-19 in
severely affected patients.

Funding
This work was supported by the Ministry of Science and Higher Education of the Russian
Federation (grant 075-15-2019-1660).
Conflict of interest
The authors declare that there is no conflict of interest.

of
Acknowledgments
We thank Prof. S. A. Nedospasov and Dr. K.-S. N. Atretkhany for critical reading of manuscript.

ro
We thank P.V. Matveev for assistance with figure preparation.

References
[1] World Health Organization.
-p
Coronavirus disease (COVID-19) outbreak situation.
https://www.who.int/docs/default-source/coronaviruse/situation-reports/ 2020, April 6.
re
[2] Worldometer. COVID-19 CORONAVIRUS PANDEMIC. https://www.worldometers.info/coronavirus/
2020, April 6.
lP

[3] World Health Organization. Update 49 - SARS case fatality ratio, incubation period.
https://www.who.int/csr/sars/archive/2003_05_07a/en/ 2003, May 7.

[4] World Health Organization. Middle East respiratory syndrome coronavirus (MERS-CoV).
https://www.who.int/emergencies/mers-cov/en/ 2019, March 11.
na

[5] Ruan S. Likelihood of survival of coronavirus disease 2019. The Lancet Infectious Diseases. 2020,
March 30.

[6] Woo PC, Lau SK, Huang Y, Yuen KY. Coronavirus diversity, phylogeny and interspecies jumping. Exp
Biol Med (Maywood). 2009;234:1117-27.
ur

[7] Weiss SR, Leibowitz JL. Coronavirus pathogenesis. Adv Virus Res. 2011;81:85-164.

[8] Guo W, Li M, Dong Y, Zhou H, Zhang Z, Tian C, et al. Diabetes is a risk factor for the progression and
Jo

prognosis of COVID-19. Diabetes Metab Res Rev. 2020:e3319.


[9] Guan WJ, Ni ZY, Hu Y, Liang WH, Ou CQ, He JX, et al. Clinical Characteristics of Coronavirus
Disease 2019 in China. N Engl J Med. 2020;382:1708-20.

[10] Zhu N, Zhang D, Wang W, Li X, Yang B, Song J, et al. A Novel Coronavirus from Patients with
Pneumonia in China, 2019. N Engl J Med. 2020;382:727-33.

[11] Xu X, Yu C, Qu J, Zhang L, Jiang S, Huang D, et al. Imaging and clinical features of patients with
2019 novel coronavirus SARS-CoV-2. Eur J Nucl Med Mol Imaging. 2020;47:1275-80.

[12] Zhou P, Yang XL, Wang XG, Hu B, Zhang L, Zhang W, et al. A pneumonia outbreak associated with
a new coronavirus of probable bat origin. Nature. 2020;579:270-3.
[13] Wang Z, Ren L, Zhao X, Hung T, Meng A, Wang J, et al. Inhibition of severe acute respiratory
syndrome virus replication by small interfering RNAs in mammalian cells. J Virol. 2004;78:7523-7.
[14] Lan J, Ge J, Yu J, Shan S, Zhou H, Fan S, et al. Structure of the SARS-CoV-2 spike receptor-binding
domain bound to the ACE2 receptor. Nature. 2020.
[15] Kuba K, Imai Y, Rao S, Gao H, Guo F, Guan B, et al. A crucial role of angiotensin converting enzyme
2 (ACE2) in SARS coronavirus-induced lung injury. Nat Med. 2005;11:875-9.
[16] Ou X, Liu Y, Lei X, Li P, Mi D, Ren L, et al. Characterization of spike glycoprotein of SARS-CoV-2 on
virus entry and its immune cross-reactivity with SARS-CoV. Nat Commun. 2020;11:1620.

[17] Wang X, Xu W, Hu G, Xia S, Sun Z, Liu Z, et al. SARS-CoV-2 infects T lymphocytes through its spike
protein-mediated membrane fusion. Cell Mol Immunol. 2020.

[18] Hoffmann M, Kleine-Weber H, Schroeder S, Kruger N, Herrler T, Erichsen S, et al. SARS-CoV-2 Cell
Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor. Cell.
2020.

[19] Wrapp D, Wang N, Corbett KS, Goldsmith JA, Hsieh CL, Abiona O, et al. Cryo-EM structure of the

of
2019-nCoV spike in the prefusion conformation. Science. 2020;367:1260-3.

[20] Monteil V, Kwon H, Prado P, Hagelkruys A, Wimmer RA, Stahl M, et al. Inhibition of SARS-CoV-2
Infections in Engineered Human Tissues Using Clinical-Grade Soluble Human ACE2. Cell. 2020.

ro
[21] Wang K, Chen W, Zhou Y-S, Lian J-Q, Zhang Z, Du P, et al. SARS-CoV-2 invades host cells via a
novel route: CD147-spike protein. bioRxiv. https://doi.org/10.1101/2020.03.14.988345, 2020, March 14.
[22] Grass GD, Toole BP. How, with whom and when: an overview of CD147-mediated regulatory
-p
networks influencing matrix metalloproteinase activity. Biosci Rep. 2015;36:e00283.

[23] Crosnier C, Bustamante LY, Bartholdson SJ, Bei AK, Theron M, Uchikawa M, et al. Basigin is a
receptor essential for erythrocyte invasion by Plasmodium falciparum. Nature. 2011;480:534-7.
re
[24] Pushkarsky T, Zybarth G, Dubrovsky L, Yurchenko V, Tang H, Guo H, et al. CD147 facilitates HIV-1
infection by interacting with virus-associated cyclophilin A. Proc Natl Acad Sci U S A. 2001;98:6360-5.
lP

[25] Chen Z, Mi L, Xu J, Yu J, Wang X, Jiang J, et al. Function of HAb18G/CD147 in invasion of host cells
by severe acute respiratory syndrome coronavirus. J Infect Dis. 2005;191:755-60.

[26] Hamming I, Timens W, Bulthuis ML, Lely AT, Navis G, van Goor H. Tissue distribution of ACE2
protein, the functional receptor for SARS coronavirus. A first step in understanding SARS pathogenesis. J
na

Pathol. 2004;203:631-7.

