ArticlePDF AvailableLiterature Review

A Vicious Cycle: In Severe and Critically Ill COVID-19 Patients

Frontiers
Frontiers in Immunology
Authors:

Abstract and Figures

The coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus, is one of the fastest-evolving viral diseases that has instigated a worldwide pandemic. Severe inflammatory syndrome and venous thrombosis are commonly noted in COVID-19 patients with severe and critical illness, contributing to the poor prognosis. Interleukin (IL)-6, a major complex inflammatory cytokine, is an independent factor in predicting the severity of COVID-19 disease in patients. IL-6 and tumor necrosis factor (TNF)-α participate in COVID-19-induced cytokine storm, causing endothelial cell damage and upregulation of plasminogen activator inhibitor-1 (PAI-1) levels. In addition, IL-6 and PAI-1 form a vicious cycle of inflammation and thrombosis, which may contribute to the poor prognosis of patients with severe COVID-19. Targeted inhibition of IL-6 and PAI-1 signal transduction appears to improve treatment outcomes in severely and critically ill COVID-19 patients suffering from cytokine storms and venous thrombosis. Motivated by studies highlighting the relationship between inflammatory cytokines and thrombosis in viral immunology, we provide an overview of the immunothrombosis and immunoinflammation vicious loop between IL-6 and PAI-1. Our goal is that understanding this ferocious circle will benefit critically ill patients with COVID-19 worldwide.
This content is subject to copyright.
A Vicious Cycle: In Severe and
Critically Ill COVID-19 Patients
Peifeng Huang
1
, Qingwei Zuo
1
, Yue Li
2
, Patrick Kwabena Oduro
3
, Fengxian Tan
1
,
Yuanyuan Wang
1
, Xiaohui Liu
1
, Jing Li
2
, Qilong Wang
3
, Fei Guo
4
*,
Yue Li
3
*
and Long Yang
1,5
*
1
School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China,
2
School of Department of
Clinical Training and Teaching of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin,
China,
3
State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine,
Tianjin, China,
4
National Health Commission of the Peoples Republic of China Key Laboratory of Systems Biology of
Pathogens, Institute of Pathogen Biology and Center for AIDS Research, Chinese Academy of Medical Sciences & Peking
Union Medical College, Beijing, China,
5
Research Center for Infectious Diseases, Tianjin University of Traditional Chinese
Medicine, Tianjin, China
The coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory
syndrome coronavirus 2 (SARS-CoV-2) virus, is one of the fastest-evolving viral
diseases that has instigated a worldwide pandemic. Severe inammatory syndrome
and venous thrombosis are commonly noted in COVID-19 patients with severe and
critical illness, contributing to the poor prognosis. Interleukin (IL)-6, a major complex
inammatory cytokine, is an independent factor in predicting the severity of COVID-19
disease in patients. IL-6 and tumor necrosis factor (TNF)-aparticipate in COVID-19-
induced cytokine storm, causing endothelial cell damage and upregulation of
plasminogen activator inhibitor-1 (PAI-1) levels. In addition, IL-6 and PAI-1 form a
vicious cycle of inammation and thrombosis, which may contribute to the poor
prognosis of patients with severe COVID-19. Targeted inhibition of IL-6 and PAI-1
signal transduction appears to improve treatment outcomes in severely and critically ill
COVID-19 patients suffering from cytokine storms and venous thrombosis. Motivated by
studies highlighting the relationship between inammatory cytokines and thrombosis in
viral immunology, we provide an overview of the immunothrombosis and
immunoinammation vicious loop between IL-6 and PAI-1. Our goal is that
understanding this ferocious circle will benet critically ill patients with COVID-
19 worldwide.
Keywords: COVID-19, PAI-1, IL-6, inammatory reaction, venous thrombosis, tocilizumab, endothelial cells
Abbreviations: ACE, Angiotensin-converting enzyme; COVID-19, Coronavirus disease 2019; ECs, Endothelial cells; EGFR,
Epidermal growth factor receptor; HFD, High-fat diet; HPMECs, Human pulmonary microvascular endothelial cells; ICU,
Intensive care unit; IL, Interleukin; IL-6R, Interleukin-6 receptor; JAK, Janus kinase; LPS, Lipopolysaccharide; MD2, Myeloid
differentiation protein 2; NF-kB, Nuclear factor of kappa B; PAI-1, Plasminogen activator inhibitor 1; STAT3, Signal
transducer and activator of transcription 3; TCZ, Tocilizumab; TLR, Toll-like receptors; TNF, Tumor necrosis factor; tPA,
Tissue plasminogen activator; uPA, Urokinase-type plasminogen activator.
Frontiers in Immunology | www.frontiersin.org June 2022 | Volume 13 | Article 9306731
Edited by:
Chang Li,
Chinese Academy of Agricultural
Sciences (CAAS), China
Reviewed by:
Zhanbo Zhu,
Heilongjiang Bayi Agricultural
University, China
Wentao Qiao,
Nankai University, China
*Correspondence:
Fei Guo
guofei@ipb.pumc.edu.cn
Yue Li
liyue2018@tjutcm.edu.cn
Long Yang
long.yang@tjutcm.edu.cn
ORCID:
Yue Li
orcid.org/0000-0001-8198-9911
These authors have contributed
equally to this work and share
rst authorship
Specialty section:
This article was submitted to
Molecular Innate Immunity,
a section of the journal
Frontiers in Immunology
Received: 28 April 2022
Accepted: 12 May 2022
Published: 15 June 2022
Citation:
Huang P, Zuo Q, Li Y, Oduro PK,
Tan F, Wang Y, Liu X, Li J, Wang Q,
Guo F, Li Y and Yang L (2022)
A Vicious Cycle: In Severe and
Critically Ill COVID-19 Patients.
Front. Immunol. 13:930673.
doi: 10.3389/fimmu.2022.930673
REVIEW
published: 15 June 2022
doi: 10.3389/fimmu.2022.930673
INTRODUCTION
Since late December, coronavirus disease 2019 (COVID-19) (1)
has spread worldwide and instigated a pandemic. Globally, as of
April 12, 2022, more than ve hundred million people have been
diagnosed with COVID-19 disease, including more than 6
million deaths from the disease (WHO, https://covid19.who.
int/), posing a great challenge to the health system around the
world. The causative agent of the disease is the SARS-CoV-2
virus. Based on the clinical presentation of the COVID-19
disease, the mild-to-moderate disease accounts for 81% of
COVID-19 infections and is accompanied by symptoms such
as cough, fever, fatigue, and others. Meanwhile, only about 14%
of cases have severe symptoms such as dyspnea and hypoxemia,
while 5% present with respiratory failure, shock failure, multiple
organ failure, and other severe conditions that can result in
death. In addition, 14.8% of patients are classied as severe or
critically ill patients (Table 1)(2). Emerging laboratory and
pathological examination data indicate that cytokine storms and
thrombosis were closely related to the disease progression,
accounting for the poor prognosis in COVID-19 patients (38).