[27] Bertram S, Heurich A, Lavender H, Gierer S, Danisch S, Perin P, et al. Influenza and SARS-
coronavirus activating proteases TMPRSS2 and HAT are expressed at multiple sites in human respiratory
and gastrointestinal tracts. PLoS One. 2012;7:e35876.
ur

[28] Lukassen S, Chua RL, Trefzer T, Kahn NC, Schneider MA, Muley T, et al. SARS-CoV-2 receptor
ACE2 and TMPRSS2 are primarily expressed in bronchial transient secretory cells. EMBO J.
2020:e105114.
Jo

[29] Chen Y, Feng Z, Diao B, Wang R, Wang G, Wang C, et al. The Novel Severe Acute Respiratory
Syndrome Coronavirus 2 (SARS-CoV-2) Directly Decimates Human Spleens and Lymph Nodes.
medRxiv. https://doi.org/10.1101/2020.03.27.20045427, 2020, March 27.

[30] Walls AC, Park YJ, Tortorici MA, Wall A, McGuire AT, Veesler D. Structure, Function, and
Antigenicity of the SARS-CoV-2 Spike Glycoprotein. Cell. 2020.

[31] Muniyappa R, Gubbi S. COVID-19 Pandemic, Corona Viruses, and Diabetes Mellitus. Am J Physiol
Endocrinol Metab. 2020.

[32] Wu C, Chen X, Cai Y, Xia J, Zhou X, Xu S, et al. Risk Factors Associated With Acute Respiratory
Distress Syndrome and Death in Patients With Coronavirus Disease 2019 Pneumonia in Wuhan, China.
JAMA Intern Med. 2020.
[33] Yang X, Yu Y, Xu J, Shu H, Xia Ja, Liu H, et al. Clinical course and outcomes of critically ill patients
with SARS-CoV-2 pneumonia in Wuhan, China: a single-centered, retrospective, observational study.
Lancet Respir Med. 2020;8:e26.
[34] Huang C, Wang Y, Li X, Ren L, Zhao J, Hu Y, et al. Clinical features of patients infected with 2019
novel coronavirus in Wuhan, China. Lancet. 2020;395:497-506.

[35] Worldometer. Age, Sex, Existing Conditions of COVID-19 Cases and Deaths. 2020, February 29.
[36] Chen G, Wu D, Guo W, Cao Y, Huang D, Wang H, et al. Clinical and immunologic features in severe
and moderate Coronavirus Disease 2019. J Clin Invest. 2020.

[37] Herold T, Jurinovic V, Arnreich C, Hellmuth JC, von Bergwelt-Baildon M, Klein M, et al. Level of IL-6
predicts respiratory failure in hospitalized symptomatic COVID-19 patients. medRxiv.
https://doi.org/10.1101/2020.04.01.20047381, 2020, April 1.

[38] Zhou F, Yu T, Du R, Fan G, Liu Y, Liu Z, et al. Clinical course and risk factors for mortality of adult
inpatients with COVID-19 in Wuhan, China: a retrospective cohort study. Lancet. 2020;395:1054-62.

[39] Gao Y, Li T, Han M, Li X, Wu D, Xu Y, et al. Diagnostic utility of clinical laboratory data

of
determinations for patients with the severe COVID-19. J Med Virol. 2020.

[40] Gong J, Dong H, Xia SQ, Huang YZ, Wang D, Zhao Y, et al. Correlation Analysis Between Disease
Severity and Inflammation-related Parameters in Patients with COVID-19 Pneumonia. medRxiv.

ro
https://doi.org/10.1101/2020.02.25.20025643, 2020, February 25.
[41] Xu X. A multicenter, randomized controlled trial for the efficacy and safety of tocilizumab in the
treatment of new coronavirus pneumonia (COVID-19). ChiCTR2020,
-p
https://clinicaltrials.gov/ct2/show/NCT04335071, February 13.

[42] Chomarat P, Banchereau J, Davoust J, Palucka AK. IL-6 switches the differentiation of monocytes
from dendritic cells to macrophages. Nat Immunol. 2000;1:510-4.
re
[43] Urashima M, Chauhan D, Hatziyanni M, Ogata A, Hollenbaugh D, Aruffo A, et al. CD40 ligand
triggers interleukin-6 mediated B cell differentiation. Leuk Res. 1996;20:507-15.
[44] Yang R, Masters AR, Fortner KA, Champagne DP, Yanguas-Casas N, Silberger DJ, et al. IL-6
lP

promotes the differentiation of a subset of naive CD8+ T cells into IL-21-producing B helper CD8+ T cells.
J Exp Med. 2016;213:2281-91.

[45] Diehl S, Rincon M. The two faces of IL-6 on Th1/Th2 differentiation. Mol Immunol. 2002;39:531-6.
na

[46] Coomes EA, Haghbayan H. Interleukin-6 in COVID-19: A Systematic Review and Meta-Analysis.
medRxiv. https://doi.org/10.1101/2020.03.30.20048058, 2020, March 30.

[47]. Chen X, Zhao B, Qu Y, Chen Y, Xiong J, Feng Y, Men D, Huang Q, Liu Y, Yang B, Ding J.
Detectable serum SARS-CoV-2 viral load (RNAaemia) is closely correlated with drastically elevated
ur

interleukin 6 (IL-6) level in critically ill COVID-19 patients. Clin Infect Dis. 2020 Apr 17.
[48] Zhang C, Wu Z, Li JW, Zhao H, Wang GQ. The cytokine release syndrome (CRS) of severe COVID-
19 and Interleukin-6 receptor (IL-6R) antagonist Tocilizumab may be the key to reduce the mortality. Int J
Jo

Antimicrob Agents. 2020:105954.

[49] Ulhaq ZS, Soraya GV. Interleukin-6 as a potential biomarker of COVID-19 progression. . Med Mal
Infect. 2020.

[50] Rose-John S, Winthrop K, Calabrese L. The role of IL-6 in host defence against infections:
immunobiology and clinical implications. Nat Rev Rheumatol. 2017;13:399-409.

[51] Cox MA, Kahan SM, Zajac AJ. Anti-viral CD8 T cells and the cytokines that they love. Virology.
2013;435:157-69.

[52] Bottcher JP, Schanz O, Garbers C, Zaremba A, Hegenbarth S, Kurts C, et al. IL-6 trans-signaling-
dependent rapid development of cytotoxic CD8+ T cell function. Cell Rep. 2014;8:1318-27.
[53] Eskilsson A, Mirrasekhian E, Dufour S, Schwaninger M, Engblom D, Blomqvist A. Immune-induced
fever is mediated by IL-6 receptors on brain endothelial cells coupled to STAT3-dependent induction of
brain endothelial prostaglandin synthesis. J Neurosci. 2014;34:15957-61.
[54] Evans SS, Repasky EA, Fisher DT. Fever and the thermal regulation of immunity: the immune
system feels the heat. Nat Rev Immunol. 2015;15:335-49.

[55] Yang ML, Wang CT, Yang SJ, Leu CH, Chen SH, Wu CL, et al. IL-6 ameliorates acute lung injury in
influenza virus infection. Sci Rep. 2017;7:43829.
[56] Dienz O, Rud JG, Eaton SM, Lanthier PA, Burg E, Drew A, et al. Essential role of IL-6 in protection
against H1N1 influenza virus by promoting neutrophil survival in the lung. Mucosal Immunol. 2012;5:258-
66.
[57] Lauder SN, Jones E, Smart K, Bloom A, Williams AS, Hindley JP, et al. Interleukin-6 limits influenza-
induced inflammation and protects against fatal lung pathology. Eur J Immunol. 2013;43:2613-25.
[58] Pyle CJ, Uwadiae FI, Swieboda DP, Harker JA. Early IL-6 signalling promotes IL-27 dependent
maturation of regulatory T cells in the lungs and resolution of viral immunopathology. PLoS Pathog.
2017;13:e1006640.

of
[59] Indalao IL, Sawabuchi T, Takahashi E, Kido H. IL-1beta is a key cytokine that induces trypsin
upregulation in the influenza virus-cytokine-trypsin cycle. Arch Virol. 2017;162:201-11.

ro
[60] Velazquez-Salinas L, Verdugo-Rodriguez A, Rodriguez LL, Borca MV. The Role of Interleukin 6
During Viral Infections. Front Microbiol. 2019;10:1057.
[61] Wu W, Dietze KK, Gibbert K, Lang KS, Trilling M, Yan H, et al. TLR ligand induced IL-6 counter-

2015;5:10501.
-p
regulates the anti-viral CD8(+) T cell response during an acute retrovirus infection. Sci Rep.

[62] Hou W, Kang HS, Kim BS. Th17 cells enhance viral persistence and inhibit T cell cytotoxicity in a
re
model of chronic virus infection. J Exp Med. 2009;206:313-28.

[63] Chuammitri P, Wongsawan K, Pringproa K, Thanawongnuwech R. Interleukin 17 (IL-17) manipulates


mouse bone marrow- derived neutrophils in response to acute lung inflammation. Comp Immunol
lP

Microbiol Infect Dis. 2019;67:101356.

[64] Liu R, Lauridsen HM, Amezquita RA, Pierce RW, Jane-Wit D, Fang C, et al. IL-17 Promotes
Neutrophil-Mediated Immunity by Activating Microvascular Pericytes and Not Endothelium. J Immunol.
2016;197:2400-8.
na

[65] Barnes BJ, Adrover JM, Baxter-Stoltzfus A, Borczuk A, Cools-Lartigue J, Crawford JM, et al.
Targeting potential drivers of COVID-19: Neutrophil extracellular traps. J Exp Med. 2020;217.

[66] Percopo CM, Ma M, Brenner TA, Krumholz JO, Break TJ, Laky K, et al. Critical Adverse Impact of IL-
6 in Acute Pneumovirus Infection. J Immunol. 2019;202:871-82.
ur

[67] Ruan Q, Yang K, Wang W, Jiang L, Song J. Clinical predictors of mortality due to COVID-19 based
on an analysis of data of 150 patients from Wuhan, China. Intensive Care Med. 2020.
Jo

[68] Monteleone G, Sarzi-Puttini PC, Ardizzone S. Preventing COVID-19-induced pneumonia with


anticytokine therapy. Lancet Rheumatol. 2020, April 6.
[69] Guo C, Li B, Ma H, Wang X, Cai P, Yu Q, et al. Tocilizumab treatment in severe COVID-19 patients
attenuates the inflammatory storm incited by monocyte centric immune interactions revealed by single-
cell analysis. bioRxiv. https://doi.org/10.1101/2020.04.08.029769, 2020, April 8.
[70] Tisoncik JR, Korth MJ, Simmons CP, Farrar J, Martin TR, Katze MG. Into the eye of the cytokine
storm. Microbiol Mol Biol Rev. 2012;76:16-32.

[71] Liao M, Liu Y, Yuan J, Wen Y, Xu G, Zhao J, Cheng L, Li J, Wang X, Wang F, Liu L. Single-cell
landscape of bronchoalveolar immune cells in patients with COVID-19. Nat Med. 2020 May 12:1-3.
[72] Channappanavar R, Fehr AR, Vijay R, Mack M, Zhao J, Meyerholz DK, et al. Dysregulated Type I
Interferon and Inflammatory Monocyte-Macrophage Responses Cause Lethal Pneumonia in SARS-CoV-
Infected Mice. Cell Host Microbe. 2016;19:181-93.
[73] Tseng CT, Perrone LA, Zhu H, Makino S, Peters CJ. Severe acute respiratory syndrome and the
innate immune responses: modulation of effector cell function without productive infection. J Immunol.
2005;174:7977-85.

[74] Law HK, Cheung CY, Ng HY, Sia SF, Chan YO, Luk W, et al. Chemokine up-regulation in SARS-
coronavirus-infected, monocyte-derived human dendritic cells. Blood. 2005;106:2366-74.
[75] Diao B, Wang C, Tan Y, Chen X, Liu Y, Ning L, Chen L, Li M, Liu Y, Wang G, Yuan Z. Reduction and
functional exhaustion of T cells in patients with coronavirus disease 2019 (COVID-19). Front Immunol.
2020 May 1;11:827.