A signicant reduction in spontaneous clot dissolution after
activation of the external clotting pathway and increased resistance
to tissue plasminogen activator (tPA) suggests a potential link
between brinolytic disorder and thrombosis (9). Serum
proteomics studies in patients with COVID-19 have found that
abnormal increases in IL-6 correlate with increases in the
coagulation and complement cascade components (10). PAI-1 is
a serine protease inhibitor that acts as a principal inhibitor of tPA
and urokinase-type plasminogen activator (uPA) to inhibit
brinolysis. Based on PAI-1s primary function, diseases, or
disorders that increase PAI-1 levels appear to result in high
coagulation states (1113). Interestingly, in patients with mild-
to-moderate disease, plasma levels of PAI-1 were normal
compared to critically ill COVID-19 patients (14,15). However,
reports from studies suggested that PAI-1 levels signicantly
increase in critically ill (14) and hospitalized COVID-19 patients
(Figure 1). In addition, previous analyses on the detection of
inammatory and prethrombotic biomarkers in the blood showed
signicant differences between IL-6 and PAI-1 levels. The mean
concentration of IL-6 in the non-severe COVID-19 group was
430.3 pg/ml, whereas that of the control group was 419.5 pg/ml.
Meanwhile, the concentration of IL-6 in severe COVID-19 and
death group was 1,463 and 2,200 pg/ml, respectively (14). PAI-1 is
a widely recognized biomarker of endothelial dysfunction and has
been shown that increased concentration is associated with the
severity of the disease (16,17). The expression of PAI-1 may
reect the severity of SARS-CoV-2 infection to some extent (18).
The plasma concentration of PAI-1 detected in patients with
severe COVID-19 was 713.3 ng/ml, while in the COVID-19
death group, it was 1,223.5 ng/ml. Then again, in the non-severe
COVID-19 group, the plasma concentration of PAI-1 was 465.2
ng/ml and that of healthy donors was 183.7 ng/ml (14). It is
important to note that severe and critically ill patients with
COVID-19oftensufferfromunderlyingdiseases(19,20).
Evidence has also suggested that most of the underlying diseases
present with elevated levels of PAI-1 (21). For example, among
diabetes and acute cerebral infarction patients without COVID-19,
PAI-1 levels averaged 36.5 and 63.95 ng/ml (22,23). Nonetheless,
COVID-19-infected individuals have signicantly higher levels of
PAI-1 than those with diabetes or acute cerebral infarction,
providing indirect evidence that COVID-19 could increase PAI-
1levels(Table 2).
Studies on coexpression-induced IL-6 and PAI-1 through the
nuclear factor-kappa B (NF-kB) pathway and ligand-dependent
epidermal growth factor receptor (EGFR) activation conrmed a
signicant correlation between IL-6 and PAI-1 (26). The same
phenomenon has revealed signicant differences between IL-6
and PAI-1 levels in severe and mild-to-moderate COVID-19
FIGURE 1 | SARS-Co-2 upregulates plasma IL-6, TNF- a, and PAI-1 levels. The levels of IL-6, PAI-1, and TNF-ain the serum of severely and critically ill COVID-19
patients with SARS-CoV-2 pulmonary infection via the respiratory tract were signicantly increased.
TABLE 1 | The distribution of age, degree, and fatality rate of COVID-19 (2).
Categories Subgroup Cases Distribution
Age 80 years 1,408 3%
3079 years 38,680 87%
1029 years 4,168 6%
<10 years 416 1%
Degree Mild 36,160 81%
Severe 6,168 4%
Critically ill 2,087 5%
Fatality rate 44,672 conrmed cases 1,023 2.3%
Aged 80 years 208 14.8%
7079 years 312 8.0%
Critically cases 1,023 49.0%
Bold values highlight the proportion and mortality of critically ill patients and emphasize the
lethality of COVID-19.
Huang et al. COVID-19 Patients Suffer Vicious Cycle
Frontiers in Immunology | www.frontiersin.org June 2022 | Volume 13 | Article 9306732
patients (14). Treatment with anti-TNFs can reduce the death
rate and poor outcomes of COVID-19 patients (27). Below, we
review the possible relationship between inammatory levels and
thrombosis in severe and critically ill COVID-19 patients.
SARS-COV-2 RAISES THE EXPRESSION
OF PAI-1, IL-6, AND TNF-Α
The SARS-CoV-2 infection has a devastating effect on immune
regulation, leading to a life-threatening systemic inammatory
syndrome called the cytokine storm. This systemic inammatory
syndrome involves abnormal immune-cell hyperactivation and
uncontrolled release of circulatory cytokines. Elegant evidence
from the COVID-19 pandemic shows that IL-6 and TNF-aare
involved in the COVID-19-induced cytokine storm (28). In
severe disease, IL-6 and TNF-aare major contributing factors
that worsen the condition and cause poor clinical outcomes and
even death (2931). IL-6 is a multifunctional cytokine capable of
transmitting cell signals. It is the main trigger of endothelial
cytokine storm and an intervention target for clinical therapy
(32,33). Almost all stromal cells and immune system cells can
produce IL-6, and the primary activator is IL-1bor TNF-a(34).
Toll-like receptor (TLR)-stimulated monocytes and
macrophages can also promote the expression of IL-6 (35).
During propagation of the SARS-CoV-2 virus, the envelope
spike glycoprotein of the SARS-CoV-2 virus attaches to the
angiotensin-converting enzyme (ACE)-2 on the target cell
surface, resulting in ACE-2 loss (36). ACE-2 is a negative
regulator that functions by activating tPA. ACE-2 deciency
disrupts the effective ACE-2/angiotensin (17)/Mas receptor
axis, making Ang II more active and decreasing tPA activity,
prompting endothelial and smooth muscle cells to synthesize
and release PAI-1, leading to the balance of PAI-1/tPA to revert
to its prethrombotic state (37,38). Studies on intensive care unit
(ICU) patients with critically ill COVID-19 found that low
brinolysis was mainly associated with elevated PAI-1 levels
(39). The action of recombinant SARS-CoV-2 on the ACE-2
receptor is comparable to that of live viruses, and its spiking
glycoprotein induces the expression of PAI-1 in human
pulmonary microvascular endothelial cells (HPMECs) (40). In
individuals with severe COVID-19 illness, increased PAI-1
expression reduces tPA activity and increases thrombosis while
perhaps worsening the inammatory response (Table 3).
PAI-1 Upregulates the Expressions of IL-6
and TNF-a
In several studies, PAI-1 has been found at the inammatory site
after tissue damage (47,48). PAI-1 inhibitors reduce TNF-a
expression and, at the same time, decrease PAI-1 expression in
diabetic mice (49). PAI-1 upregulation may be related to its
capacity to activate macrophages. PAI-1 helps to regulate the
lipopolysaccharide (LPS)-induced inammatory response in
NR8383 cells, possibly by inuencing the TLR4-myeloid
differentiation protein 2 (MD-2)/NF-kBsignalingtransfer
pathway (50). PAI-1-induced TLR4 activation causes monocyte
macrophages to release signicant quantities of IL-6 and TNF-a,
exacerbating the inammatory response (51,52). This shows that
TLR4 is an essential medium for PAI-1 to activate macrophages
and promote TNF-aexpression. The expression spectrum of
macrophages stimulated by PAI-1 occurs 2 h after the peak
transcription of PAI-1 (53). PAI-1 can promote macrophage
activation and may also be an initial response gene
for predicting inammation. PAI-1 promotes the recruitment
of monocytes/macrophages in tumor cells. Its lipoprotein-
receptor-related protein 1 interaction domain regulates
macrophage migration, whereas its C-terminal uPA interaction
domain auto-secretes IL-6 by activating the p38MAPK and NF-kB
pathway and inducing macrophage polarization (54). There was a
considerable increase in the expression of M1 macrophages in
obese mice caused by a high-fat diet (HFD), but PAI-1 deciency
and PAI-039therapy prevented the development of these markers,
demonstrating that PAI-1 is required for macrophage
polarization. Meanwhile, PAI-1 activates TLR4, triggering a
robust inammatory response in endothelial cells (ECs),
allowing ECs to continuously secrete IL-6 (55). PAI-1 may
interact with TLR4 to activate NF-kB, leading ECs to generate
TABLE 3 | The expressions of PAI-1 and IL-6 in severe COVID-19 patients.