[76] Zhao J, Zhao J, Van Rooijen N, Perlman S. Evasion by stealth: inefficient immune activation
underlies poor T cell response and severe disease in SARS-CoV-infected mice. PLoS Pathog.
2009;5:e1000636.
[77] Bost P, Giladi A, Liu Y, Bendjelal Y, Xu G, David E, et al. Host-viral infection maps reveal signatures

of
of severe COVID-19 patients. Cell. 2020.

[78] Karnowski A, Chevrier S, Belz GT, Mount A, Emslie D, D'Costa K, et al. B and T cells collaborate in
antiviral responses via IL-6, IL-21, and transcriptional activator and coactivator, Oct2 and OBF-1. J Exp

ro
Med. 2012;209:2049-64.

[79] Liu ZH, Wei R, Wu YP, Lisman T, Wang ZX, Han JJ, et al. Elevated plasma tissue-type plasminogen
activator (t-PA) and soluble thrombomodulin in patients suffering from severe acute respiratory syndrome
-p
(SARS) as a possible index for prognosis and treatment strategy. Biomed Environ Sci. 2005;18:260-4.

[80] Tang N, Li D, Wang X, Sun Z. Abnormal coagulation parameters are associated with poor prognosis
in patients with novel coronavirus pneumonia. J Thromb Haemost. 2020;18:844-7.
re
[81] Giannis D, Ziogas IA, Gianni P. Coagulation disorders in coronavirus infected patients: COVID-19,
SARS-CoV-1, MERS-CoV and lessons from the past. J Clin Virol. 2020;127:104362.
lP

[82] Han H, Yang L, Liu R, Liu F, Wu KL, Li J, et al. Prominent changes in blood coagulation of patients
with SARS-CoV-2 infection. Clin Chem Lab Med. 2020.

[83] Shi C, Wang C, Wang H, Yang C, Cai F, Zeng F, et al. The potential of low molecular weight heparin
to mitigate cytokine storm in severe COVID-19 patients: a retrospective clinical study. medRxiv.
na

https://doi.org/10.1101/2020.03.28.20046144, 2020, March 28.


[84] Bester J, Matshailwe C, Pretorius E. Simultaneous presence of hypercoagulation and increased clot
lysis time due to IL-1beta, IL-6 and IL-8. Cytokine. 2018;110:237-42.

[85] Nosaka M, Ishida Y, Kimura A, Kuninaka Y, Taruya A, Ozaki M, et al. Crucial Involvement of IL-6 in
ur

Thrombus Resolution in Mice via Macrophage Recruitment and the Induction of Proteolytic Enzymes.
Front Immunol. 2019;10:3150.
[86] Sackett K, Cunderlik M, Sahni N, Killeen AA, Olson AP. Extreme Hyperferritinemia: Causes and
Jo

Impact on Diagnostic Reasoning. Am J Clin Pathol. 2016;145:646-50.

[87] Crayne CB, Albeituni S, Nichols KE, Cron RQ. The Immunology of Macrophage Activation
Syndrome. Front Immunol. 2019;10:119.
[88] Haga S, Yamamoto N, Nakai-Murakami C, Osawa Y, Tokunaga K, Sata T, et al. Modulation of TNF-
alpha-converting enzyme by the spike protein of SARS-CoV and ACE2 induces TNF-alpha production
and facilitates viral entry. Proc Natl Acad Sci U S A. 2008;105:7809-14.
[89] Feldmann M, Maini RN, Woody JN, Holgate ST, Winter G, Rowland M, et al. Trials of anti-tumour
necrosis factor therapy for COVID-19 are urgently needed. Lancet. 2020.

[90] Hussell T, Pennycook A, Openshaw PJ. Inhibition of tumor necrosis factor reduces the severity of
virus-specific lung immunopathology. Eur J Immunol. 2001;31:2566-73.
[91] Fong Y, Tracey KJ, Moldawer LL, Hesse DG, Manogue KB, Kenney JS, et al. Antibodies to
cachectin/tumor necrosis factor reduce interleukin 1 beta and interleukin 6 appearance during lethal
bacteremia. J Exp Med. 1989;170:1627-33.
[92] Brenner EJ, Ungaro RC, Colombel JF, Kappelman MD. SECURE-IBD Database Public Data Update.
2020, April 30.

[93] Neurath MF. Covid-19 and immunomodulation in IBD. Gut. 2020.


[94] Wu Z, McGoogan JM. Characteristics of and Important Lessons From the Coronavirus Disease 2019
(COVID-19) Outbreak in China: Summary of a Report of 72314 Cases From the Chinese Center for
Disease Control and Prevention. JAMA. 2020.

[95] Chae CU, Lee RT, Rifai N, Ridker PM. Blood pressure and inflammation in apparently healthy men.
Hypertension. 2001;38:399-403.

[96] Gibas-Dorna M, Nowak D, Piatek J, Pupek-Musialik D, Krauss H, Kopczynski P. Plasma ghrelin and
interleukin-6 levels correlate with body mass index and arterial blood pressure in males with essential
hypertension. J Physiol Pharmacol. 2015;66:367-72.

of
[97] Naya M, Tsukamoto T, Morita K, Katoh C, Furumoto T, Fujii S, et al. Plasma interleukin-6 and tumor
necrosis factor-alpha can predict coronary endothelial dysfunction in hypertensive patients. Hypertens
Res. 2007;30:541-8.

ro
[98] Suematsu S, Matsuda T, Aozasa K, Akira S, Nakano N, Ohno S, et al. IgG1 plasmacytosis in
interleukin 6 transgenic mice. Proc Natl Acad Sci U S A. 1989;86:7547-51.
[99] Steiner MK, Syrkina OL, Kolliputi N, Mark EJ, Hales CA, Waxman AB. Interleukin-6 overexpression
-p
induces pulmonary hypertension. Circ Res. 2009;104:236-44, 28p following 44.

[100] Hampl V, Herget J, Bibova J, Banasova A, Huskova Z, Vanourkova Z, et al. Intrapulmonary


activation of the angiotensin-converting enzyme type 2/angiotensin 1-7/G-protein-coupled Mas receptor
re
axis attenuates pulmonary hypertension in Ren-2 transgenic rats exposed to chronic hypoxia. Physiol
Res. 2015;64:25-38.
[101] Yang P, Gu H, Zhao Z, Wang W, Cao B, Lai C, et al. Angiotensin-converting enzyme 2 (ACE2)
lP

mediates influenza H7N9 virus-induced acute lung injury. Sci Rep. 2014;4:7027.