Factors Expressing and working Reference
PAI-1 rSARS-CoV-2-S1 infect HPMECs exhibited robust
induction of PAI-1
(40)
Circulating levels of PAI-1 upregulate and function as an
independent predictor of the severity of COVID-19
disease in patients
(41)
Decreased the PAI-1 levels and alleviated critical illness in
severe COVID-19 patients
(42)
Signicant expression of PAI-1 exists only in severe
COVID-19 patients and promotes patient thrombosis
(14)
Hypercoagulability and hypobrinolysis are connected to
the elevated level of PAI-1 in COVID-19
(39)
IL-6 IL-6 can serve as an independent factor predictor of the
severity of COVID-19 disease in patients
(4346)
Seroproteomics studies found IL-6 signicant
upregulation, and IL-6 signal transduction is the most
upstream upregulation pathway in severe patients with
COVID-19 patients
(10)
IL-6 is the main trigger of endothelial cytokine storms in
COVID-19 patients
(32)
TABLE 2 | The expression of IL-6 and PAI-1 in COVID-19 and underlying diseases.
Disease IL-6 (mean
pg/ml)
PAI-1 (mean
ng/ml)
COVID-19 Healthy donors 419.5 183.7
Non-severe
COVID-19 group
430.3 465.2
Severe COVID-19
group
1463 713.3
Death group 2200 1,223.5
Type 2
diabetes
<20 (24) 36.5
Acute cerebral
infarction
<1,000 (25) 63.95
Huang et al. COVID-19 Patients Suffer Vicious Cycle
Frontiers in Immunology | www.frontiersin.org June 2022 | Volume 13 | Article 9306733
cytokines such as IL-6 (56,57). This shows that PAI-1 can
stimulate macrophages and endothelial cells in various ways,
promoting inammatory responses (Table 4).
There is no clinical use of PAI-1 inhibitors in COVID-19
patients. However, it is worth noting that bortezomib upregulates
KLF2 to suppress PAI-1 expression and reduce EC damage in
HPME cells stimulated with rSARS-CoV-2-S1 glycoprotein (58).
The IL-6 Increases the Expression
of the PAI-1
Severe clotting disorder in patients with COVID-19 is closely
related to the increased risk of death (5962). Venous
thromboembolism was prevalent in COVID-19 patients, with a
total incidence of 31% in 184 patients with severe COVID-19 (63),
and a preliminary autopsy on 11 of the COVID-19 patients revealed
thrombus in the pulmonary arterioles (64). The D-dimer is a brin
degradation product used as an alternative marker of brinolysis
and is often elevated in thrombotic events (65). Relevant studies on
COVID-19 report that D-dimer elevation is a prevalent feature (66).
Low brinolysis is the primary cause of increased blood viscosity
and is associated with elevated PAI-1 levels (39). PAI-1 circulating
levels may be used as an independent predictor of severity in
COVID-19 patients (41), and regulating PAI-1 expression can
benetpatientswithCOVID-19(42).
Alongside PAI-1, IL-6 is an independent predictor of COVID-
19 severity (4346). IL-6 levels have a substantial predictive value
for mortality in COVID-19 ICUs (67). Patients with severe
COVID-19 have considerable IL-6 overexpression, and IL-6
signal transduction is the most upregulated pathway in COVID-
19 patients (10). IL-6 may have a signicant role in the progression
of severe COVID-19 disease in patients. PAI-1 expression is only
found in severe COVID-19 patients and increases thrombosis
(14). PAI-1 is linked to elevated levels of IL-6 in critically ill
COVID-19 patients. IL-6 signals through two central pathways.
The rst is the classic cis signaling, and the second is the trans-
signaling. In the classic cis pathway, IL-6 attaches to cells, mainly
immune cells, expressing the membrane-bound interleukin-6
receptor (IL-6R) to initiate a downstream signaling response (68,
69). On the other hand, in trans-signaling, IL-6 binds to the
soluble form of IL-6R, which is released from IL-6R expressing cell
surfaces by proteolysis and IL-6R mRNA to form an exciting
complex that associates with membrane-bound gp130 (7072). In
the presence of high circulating levels of IL-6, trans-signaling
typically occurs. For instance, ECs express the membrane-bound
gp130 but not the membrane-bound IL-6R (7376), allowing for
IL-6/soluble-IL-6R/gp130 downstream signaling activation.
The detection of PAI-1 expression before and after tocilizumab
(TCZ) treatment demonstrates that IL-6 signaling transduction
can promote PAI-1 expression in ECs (18,42). LPS stimulates the
NF-kB classical pathway to increase the PAI-1 expression and
promote alveolar hypercoagulation and brinolysis inhibitory
states. PAI-1 expression is dramatically reduced following NF-
kB knockout (77,78), indicating that the NF-kB pathway can
control PAI-1 expression to some extent. At the same time,
elevated plasma IL-6 levels promote NF-kB activation (79),
resulting in EC-induced PAI-1 overexpression. In hepatocytes,
IL-6 signals via the Janus kinase (JAK) pathway to promote C/
EBPd-induced PAI-1 expression (80). In addition, IL-6 signals and
activates the IL-6R/signal transducer and activator of transcription
3 (STAT3) pathway (54), which can indirectly upregulate PAI-1
via miR-34a (81). TNF-acan also upregulate PAI-1 (82).
However, it is less commonly documented in the literature, and
the mechanism remains unknown (Table 5).
According to the preceding discussion, elevated and
persistent IL-6, TNF-a, and PAI-1 levels in severe COVID-19
patients potentially generate a vicious cycle of inammatory
response and thrombosis (Figure 2).
CLINICAL SIGNIFICANCE
The probable inammatory response and thrombus interaction
mechanisms are rst described in critically ill COVID-19
patients. TCZ is a recombinant human-resistant human IL-6R
IgG1 monoclonal antibody (83). The use of TCZ in critically ill
COVID-19 patients can decrease PAI-1 levels and improve the
condition of severe COVID-19 patients (42). TCZ is authorized
for the treatment of rheumatoid arthritis (84) and systemic
juvenile idiopathic arthritis (85) because it selectively binds
soluble and membrane-bound IL-6 receptors and inhibits IL-6-
mediated classic cis and trans-signaling (86). IL-6 levels in severe
COVID-19 patients are signicantly higher than in other
patients, prompting several researchers to recommend TCZ to
TABLE 4 | PAI-1 upregulate the expressions of IL-6 and TNF-a.
Targets Cell/host Model Mechanism Reference
PAI-1 upregulates
TNF-ɑ
NR8383 cells Inammatory model induced by LPS TLR4-MD-2/NF-kB signaling transduction pathway (50)
Mouse Type 2 diabetes mellitus PAItrap3 decreases the levels of both PAI-1 and TNF-a(49)
Mouse Systemic inammation model PAI-1 regulates inammatory responses through TLR4
mediated macrophage activation
(53)
PAI-1 upregulates
IL-6
C57 mouse/HT-1080 brosarcoma
cancer cell line
Rag1
/
PAI1
/
/Rag1
/
PAI-1 mice PAI-1 promotes the recruitment and polarization of
macrophages in cancer
(54)
Microvascular (MIC) and
macrovascular (MAC) endothelial
cells (ECs)
Inammatory model induced by LPS PAI-1 was necessary for macrophage polarization (55)
Mice/human aortic endothelial cells
(HAECs)
Endotoxemia of mouse/Inammatory
model induced by LPS
PAI-1 combines with TLR4 to promote NF-kB
activation so that ECs produce chemokines, such as
IL-6
(56,57)
Huang et al. COVID-19 Patients Suffer Vicious Cycle
Frontiers in Immunology | www.frontiersin.org June 2022 | Volume 13 | Article 9306734
inhibit IL-6 signaling in patients with severe COVID-19 to
improve patient symptoms (35,87). According to reports, TCZ
can be used as an alternative therapy for COVID-19 patients who
are at risk of cytokine storms (88). It is advised that in critically ill
patients with elevated IL-6 levels, a repeated dose of TCZ will be
necessary to reduce IL-6 levels signicantly (88). However, TCZ
is ineffective for patients with moderate COVID-19 (89) but can
improve clinical symptoms in severely and critically ill COVID-
19 patients (90). Breathing and bilateral diffuse turbidity
disappear by intravenous TCZ in severe COVID-19 patients
with pneumonia and acute respiratory distress syndrome
(ARDS) (91). Unfortunately, thrombosis in severe COVID-19
patients was not mentioned. PAI-1 inhibition can improve the
level of IL-6 and the damage to ECs. Treatment with TM5614
(PAI-1 inhibitor) eliminates the elevated circulating levels of
PAI-1 and thrombin in plasma produced by particulate matter
(PM) 2.5 (92). Meanwhile, TM5614 signicantly reduces the
elevated level of IL-6 (92). Bortezomib, a proteasomal
degradation inhibitor, enhances KLF2, decreases PAI-1
expression, and reduces EC damage in HPMECs stimulated
with rSARS-CoV-2-S1 glycoprotein (58). PAI-1 may have a
role in prothrombotic events and inammation in COVID-19
patients. This asserts the vicious cycle of PAI-1 and IL-6 in
COVID-19.
CONCLUSION AND
FUTURE PERSPECTIVE
In this review, we briey discussed the possible link between
elevated IL-6 levels and thrombosis in COVID-19 patients. From
non-viral contexts, the link between PAI-1 and IL-6 forms an
inammatorythrombus circuit (42). PAI-1 and IL-6 were not
shown to be strongly connected in COVID-19 case reports,
although autopsy demonstrated substantial damage to ECs (93).
In COVID-19patients, inammation and thrombosis are two of the
FIGURE 2 | Relationship between PAI-1 and IL-6 after SARS-Co-2 infection. SARS-CoV-2 binds to ACE-2 on the target cell surface, resulting in the loss of ACE-2.
ACE-2 is a negative regulator that works by activating tPA. ACE-2 deciency loses the effective ACE-2/angiotensin (17)/Mas receptor axis and increases the level of
Ang1. ACE converts Ang I to Ang II and decreases tPA activity, causing endothelial cells and smooth muscle cells to synthesize and release PAI-1. Ang II binds to
AT1/AT2 to break the balance of PAI-1/tPA to its prethrombotic state. Elevated levels of PAI-1 in severely and critically ill COVID-19 patients may upregulate IL-6
expression through TLR4/NF-kB pathway and activate macrophages to upregulate IL-6 and TNF-aexpression. At the same time, TNF-acan also upregulate PAI-1
expression. IL-6 upregulates the expression of PAI-1 via STAT3/miR-29a.
TABLE 5 | IL-6 and TNF-apromote the expression of PAI-1.
Promote expres-
sion
Cell/host Model Possible mechanism Reference
TNF-aupregulates
PAI-1
Clinic patients Atherosclerosis TNF-ainhibition with iniximab decreases PAI-1 Ag level (82)
IL-6 upregulates
PAI-1
Clinic patients/HUVECs Patients diagnosed with
CRS from sepsis
Tocilizumab treatment decreased the PAI-1 levels and alleviated
critical illness in severe COVID-19 patients
(18,42)
Human hepatoma/primary mouse
hepatocytes
IL-6 induces PAI-1 expression through JAK signaling pathways
converging on C/EBPd
(80)
Human colorectal cancer/breast
cancer/prostate cancer
Rag1
/
PAI1
/
/Rag1
/
PAI-1 mice (54)
IL-6 activates the IL-6/STAT3 pathway and, through miR-34a,
upregulates PAI-1
(54,81)
Huang et al. COVID-19 Patients Suffer Vicious Cycle
Frontiers in Immunology | www.frontiersin.org June 2022 | Volume 13 | Article 9306735
most signicant deleterious responses (94,95). The development of
blood clotsin the heart can be explainedby the distributionof ECs in
the heart and by the above process (96). In critically ill COVID-19
patients, EC dysfunction increases PAI-1 expression (17) and
promotes macrophage recruitment and activation (54). This
raises the amount of IL-6 and TNF-ain the blood, increasing the
odds of a cytokine storm(28). TCZ can decrease IL-6 signal
transduction via IL-6R and soluble IL-6R. TNF-a, on the other
hand, stimulates endothelial PAI-1 production and activates
macrophages, exposing ECs to prominent levels of IL-6 and TNF-
aand causing sustained tissue and organ damage. In thrombosis,
therapeutic use of thrombolytic treatment merely lowers brin
production. Inability to directly suppress PAI-1 expression and
break the vicious cycle between PAI-1 and IL-6 results in serum
PAI-1 and IL-6 buildup, facilitating tissue damage and thrombosis
development. IL-6 trans-signaling has been shown to increase PAI-
1 expression. When IL-6 is coupled with soluble IL-6R and gp130, it
activates the downstream JAK/STAT signal pathway and promotes
the expression of IL-6 and PAI-1 (54,97,98)(Figure 3). STAT3-
dependent transcription inhibition signicantly reduces VEGF-
induced vascular permeability in zebrash, mouse, and human
endothelial cells (99). Increased endothelial cell permeability can
aggravate pulmonary edema and dyspnea in COVID-19 patients
(100). Although the connectionbetween PAI-1 and IL-6 has not yet
been shown,the possibility of a malignantinteraction between PAI-
1 and IL-6 in critically ill COVID-19 patients should not be
overlooked. PAI-1 and IL-6 may produce a vicious cycle in which
their expression is mutually induced, but the mechanism involved
remains unclear. Thrombosis and inammatory responses in
patients with severe COVID-19 are discussed from a new
perspective, which provides innovative ideas for future studies.
AUTHOR CONTRIBUTIONS
All authors have read and approved the manuscript. FG, YL
(11th author), and LY supervised and edited the nal
manuscript with comments from co-authors. PH, QZ, YL
(3rd author), and PO conceptualized and wrote the initial
draft, which was further reviewed and edited by FT, YW, XL,
JL, and QW for intellectual content. All authors provided
crucial revisions in subsequent drafts.
FUNDING
This work was supported by the Tianjin Municipal Education
Commission Scientic Research Project (Natural Science, Grant
No. 2019ZD11 to LY), Science and Technology Program of
Tianjin (21ZYJDJC00070), the National Key Research and
Development Program of China (2019YFC1708803), and
Innovation Team and Talents Cultivation Program of National
Administration of Traditional Chinese Medicine (ZYYCXTD-
C-202203).
ACKNOWLEDGMENTS
We thank LY, YL, and FG for their assistance with
conceptualization and helpful discussion. We are also grateful
to the Tianjin Municipal Education Commission Scientic
Research Project for the funding support.