[102] Meng J, Xiao G, Zhang J, He X, Ou M, Bi J, et al. Renin-angiotensin system inhibitors improve the
clinical outcomes of COVID-19 patients with hypertension. Emerg Microbes Infect. 2020;9:757-60.

[103] Liu Y, Yang Y, Zhang C, Huang F, Wang F, Yuan J, et al. Clinical and biochemical indexes from
na

2019-nCoV infected patients linked to viral loads and lung injury. Sci China Life Sci. 2020;63:364-74.

[104] Wang X, Khaidakov M, Ding Z, Mitra S, Lu J, Liu S, et al. Cross-talk between inflammation and
angiotensin II: studies based on direct transfection of cardiomyocytes with AT1R and AT2R cDNA. Exp
Biol Med (Maywood). 2012;237:1394-401.
ur

[105] Funakoshi Y, Ichiki T, Ito K, Takeshita A. Induction of interleukin-6 expression by angiotensin II in


rat vascular smooth muscle cells. Hypertension. 1999;34:118-25.
Jo

[106] Han Y, Runge MS, Brasier AR. Angiotensin II induces interleukin-6 transcription in vascular smooth
muscle cells through pleiotropic activation of nuclear factor-kappa B transcription factors. Circ Res.
1999;84:695-703.

[107] Sano M, Fukuda K, Sato T, Kawaguchi H, Suematsu M, Matsuda S, et al. ERK and p38 MAPK, but
not NF-kappaB, are critically involved in reactive oxygen species-mediated induction of IL-6 by
angiotensin II in cardiac fibroblasts. Circ Res. 2001;89:661-9.
[108] Gomolak JR, Didion SP. Angiotensin II-induced endothelial dysfunction is temporally linked with
increases in interleukin-6 and vascular macrophage accumulation. Front Physiol. 2014;5:396.

[109] Ala Y, Palluy O, Favero J, Bonne C, Modat G, Dornand J. Hypoxia/reoxygenation stimulates


endothelial cells to promote interleukin-1 and interleukin-6 production. Effects of free radical scavengers.
Agents Actions. 1992;37:134-9.
[110] Liu PP, Blet A, Smyth D, Li H. The Science Underlying COVID-19: Implications for the
Cardiovascular System. Circulation. 2020.
[111] Zou Z, Yan Y, Shu Y, Gao R, Sun Y, Li X, et al. Angiotensin-converting enzyme 2 protects from
lethal avian influenza A H5N1 infections. Nat Commun. 2014;5:3594.
[112] Yang G, Tan Z, Zhou L, Yang M, Peng L, Liu J, et al. Angiotensin II Receptor Blockers and
Angiotensin-Converting Enzyme Inhibitors Usage is Associated with Improved Inflammatory Status and
Clinical Outcomes in COVID-19 Patients With Hypertension. medRxiv.
https://doi.org/10.1101/2020.03.31.20038935, 2020, March 31.
[113] Onder G, Rezza G, Brusaferro S. Case-Fatality Rate and Characteristics of Patients Dying in
Relation to COVID-19 in Italy. JAMA. 2020.
[114] Akbari M, Hassan-Zadeh V. IL-6 signalling pathways and the development of type 2 diabetes.
Inflammopharmacology. 2018;26:685-98.
[115] Zhu L, She ZG, Cheng X, Qin JJ, Zhang XJ, Cai J, et al. Association of Blood Glucose Control and
Outcomes in Patients with COVID-19 and Pre-existing Type 2 Diabetes. Cell Metab. 2020.

of
[116] Bindom SM, Lazartigues E. The sweeter side of ACE2: physiological evidence for a role in diabetes.
Mol Cell Endocrinol. 2009;302:193-202.

[117] Kulcsar KA, Coleman CM, Beck SE, Frieman MB. Comorbid diabetes results in immune

ro
dysregulation and enhanced disease severity following MERS-CoV infection. JCI Insight. 2019;4.
[118] Zhang JJ, Dong X, Cao YY, Yuan YD, Yang YB, Yan YQ, et al. Clinical characteristics of 140
patients infected with SARS-CoV-2 in Wuhan, China. Allergy. 2020.
-p
[119] Simonnet A, Chetboun M, Poissy J, Raverdy V, Noulette J, Duhamel A, et al. High prevalence of
obesity in severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) requiring invasive mechanical
ventilation. Obesity (Silver Spring). 2020.
re
[120] Huang JF, Wang XB, Zheng KI, Liu WY, Chen JJ, George J, et al. Obesity hypoventilation
syndrome and severe COVID-19. Metabolism. 2020:154249.
[121] Richard C, Wadowski M, Goruk S, Cameron L, Sharma AM, Field CJ. Individuals with obesity and
lP

type 2 diabetes have additional immune dysfunction compared with obese individuals who are
metabolically healthy. BMJ Open Diabetes Res Care. 2017;5:e000379.

[122] Maachi M, Pieroni L, Bruckert E, Jardel C, Fellahi S, Hainque B, et al. Systemic low-grade
inflammation is related to both circulating and adipose tissue TNFalpha, leptin and IL-6 levels in obese
na

women. Int J Obes Relat Metab Disord. 2004;28:993-7.

[123] Song J, Hu B, Qu H, Wang L, Huang X, Li M, et al. Upregulation of angiotensin converting enzyme


2 by shear stress reduced inflammation and proliferation in vascular endothelial cells. Biochem Biophys
Res Commun. 2020;525:812-8.
ur

[124] Guy JL, Lambert DW, Turner AJ, Porter KE. Functional angiotensin-converting enzyme 2 is
expressed in human cardiac myofibroblasts. Exp Physiol. 2008;93:579-88.
Jo

[125] Sargiacomo C, Sotgia F, Lisanti MP. COVID-19 and chronological aging: senolytics and other anti-
aging drugs for the treatment or prevention of corona virus infection? Aging (Albany NY). 2020.
[126] Mosteiro L, Pantoja C, de Martino A, Serrano M. Senescence promotes in vivo reprogramming
through p16(INK)(4a) and IL-6. Aging Cell. 2018;17.
[127] Tishler M, Yaron I, Shirazi I, Yaron M. Hydroxychloroquine treatment for primary Sjogren's
syndrome: its effect on salivary and serum inflammatory markers. Ann Rheum Dis. 1999;58:253-6.

[128] Ozsvari B, Nuttall JR, Sotgia F, Lisanti MP. Azithromycin and Roxithromycin define a new family of
"senolytic" drugs that target senescent human fibroblasts. Aging (Albany NY). 2018;10:3294-307.

[129] Tang F, Li R, Xue J, Lan J, Xu H, Liu Y, et al. Azithromycin attenuates acute radiation-induced lung
injury in mice. Oncol Lett. 2017;14:5211-20.
[130] Mosquera RA, De Jesus-Rojas W, Stark JM, Yadav A, Jon CK, Atkins CL, et al. Role of prophylactic
azithromycin to reduce airway inflammation and mortality in a RSV mouse infection model. Pediatr
Pulmonol. 2018;53:567-74.
[131] Elyahu Y, Hekselman I, Eizenberg-Magar I, Berner O, Strominger I, Schiller M, et al. Aging
promotes reorganization of the CD4 T cell landscape toward extreme regulatory and effector phenotypes.
Sci Adv. 2019;5:eaaw8330.

[132] Kato T, Nishida T, Murase M, Ito Y, Murata M, Naoe T. Correlation of Serum IL-6 Level with
Exhaustion of Cytomegalovirus-specific T Cells After Hematopoietic Stem Cell Transplantation. Blood.
2011;118:360-.
[133] Naseem S, Manzoor S, Javed A, Abbas S. Interleukin-6 Rescues Lymphocyte from Apoptosis and
Exhaustion Induced by Chronic Hepatitis C Virus Infection. Viral Immunol. 2018;31:624-31.

[134] Lawrence H, Hunter A, Murray R, Lim WS, McKeever T. Cigarette smoking and the occurrence of
influenza - Systematic review. J Infect. 2019;79:401-6.
[135] Han L, Ran J, Mak YW, Suen LK, Lee PH, Peiris JSM, et al. Smoking and Influenza-associated
Morbidity and Mortality: A Systematic Review and Meta-analysis. Epidemiology. 2019;30:405-17.

of
[136] Ji W, Lim MN, Bak SH, Hong SH, Han SS, Lee SJ, et al. Differences in chronic obstructive
pulmonary disease phenotypes between non-smokers and smokers. Clin Respir J. 2018;12:666-73.

ro
[137] Cai G, Bosse Y, Xiao F, Kheradmand F, Amos CI. Tobacco Smoking Increases the Lung Gene
Expression of ACE2, the Receptor of SARS-CoV-2. Am J Respir Crit Care Med. 2020.
[138] Brake SJ, Barnsley K, Lu W, McAlinden KD, Eapen MS, Sohal SS. Smoking Upregulates

CoV-2 (Covid-19). J Clin Med. 2020;9.


-p
Angiotensin-Converting Enzyme-2 Receptor: A Potential Adhesion Site for Novel Coronavirus SARS-

[139] Smith JC, Sausville EL, Girish V, Yuan ML, John KM, Sheltzer JM. Cigarette smoke exposure and
re
inflammatory signaling increase the expression of the SARS-CoV-2 receptor ACE2 in the respiratory
tract. Dev Cell. 2020, May 16.
[140] Leung JM, Yang CX, Tam A, Shaipanich T, Hackett TL, Singhera GK, et al. ACE-2 Expression in
lP

the Small Airway Epithelia of Smokers and COPD Patients: Implications for COVID-19. Eur Respir J.
2020.

[141] Zhao Q, Meng M, Kumar R, Wu Y, Huang J, Lian N, et al. The impact of COPD and smoking history
on the severity of Covid-19: A systemic review and meta-analysis. J Med Virol. 2020.
na

[142] Wang B, Li R, Lu Z, Huang Y. Does comorbidity increase the risk of patients with COVID-19:
evidence from meta-analysis. Aging (Albany NY). 2020;12:6049-57.

[143] Li X, Xu S, Yu M, Wang K, Tao Y, Zhou Y, et al. Risk factors for severity and mortality in adult
COVID-19 inpatients in Wuhan. J Allergy Clin Immunol. 2020.
ur

[144] Lippi G, Henry BM. Active smoking is not associated with severity of coronavirus disease 2019
(COVID-19). Eur J Intern Med. 2020;75:107-8.
Jo

[145] Sajuthi SP, DeFord P, Jackson ND, Montgomery MT, Everman Jamie L. , Rios CL, et al. Type 2
and interferon inflammation strongly regulate SARS-CoV-2 related gene expression in the airway
epithelium. bioRxiv. https://doi.org/10.1101/2020.04.09.034454, 2020, April 9.

[146] Jackson DJ, Busse WW, Bacharier LB, Kattan M, O'Connor GT, Wood RA, et al. Association of
Respiratory Allergy, Asthma and Expression of the SARS-CoV-2 Receptor, ACE2. J Allergy Clin
Immunol. 2020.
[147] Li LQ, Huang T, Wang YQ, Wang ZP, Liang Y, Huang TB, et al. COVID-19 patients' clinical
characteristics, discharge rate, and fatality rate of meta-analysis. J Med Virol. 2020.

[148] Karlberg J, Chong DS, Lai WY. Do men have a higher case fatality rate of severe acute respiratory
syndrome than women do? Am J Epidemiol. 2004;159:229-31.
[149] Leong HN, Earnest A, Lim HH, Chin CF, Tan C, Puhaindran ME, et al. SARS in Singapore--
predictors of disease severity. Ann Acad Med Singapore. 2006;35:326-31.
[150] Badawi A, Ryoo SG. Prevalence of comorbidities in the Middle East respiratory syndrome
coronavirus (MERS-CoV): a systematic review and meta-analysis. Int J Infect Dis. 2016;49:129-33.
[151] Alghamdi IG, Hussain, II, Almalki SS, Alghamdi MS, Alghamdi MM, El-Sheemy MA. The pattern of
Middle East respiratory syndrome coronavirus in Saudi Arabia: a descriptive epidemiological analysis of
data from the Saudi Ministry of Health. Int J Gen Med. 2014;7:417-23.
[152] Burrell LM, Harrap SB, Velkoska E, Patel SK. The ACE2 gene: its potential as a functional
candidate for cardiovascular disease. Clin Sci (Lond). 2013;124:65-76.