FIGURE 3 | IL-6 promotes PAI-1 expression via trans signaling. High concentration of IL-6 combined with soluble IL-6R can activate the JAK/STAT3 signal pathway
through gp130 and upregulate the expression of PAI-1 and IL-6. TCZ canreduce the expression of PAI-1 and IL-6 by inhibiting the binding of IL-6 and soluble IL-6R.
Huang et al. COVID-19 Patients Suffer Vicious Cycle
Frontiers in Immunology | www.frontiersin.org June 2022 | Volume 13 | Article 9306736
REFERENCES
1. Li Q, Guan X, Wu P, Wang X, Zhou L, Tong Y, et al. Early Transmission
Dynamics in Wuhan, China, of Novel Coronavirus-Infected Pneumonia.
N Engl J Med (2020) 382(13):1199207. doi: 10.1056/NEJMoa2001316
2. Wu ZY, McGoogan JM. Characteristics of and Important Lessons From the
Coronavirus Disease 2019 (COVID-19) Outbreak in China Summary of a Report
of 72 314 Cases From the Chinese Center for Disease Controland Prevention.Jama-J
Am Med Assoc (2020) 323(13):123942. doi: 10.1001/jama.2020.2648
3. 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(18):170820.
doi: 10.1056/NEJMoa2002032
4. Liu K, Fang YY, Deng Y, Liu W, Wang MF, Ma JP, et al. Clinical Characteristics
of Novel Coronavirus Cases in Tertiary Hospitals in Hubei Province. Chin Med
J (Engl) (2020) 133(9):102531. doi: 10.1097/CM9.0000000000000744
5. Diao B, Wang C, Tan Y, Chen X, Liu Y, Ning L, et al. Reduction and Functional
Exhaustion of T Cells in Patients With Coronavirus Disease 2019 (COVID-19).
Front Immunol (2020) 11:827. doi: 10.3389/mmu.2020.00827
6. Song F, Shi N, Shan F, Zhang Z, Shen J, Lu H, et al. Emerging 2019 Novel
Coronavirus (2019-Ncov) Pneumonia. Radiology (2020) 295(1):2107.
doi: 10.1148/radiol.2020200274
7. Tian S, Xiong Y, Liu H, Niu L, Guo J, Liao M, et al. Pathological Study of the
2019 Novel Coronavirus Disease (COVID-19) Through Postmortem Core
Biopsies. Mod Pathol (2020) 33(6):100714. doi: 10.1038/s41379-020-0536-x
8. Belen-Apak FB, Sarialioglu F. Pulmonary Intravascular Coagulation in
COVID-19: Possible Pathogenesis and Recommendations on Anticoagulant/
Thrombolytic Therapy. J Thromb Thrombolysis (2020) 50(2):27880. doi:
10.1007/s11239-020-02129-0
9. Hammer S, Haberle H, Schlensak C, Bitzer M, Malek NP, Handgretinger R, et al.
Severe SARS-CoV-2 Infection Inhibits Fibrinolysis Leading to Changes in
Viscoelastic Properties of Blood Clot: A Descriptive Study of Fibrinolysis in
COVID-19. Thromb Haemost (2021) 121(11):141726. doi: 10.1055/a-1 400-6034
10. D'Alessandro A, Thomas T, Dzieciatkowska M, Hill RC, Francis RO, Hudson
KE, et al. Serum Proteomics in COVID-19 Patients: Altered Coagulation and
Complement Status as a Function of IL-6 Level. J Proteome Res (2020) 19
(11):441727. doi: 10.1021/acs.jproteome.0c00365
11. Cesari M, Pahor M, Incalzi RA. Plasminogen Activator Inhibitor-1 (PAI-1): A
Key Factor Linking Fibrinolysis and Age-Related Subclinical and Clinical
Conditions. Cardiovasc Ther (2010) 28(5):e7291. doi: 10.1111/j.1755-
5922.2010.00171.x
12. Binder BR, Christ G, Gruber F, Grubic N, Hufnagl P, Krebs M, et al.
Plasminogen Activator Inhibitor 1: Physiological and Pathophysiological
Roles. News Physiol Sci (2002) 17:5661. doi: 10.1152/nips.01369.2001
13. Ghosh AK, Vaughan DE. PAI-1 in Tissue Fibrosis. J Cell Physiol (2012) 227
(2):493507. doi: 10.1002/jcp.22783
14. Lopez-Castaneda S, Garcia-Larragoiti N, Cano-Mendez A, Blancas-Ayala K,
Damian-Vazquez G, Perez-Medina AI, et al. Inammatory and
Prothrombotic Biomarkers Associated With the Severity of COVID-19
Infection. Clin Appl Thromb Hemost (2021) 27:1076029621999099.
doi: 10.1177/1076029621999099
15. Pine AB, Meizlish ML, Goshua G, Chang CH, Zhang H, Bishai J, et al.
Circulating Markers of Angiogenesis and Endotheliopathy in COVID-19.
Pulm Circ (2020) 10(4):2045894020966547. doi: 10.1177/2045894020966547
16. Bernard I, Limonta D, Mahal LK, Hobman TC. Endothelium Infection and
Dysregulation by SARS-CoV-2: Evidence and Caveats in COVID-19. Viruses
(2020) 13(1):29. doi: 10.3390/v13010029
17. Norooznezhad AH, Mansouri K. Endothelial Cell Dysfunction, Coagulation,
and Angiogenesis in Coronavirus Disease 2019 (COVID-19). Microvasc Res
(2021) 137:104188. doi: 10.1016/j.mvr.2021.104188
18. Kellici TF, Pilka ES, Bodkin MJ. Therapeutic Potential of Targeting
Plasminogen Activator Inhibitor-1 in COVID-19. Trends Pharmacol Sci
(2021) 42(6):4313. doi: 10.1016/j.tips.2021.03.006
19. Vogrig A, Gigli GL, Bna C, Morassi M. Stroke in Patients With COVID-19:
Clinical and Neuroimaging Characteristics. Neurosci Lett (2021) 743:135564.
doi: 10.1016/j.neulet.2020.135564
20. Zhou Y, Chi J, Lv W, Wang Y. Obesity and Diabetes as High-Risk Factors for
Severe Coronavirus Disease 2019 (Covid-19). Diabetes Metab Res Rev (2021)
37(2):e3377. doi: 10.1002/dmrr.3377
21. Bayomy O, Rao AD, Garg R, Vaidya A, Kotin AR, Reiber B, et al. Plasminogen
Activator Inhibitor-1 and Pericardial Fat in Individuals With Type 2 Diabetes
Mellitus. Metab Syndr Relat Disord (2017) 15(6):26975. doi: 10.1089/
met.2017.0031
22. Sakurai S, Jojima T, Iijima T, Tomaru T, Usui I, Aso Y. Empagliozin
Decreases the Plasma Concentration of Plasminogen Activator Inhibitor-1
(PAI-1) in Patients With Type 2 Diabetes: Association With Improvement of
Fibrinolysis. J Diabetes Complications (2020) 34(11):107703. doi: 10.1016/
j.jdiacomp.2020.107703
23. Li L, Ren S, Hao X, Zhen Z, Ji H. Efcacy of Minimally Invasive Intervention
in Patients With Acute Cerebral Infarction. J Cardiovasc Pharmacol (2019) 73
(1):226. doi: 10.1097/FJC.0000000000000625
24. Mallard AR, Hollekim-Strand SM, Ingul CB, Coombes JS. High Day-to-Day
and Diurnal Variability of Oxidative Stress and Inammation Biomarkers in
People With Type 2 Diabetes Mellitus and Healthy Individuals. Redox Rep
(2020) 25(1):649. doi: 10.1080/13510002.2020.1795587
25. Liu Y, Qu M, Wang N, Wang L. Effects of an Evidence-Based Nursing
Intervention on Neurological Function and Serum Inammatory Cytokines in
Patients With Acute Cerebral Infarction: A Randomized Controlled Trial.
Restor Neurol Neurosci (2021) 39(2):12937. doi: 10.3233/RNN-201080
26. Alberti C, Pinciroli P, Valeri B, Ferri R, Ditto A, Umezawa K, et al. Ligand-
Dependent EGFR Activation Induces the Co-Expression of IL-6 and PAI-1 via
the NFkB Pathway in Advanced-Stage Epithelial Ovarian Cancer. Oncogene
(2012) 31(37):413949. doi: 10.1038/onc.2011.572
27. Robinson PC, Liew DFL, Liew JW, Monaco C, Richards D, Shivakumar S,
et al. The Potential for Repurposing Anti-TNF as a Therapy for the
Treatment of COVID-19. Med (NY) (2020) 1(1):90102. doi: 10.1016/
j.medj.2020.11.005
28. Azevedo RB, Botelho BG, Hollanda JVG, Ferreira LVL, Junqueira de Andrade
LZ, Oei S, et al. Covid-19 and the Cardiovascular System: A Comprehensive
Review. J Hum Hypertens (2021) 35(1):411. doi: 10.1038/s41371-020-0387-4
29. Li X, Geng M, Peng Y, Meng L, Lu S. Molecular Immune Pathogenesis and
Diagnosis of COVID-19. J Pharm Anal (2020) 10(2):1028. doi: 10.1016/
j.jpha.2020.03.001
30. Godeau D, Petit A, Richard I, Roquelaure Y, Descatha A. Return-To-Work,
Disabilities and Occupational Health in the Age of COVID-19. Scand J Work
Environ Health (2021) 47(5):4089. doi: 10.5271/sjweh.3960
31. 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(1):1632.
doi: 10.1128/MMBR.05015-11
32. Hantoushzadeh S, Norooznezhad AH. Possible Cause of Inammatory Storm
and Septic Shock in Patients Diagnosed With (COVID-19). Arch Med Res
(2020) 51(4):3478. doi: 10.1016/j.arcmed.2020.03.015
33. Schett G, Elewaut D, McInnes IB, Dayer JM, Neurath MF. How Cytokine
Networks Fuel Inammation: Toward a Cytokine-Based Disease Taxonomy.
Nat Med (2013) 19(7):8224. doi: 10.1038/nm.3260
34. Hunter CA, Jones SA. IL-6 as a Keystone Cytokine in Health and Disease. Nat
Immunol (2015) 16(5):44857. doi: 10.1038/ni.3153
35. Zhang C, Wu Z, Li JW, Zhao H, Wang GQ. Cytokine Release Syndrome in
Severe COVID-19: Interleukin-6 Receptor Antagonist Tocilizumab may be
the Key to Reduce Mortality. Int J Antimicrob Agents (2020) 55(5):105954.
doi: 10.1016/j.ijantimicag.2020.105954
36. 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) 181(2):271
80.e278. doi: 10.1016/j.cell.2020.02.052
37. Nishimura H, Tsuji H, Masuda H, Nakagawa K, Nakahara Y, Kitamura H,
et al. Angiotensin II Increases Plasminogen Activator Inhibitor-1 and Tissue
Factor mRNA Expression Without Changing That of Tissue Type
Plasminogen Activator or Tissue Factor Pathway Inhibitor in Cultured Rat
Aortic Endothelial Cells. Thromb Haemost (1997) 77(6):118995. doi:
10.1055/s-0038-1656136
38. van Leeuwen RT, Kol A, Andreotti F, Kluft C, Maseri A, Sperti G. Angiotensin
II Increases Plasminogen Activator Inhibitor Type 1 and Tissue-Type
Plasminogen Activator Messenger RNA in Cultured Rat Aortic Smooth
Muscle Cells. Circulation (1994) 90(1):3628. doi: 10.1161/01.CIR.90.1.362
39. Nougier C, Benoit R, Simon M, Desmurs-Clavel H, Marcotte G, Argaud L,
et al. Hypobrinolytic State and High Thrombin Generation may Play a Major
Huang et al. COVID-19 Patients Suffer Vicious Cycle
Frontiers in Immunology | www.frontiersin.org June 2022 | Volume 13 | Article 9306737
Role in SARS-COV2 Associated Thrombosis. J Thromb Haemost (2020) 18
(9):22159. doi: 10.1111/jth.15016
40. Han M, Pandey D. ZMPSTE24 Regulates SARS-CoV-2 Spike Protein-
Enhanced Expression of Endothelial Plasminogen Activator Inhibitor-1. Am
J Respir Cell Mol Biol (2021) 65(3):3008. doi: 10.1165/rcmb.2020-0544OC
41. Henry BM, Cheruiyot I, Benoit JL, Lippi G, Prohaszka Z, Favaloro EJ, et al.
Circulating Levels of Tissue Plasminogen Activator and Plasminogen
Activator Inhibitor-1 Are Independent Predictors of Coronavirus Disease
2019 Severity: A Prospective, Observational Study. Semin Thromb Hemost
(2021) 47(4):4515. doi: 10.1055/s-0040-1722308
42. Kang S, Tanaka T, Inoue H, Ono C, Hashimoto S, Kioi Y, et al. IL-6 Trans-
Signaling Induces Plasminogen Activator Inhibitor-1 From Vascular
Endothelial Cells in Cytokine Release Syndrome. Proc Natl Acad Sci USA
(2020) 117(36):223516. doi: 10.1073/pnas.2010229117
43. Aziz M, Fatima R, Assaly R. Elevated Interleukin-6 and Severe COVID-19: A
Meta-Analysis. J Med Virol (2020) 92(11):22835. doi: 10.1002/jmv.25948
44. Santa Cruz A, Mendes-Frias A, Oliveira AI, Dias L, Matos AR, Carvalho A,
et al. Interleukin-6 Is a Biomarker for the Development of Fatal Severe Acute
Respiratory Syndrome Coronavirus 2 Pneumonia. Front Immunol (2021)
12:613422. doi: 10.3389/mmu.2021.613422
45. Liu F, Li L, Xu M, Wu J, Luo D, Zhu Y, et al. Prognostic Value of Interleukin-6,
C-Reactive Protein, and Procalcitonin in Patients With COVID-19. J Clin
Virol (2020) 127:104370. doi: 10.1016/j.jcv.2020.104370
46. Henry BM, de Oliveira MHS, Benoit S, Plebani M, Lippi G. Hematologic,
Biochemical and Immune Biomarker Abnormalities Associated With Severe
Illness and Mortality in Coronavirus Disease 2019 (COVID-19): A Meta-
Analysis. Clin Chem Lab Med (2020) 58(7):10218. doi: 10.1515/cclm-2020-
0369
47. Juhan-Vague I, Moerman B, De Cock F, Aillaud MF, Collen D. Plasma Levels
of a Specic Inhibitor of Tissue-Type Plasminogen Activator (and Urokinase)
in Normal and Pathological Conditions. Thromb Res (1984) 33(5):52330.