[153] Klein SL, Flanagan KL. Sex differences in immune responses. Nat Rev Immunol. 2016;16:626-38.

[154] La Vignera S, Cannarella R, Condorelli RA, Torre F, Aversa A, Calogero AE. Sex-Specific SARS-
CoV-2 Mortality: Among Hormone-Modulated ACE2 Expression, Risk of Venous Thromboembolism and
Hypovitaminosis D. Int J Mol Sci. 2020;21.

[155] Biswas DK, Singh S, Shi Q, Pardee AB, Iglehart JD. Crossroads of estrogen receptor and NF-

of
kappaB signaling. Sci STKE. 2005;2005:pe27.

[156] Liu L, Zhao Y, Xie K, Sun X, Jiang L, Gao Y, et al. Estrogen inhibits LPS-induced IL-6 production in
macrophages partially via the nongenomic pathway. Immunol Invest. 2014;43:693-704.

ro
[157] Naugler WE, Sakurai T, Kim S, Maeda S, Kim K, Elsharkawy AM, et al. Gender disparity in liver
cancer due to sex differences in MyD88-dependent IL-6 production. Science. 2007;317:121-4.
[158] van den Brand JM, Haagmans BL, van Riel D, Osterhaus AD, Kuiken T. The pathology and
-p
pathogenesis of experimental severe acute respiratory syndrome and influenza in animal models. J Comp
Pathol. 2014;151:83-112.

[159] Xu YH, Dong JH, An WM, Lv XY, Yin XP, Zhang JZ, et al. Clinical and computed tomographic
re
imaging features of novel coronavirus pneumonia caused by SARS-CoV-2. J Infect. 2020;80:394-400.
[160] Florez-Sampedro L, Song S, Melgert BN. The diversity of myeloid immune cells shaping wound
repair and fibrosis in the lung. Regeneration (Oxf). 2018;5:3-25.
lP

[161] Xu Z, Shi L, Wang Y, Zhang J, Huang L, Zhang C, et al. Pathological findings of COVID-19
associated with acute respiratory distress syndrome. Lancet Respir Med. 2020;8:420-2.
[162] Cabrera-Benitez NE, Laffey JG, Parotto M, Spieth PM, Villar J, Zhang H, et al. Mechanical
na

ventilation-associated lung fibrosis in acute respiratory distress syndrome: a significant contributor to poor
outcome. Anesthesiology. 2014;121:189-98.

[163] Zhao X, Nicholls JM, Chen YG. Severe acute respiratory syndrome-associated coronavirus
nucleocapsid protein interacts with Smad3 and modulates transforming growth factor-beta signaling. J
ur

Biol Chem. 2008;283:3272-80.


[164] Li SW, Yang TC, Wan L, Lin YJ, Tsai FJ, Lai CC, et al. Correlation between TGF-beta1 expression
and proteomic profiling induced by severe acute respiratory syndrome coronavirus papain-like protease.
Jo

Proteomics. 2012;12:3193-205.

[165] Liu T, De Los Santos FG, Phan SH. The Bleomycin Model of Pulmonary Fibrosis. Methods Mol Biol.
2017;1627:27-42.

[166] Saito F, Tasaka S, Inoue K, Miyamoto K, Nakano Y, Ogawa Y, et al. Role of interleukin-6 in
bleomycin-induced lung inflammatory changes in mice. Am J Respir Cell Mol Biol. 2008;38:566-71.

[167] Kobayashi T, Tanaka K, Fujita T, Umezawa H, Amano H, Yoshioka K, et al. Bidirectional role of IL-6
signal in pathogenesis of lung fibrosis. Respir Res. 2015;16:99.

[168] Le TT, Karmouty-Quintana H, Melicoff E, Le TT, Weng T, Chen NY, et al. Blockade of IL-6 Trans
signaling attenuates pulmonary fibrosis. J Immunol. 2014;193:3755-68.
[169] Ayaub EA, Dubey A, Imani J, Botelho F, Kolb MRJ, Richards CD, et al. Overexpression of OSM and
IL-6 impacts the polarization of pro-fibrotic macrophages and the development of bleomycin-induced lung
fibrosis. Sci Rep. 2017;7:13281.
[170] Mao L, Wang M, Chen S, He Q, Chang J, Hong C, et al. Neurological Manifestations of Hospitalized
Patients with COVID-19 in Wuhan, China: a retrospective case series study. JAMA Neurol. 2020 Apr 10.

[171] Yamazato M, Yamazato Y, Sun C, Diez-Freire C, Raizada MK. Overexpression of angiotensin-


converting enzyme 2 in the rostral ventrolateral medulla causes long-term decrease in blood pressure in
the spontaneously hypertensive rats. Hypertension. 2007;49:926-31.
[172] Feng Y, Yue X, Xia H, Bindom SM, Hickman PJ, Filipeanu CM, et al. Angiotensin-converting
enzyme 2 overexpression in the subfornical organ prevents the angiotensin II-mediated pressor and
drinking responses and is associated with angiotensin II type 1 receptor downregulation. Circ Res.
2008;102:729-36.

[173] Brann DH, Tsukahara T, Weinreb C, Lipovsek M, Van den Berge K, Gong B, et al. Non-neuronal
expression of SARS-CoV-2 entry genes in the olfactory system suggests mechanisms underlying COVID-
19-associated anosmia. bioRxiv. https://doi.org/10.1101/2020.03.25.009084, 2020, March 25.

of
[174] Clark IA, Vissel B. The meteorology of cytokine storms, and the clinical usefulness of this
knowledge. Semin Immunopathol. 2017;39:505-16.

[175] Yokota S, Imagawa T, Miyamae T, Ito S, Nakajima S, Nezu A, et al. Hypothetical pathophysiology

ro
of acute encephalopathy and encephalitis related to influenza virus infection and hypothermia therapy.
Pediatr Int. 2000;42:197-203.
[176] Stebbing J, Phelan A, Griffin I, Tucker C, Oechsle O, Smith D, et al. COVID-19: combining antiviral
-p
and anti-inflammatory treatments. Lancet Infect Dis. 2020;20:400-2.

[177] McGonagle D, Sharif K, O'Regan A, Bridgewood C. The Role of Cytokines including Interleukin-6 in
COVID-19 induced Pneumonia and Macrophage Activation Syndrome-Like Disease. Autoimmun Rev.
re
2020:102537.

[178] Teachey DT, Lacey SF, Shaw PA, Melenhorst JJ, Maude SL, Frey N, et al. Identification of
Predictive Biomarkers for Cytokine Release Syndrome after Chimeric Antigen Receptor T-cell Therapy for
lP

Acute Lymphoblastic Leukemia. Cancer Discov. 2016;6:664-79.

[179] Agarwal S, June CH. Harnessing CAR T Cell Insights to Develop Treatments for Hyperinflammatory
Responses in COVID-19 patients. Cancer Discov. 2020.
na

[180] Xu X, Han M, Li T, Sun W, Wang D, Fu B, et al. Effective treatment of severe COVID-19 patients
with tocilizumab. Proc Natl Acad Sci U S A. 2020.
[181] Gritti G, Raimondi F, Ripamonti D, Riva I, Landi F, Alborghetti L, et al. Use of siltuximab in patients
with COVID-19 pneumonia requiring ventilatory support. medRxiv.
ur

https://doi.org/10.1101/2020.04.01.20048561, 2020, April 1.

[182] Sanofi. Sanofi and Regeneron begin global Kevzara® (sarilumab) clinical trial program in patients
with severe COVID-19. http://www.news.sanofi.us/2020-03-16-Sanofi-and-Regeneron-begin-global-
Jo

Kevzara-R-sarilumab-clinical-trial-program-in-patients-with-severe-COVID-19, 2020, March 16.


Ekaterina O. Gubernatorova is PhD student in Immunology at Lomonosov Moscow State
University (MSU) and junior staff scientist in Laboratory of Molecular Mechanisms of Immunity,
Center for Precision Genome Editing and Genetic Technologies for Biomedicine Engelhardt
Institute of Molecular Biology, Russian Academy of Sciences (EIMB RAS). Ekaterina is working

of
on several research projects in the areas of mucosal immunology. She is focused on the role of
cell-specific IL-6 signaling in lungs and gut homeostasis and inflammation.

ro
-p
re
lP

Ekaterina A. Gorshkova is PhD student in Immunology at Lomonosov MSU and junior staff
na

scientist in Laboratory of Molecular Mechanisms of Immunity, Center for Precision Genome


Editing and Genetic Technologies for Biomedicine, EIMB RAS. Ekaterina is involved in several
research projects, including studying the role of IL-6 overexpression in mouse model of
inflammatory disease.
ur
Jo
Almina I. Polinova is bachelor student in Immunology in Lomonosov MSU. Almina received her
bachelor training in the laboratory of Molecular Mechanisms of Immunity, participating in a
project to study the role of IL-6 in gut inflammation.

of
Marina S. Drutskaya, PhD, is Leading Staff Scientist in Laboratory of Molecular Mechanisms of
Immunity, Center for Precision Genome Editing and Genetic Technologies for Biomedicine,
EIMB RAS. Marina is studying proinflammatory cytokines, mainly TNF and IL-6, in several

ro
relevant autoimmune and inflammatory disease models in mice.

-p
re
lP
na
ur

Figure 1. Schematic representation of SARS-CoV-2 interaction with a target cell


Jo

Binding of virion to ACE2 is followed by serine protease, TMPRSS2, cleavage of the viral spike
protein and induction of internalization of the complex with subsequent reduction of ACE2
surface availability. Then another protease, Furin, releases spike fusion peptide, and the virus
enters the host cell through an endosomal pathway.
Figure 2. Role of IL-6 in respiratory viral infections.

of
IL-6 demonstrates opposing effects during the immune response to viral infections in the lungs.
IL-6 promotes highly specific reaction of adaptive immunity by stimulating of CD8+ T cells and B

ro
cells, which is balanced by T regulatory cells. Also, IL-6 facilitates survival of phagocytic
neutrophils. On the other hand, IL-6 can provide unfavorable Th2 and Th17 over Th1 helper
differentiation and facilitate tissue injury by dysregulation of extracellular matrix and attraction of
neutrophils and pro-inflammatory macrophages. -p
re
lP
na
ur
Jo
of
ro
-p
re
lP

Figure 3. Cytokine release syndrome in COVID-19.


SARS-CoV-2 replicates in alveolar epithelial cells. After the virions are released, they can infect
or are being captured by macrophages, dendritic cells and neutrophils. At the same time,
damaged epithelial cells produce alarmins, which are sensed by the neighboring epithelial and
na

myeloid cells. All these events lead to release of proinflammatory cytokines, increase of alveolar
vessel permeability and cell recruitment to the site of infection, forming positive loop for
pathological activation of IL-6, IL-1, TNF and other proinflammatory cytokines. Due to high
permeability of pulmonary vessels under inflammatory conditions, SARS-CoV-2 can inseminate
ur

distant ACE2-expressing tissues (intestine, kidney, pancreas), and trigger burst of inflammation
at distant sites.
Jo
of
ro
-p
re
lP
na
ur
Jo

Figure 4. ACE2 downregulation may drive IL-6 production in Angiotensin II-dependent


manner.
A – in healthy state pro-inflammatory signaling of Angiotensin II via Angiotensin 1 receptor
(AT1R) is stabilized by Angiotensin 1-7 (ACE2-Angiotensin I proteolysis product) signaling via
Mas receptor (MasR) that has anti-inflammatory effect.
B – virus-mediated down-regulation of ACE2 leads to Angiotensin II accumulation in the
absence of Angiotensin 1-7 and acceleration of its proinflammatory effect. AT1R downstream
activation of NADPH oxidase results in elevated ROS directly promoting IL-6 expression. In its
turn, IL-6 can positively regulate AT1R levels. Thus, by acting together these processes
exacerbate hypertension, edema, infiltration of immune cells and inflammation in COVID-19
affected lungs.

of
ro
-p
re
lP
na
ur
Jo

You might also like