doi: 10.1016/0049-3848(84)90018-5
48. Kluft C, Verheijen JH, Jie AF, Rijken DC, Preston FE, Sue-Ling HM, et al. The
Postoperative Fibrinolytic Shutdown: A Rapidly Reverting Acute Phase
Pattern for the Fast-Acting Inhibitor of Tissue-Type Plasminogen Activator
After Trauma. Scand J Clin Lab Invest (1985) 45(7):60510. doi: 10.3109/
00365518509155267
49. Tang S, Liu W, Pan X, Liu L, Yang Y, Wang D, et al. Specic Inhibition of
Plasminogen Activator Inhibitor 1 Reduces Blood Glucose Level by Lowering
TNF-A. Life Sci (2020) 246:117404. doi: 10.1016/j.lfs.2020.117404
50. Ren W, Wang Z, Hua F, Zhu L. Plasminogen Activator Inhibitor-1 Regulates
LPS-Induced TLR4/MD-2 Pathway Activation and Inammation in Alveolar
Macrophages. Inammation (2015) 38(1):38493. doi: 10.1007/s10753-014-
0042-8
51. Kawai T, Akira S. Signaling to NF-kappaB by Toll-Like Receptors. Trends Mol
Med (2007) 13(11):4609. doi: 10.1016/j.molmed.2007.09.002
52. Togbe D, Schnyder-Candrian S, Schnyder B, Doz E, Noulin N, Janot L, et al.
Toll-Like Receptor and Tumour Necrosis Factor Dependent Endotoxin-
Induced Acute Lung Injury. Int J Exp Pathol (2007) 88(6):38791.
doi: 10.1111/j.1365-2613.2007.00566.x
53. Gupta KK, Xu Z, Castellino FJ, Ploplis VA. Plasminogen Activator Inhibitor-1
Stimulates Macrophage Activation Through Toll-Like Receptor-4. Biochem
Biophys Res Commun (2016) 477(3):5038. doi: 10.1016/j.bbrc.2016.06.065
54. Kubala MH, Punj V, Placencio-Hickok VR, Fang H, Fernandez GE, Sposto R,
et al. Plasminogen Activator Inhibitor-1 Promotes the Recruitment and
Polarization of Macrophages in Cancer. Cell Rep (2018) 25(8):2177
91.e2177. doi: 10.1016/j.celrep.2018.10.082
55. Lu Z, Li Y, Jin J, Zhang X, Lopes-Virella MF, Huang Y. Toll-Like Receptor 4
Activation in Microvascular Endothelial Cells Triggers a Robust
Inammatory Response and Cross Talk With Mononuclear Cells via
Interleukin-6. Arterioscler Thromb Vasc Biol (2012) 32(7):1696706.
doi: 10.1161/ATVBAHA.112.251181
56. Obi AT, Andraska E, Kanthi Y, Kessinger CW, Eline M, Luke C, et al.
Endotoxaemia-Augmented Murine Venous Thrombosis Is Dependent on
TLR-4 and ICAM-1, and Potentiated by Neutropenia. Thromb Haemost
(2017) 117(2):33948. doi: 10.1160/TH16-03-0218
57. Dasu MR, Devaraj S, Du Clos TW, Jialal I. The Biological Effects of CRP Are
Not Attributable to Endotoxin Contamination: Evidence From TLR4
Knockdown Human Aortic Endothelial Cells. J Lipid Res (2007) 48(3):509
12. doi: 10.1194/jlr.C600020-JLR200
58. Han M, Pandey D. ZMPSTE24 Regulates SARS-CoV-2 Spike Protein-
Enhanced Expression of Endothelial PAI-1. Am J Respir Cell Mol Biol
(2021) 65(3):3008. doi: 10.1165/rcmb.2020-0544OC
59. 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(4):8447. doi: 10.1111/jth.14768
60. Chen N, Zhou M, Dong X, Qu J, Gong F, Han Y, et al. Epidemiological and
Clinical Characteristics of 99 Cases of 2019 Novel Coronavirus Pneumonia in
Wuhan, China: A Descriptive Study. Lancet (2020) 395(10223):50713.
doi: 10.1016/S0140-6736(20)30211-7
61. 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(10223):497506. doi: 10.1016/S0140-6736(20)30183-5
62. Fogarty H, Townsend L, Ni Cheallaigh C, Bergin C, Martin-Loeches I, Browne
P, et al. More on COVID-19 Coagulopathy in Caucasian Patients. Br
J Haematol (2020) 189(6):10601. doi: 10.1111/bjh.16791
63. Klok FA, Kruip M,van der Meer NJM, Arbous MS, GommersD, Kant KM, et al.
Incidence of Thrombotic Complications in Critically Ill ICU Patients With
COVID-19. Thromb Res (2020) 191:1457. doi: 10.1016/j.thromres.2020.04.013
64. Lax SF, Skok K, Zechner P, Kessler HH, Kaufmann N, Koelblinger C, et al.
Pulmonary Arterial Thrombosis in COVID-19 With Fatal Outcome : Results
From a Prospective, Single-Center, Clinicopathologic Case Series. Ann Intern
Med (2020) 173(5):35061. doi: 10.7326/M20-2566
65. Ordieres-Ortega L, Demelo-Rodriguez P, Galeano-Valle F, Kremers BMM,
Ten Cate-Hoek AJ, Ten Cate H. Predictive Value of D-Dimer Testing for the
Diagnosis of Venous Thrombosis in Unusual Locations: A Systematic Review.
Thromb Res (2020) 189:512. doi: 10.1016/j.thromres.2020.02.009
66. Lippi G, Plebani M, Henry BM. Thrombocytopenia is Associated With Severe
Coronavirus Disease 2019 (COVID-19) Infections: A Meta-Analysis. Clin
Chim Acta (2020) 506:1458. doi: 10.1016/j.cca.2020.03.022
67. Gorham J, Moreau A, Corazza F, Peluso L, Ponthieux F, Talamonti M, et al.
Interleukine-6 in Critically Ill COVID-19 Patients: A Retrospective Analysis.
PLos One (2020) 15(12):e0244628. doi: 10.1371/journal.pone.0244628
68. Jones SA, Scheller J, Rose-John S. Therapeutic Strategies for the Clinical
Blockade of IL-6/Gp130 Signaling. J Clin Invest (2011) 121(9):337583.
doi: 10.1172/JCI57158
69. Tanaka Y, Martin Mola E. IL-6 Targeting Compared to TNF Targeting in
Rheumatoid Arthritis: Studies of Olokizumab, Sarilumab and Sirukumab. Ann
Rheum Dis (2014) 73(9):15957. doi: 10.1136/annrheumdis-2013-205002
70. Rose-John S, Heinrich PC. Soluble Receptors for Cytokines and Growth
Factors: Generation and Biological Function. Biochem J (1994) 300(Pt 2):281
90. doi: 10.1042/bj3000281
71. Peters M, Jacobs S, Ehlers M, Vollmer P, Mullberg J, Wolf E, et al. The
Function of the Soluble Interleukin 6 (IL-6) Receptor In Vivo: Sensitization of
Human Soluble IL-6 Receptor Transgenic Mice Towards IL-6 and
Prolongation of the Plasma Half-Life of IL-6. JExpMed(1996) 183
(4):1399406. doi: 10.1084/jem.183.4.1399
72. Schobitz B, Pezeshki G, Pohl T, Hemmann U, Heinrich PC, Holsboer F, et al.
Soluble Interleukin-6 (IL-6) Receptor Augments Central Effects of IL-6
In Vivo.FASEB J (1995) 9(8):65964. doi: 10.1096/fasebj.9.8.7768358
73. Yoshida K, Taga T, Saito M, Suematsu S, Kumanogoh A, Tanaka T, et al.
Targeted Disruption of Gp130, a Common Signal Transducer for the
Interleukin 6 Family of Cytokines, Leads to Myocardial and Hematological
Disorders. Proc Natl Acad Sci USA (1996) 93(1):40711. doi: 10.1073/
pnas.93.1.407
74. Kopf M, Baumann H, Freer G, Freudenberg M, Lamers M, Kishimoto T, et al.
Impaired Immune and Acute-Phase Responses in Interleukin-6-Decient
Mice. Nature (1994) 368(6469):33942. doi: 10.1038/368339a0
75. Jones GW, McLoughlin RM, Hammond VJ, Parker CR, Williams JD,
Malhotra R, et al. Loss of CD4+ T Cell IL-6R Expression During
Inammation Underlines a Role for IL-6 Trans Signaling in the Local
Maintenance of Th17 Cells. J Immunol (2010) 184(4):21309. doi: 10.4049/
jimmunol.0901528
76. McElvaney OJ, Curley GF, Rose-John S, McElvaney NG. Interleukin-6:
Obstacles to Targeting a Complex Cytokine in Critical Illness. Lancet Respir
Med (2021) 9(6):64354. doi: 10.1016/S2213-2600(21)00103-X
Huang et al. COVID-19 Patients Suffer Vicious Cycle
Frontiers in Immunology | www.frontiersin.org June 2022 | Volume 13 | Article 9306738
77. Wu Y, Wang Y, Liu B, Cheng Y, Qian H, Yang H, et al. SN50 Attenuates
Alveolar Hypercoagulation and Fibrinolysis Inhibition in Acute Respiratory
Distress Syndrome Mice Through Inhibiting NF-kappaB P65 Translocation.
Respir Res (2020) 21(1):130. doi: 10.1186/s12931-020-01372-6
78. Katz JM, Tadi P. Physiology, Plasminogen Activation. Treasure Island
(FL: StatPearls (2021).
79. Li W, Sun L, Lei J, Wu Z, Ma Q, Wang Z. Curcumin Inhibits Pancreatic
Cancer Cell Invasion and EMT by Interfering With Tumorstromal Crosstalk
Under Hypoxic Conditions via the IL6/ERK/NFkappaB Axis. Oncol Rep
(2020) 44(1):38292. doi: 10.3892/or.2020.7600
80. Dong J, Fujii S, Imagawa S, Matsumoto S, Matsushita M, Todo S, et al. IL-1
and IL-6 Induce Hepatocyte Plasminogen Activator Inhibitor-1 Expression
Through Independent Signaling Pathways Converging on C/EBPdelta. Am
J Physiol Cell Physiol (2007) 292(1):C209215. doi: 10.1152/ajpcell.00157.2006
81. Rokavec M, Oner MG,Li H, Jackstadt R, Jiang L, Lodygin D, et al. IL-6r/STAT3/
miR-34a Feedback Loop Promotes EMT-Mediated Colorectal Cancer Invasion
and Metastasis. JClinInvest(2014) 124(4):185367. doi: 10.1172/JCI73531
82. Cigolini M, et al. Expression of Plasminogen Activator Inhibitor-1 in Human
AdiposeTissue: A Role for TNF-Alpha?.Atherosclerosis(1999) 143(1):8190.doi:
10.1016/s0021-9150(98)00281-0
83. Alves JD, Marinho A, Serra MJ. Tocilizumab: Is There Life Beyond Anti-TNF
Blockade? Int J Clin Pract (2011) 65(4):50813. doi: 10.1111/j.1742-
1241.2010.02612.x
84. Kaly L, Rosner I. Tocilizumab - A Novel Therapy for non-Organ-Specic
Autoimmune Diseases. Best Pract Res Clin Rheumatol (2012) 26(1):15765.
doi: 10.1016/j.berh.2012.01.001
85. Yokota S, Miyamae T, Imagawa T, Iwata N, Katakura S, Mori M, et al.
Therapeutic Efcacy of Humanized Recombinant Anti-Interleukin-6
Receptor Antibody in Children With Systemic-Onset Juvenile Idiopathic
Arthritis. Arthritis Rheum (2005) 52(3):81825. doi: 10.1002/art.20944
86. Le RQ, Li L, Yuan W, Shord SS, Nie L, Habtemariam BA, et al. FDA Approval
Summary: Tocilizumab for Treatment of Chimeric Antigen Receptor T Cell-
Induced Severe or Life-Threatening Cytokine Release Syndrome. Oncologist
(2018) 23(8):9437. doi: 10.1634/theoncologist.2018-0028
87. Magro G. SARS-CoV-2 and COVID-19: Is Interleukin-6 (IL-6) the 'Culprit
Lesion' of ARDS Onset? What is There Besides Tocilizumab? SGP130Fc.
Cytokine X (2020) 2(2):100029. doi: 10.1016/j.cytox.2020.100029
88. Luo P, Liu Y, Qiu L, Liu X, Liu D, Li J. TocilizumabTreatment in COVID-19: A
SingleCenter Experience. JMedVirol(2020) 92(7):8148.doi:10.1002/jmv.25801
89. Stone JH, Frigault MJ, Serling-Boyd NJ, Fernandes AD, Harvey L, Foulkes AS,
et al. Efcacy of Tocilizumab in Patients Hospitalized With Covid-19. N Engl
J Med (2020) 383(24):233344. doi: 10.1056/NEJMoa2028836
90. 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 USA (2020) 117
(20):109705. doi: 10.1073/pnas.2005615117
91. Toniati P, Piva S, Cattalini M, Garrafa E, Regola F, Castelli F, et al.
Tocilizumab for the Treatment of Severe COVID-19 Pneumonia With
Hyperinammatory Syndrome and Acute Respiratory Failure: A Single
Center Study of 100 Patients in Brescia, Italy. Autoimmun Rev (2020) 19
(7):102568. doi: 10.1016/j.autrev.2020.102568
92. Ghosh AK, Soberanes S, Lux E, Shang M, Aillon RP, Eren M, et al.
Pharmacological Inhibition of PAI-1 Alleviates Cardiopulmonary
Pathologies Induced by Exposure to Air Pollutants PM2.5. Environ Pollut
(2021) 287:117283. doi: 10.1016/j.envpol.2021.117283
93. Ackermann M, Verleden SE, Kuehnel M, Haverich A, Welte T, Laenger F,
et al. Pulmonary Vascular Endothelialitis, Thrombosis, and Angiogenesis in
Covid-19. N Engl J Med (2020) 383(2):1208. doi: 10.1056/NEJMoa2015432
94. Gao YD, Ding M, Dong X, Zhang JJ, Kursat Azkur A, Azkur D, et al. Risk
Factors for Severe and Critically Ill COVID-19 Patients: A Review. Allergy
(2021) 76(2):42855. doi: 10.1111/all.14657
95. Chan NC, Weitz JI. COVID-19 Coagulopathy, Thrombosis, and Bleeding.
Blood (2020) 136(4):3813. doi: 10.1182/blood.2020007335
96. Topol EJ. COVID-19 can Affect the Heart. Science (2020) 370(6515):4089.
doi: 10.1126/science.abe2813
97. Moore JB, June CH. Cytokine Release Syndrome in Severe COVID-19. Science
(2020) 368(6490):4734. doi: 10.1126/science.abb8925
98. Matsuyama T, Kubli SP, Yoshinaga SK, Pfeffer K, Mak TW. An Aberrant
STAT Pathway Is Central to COVID-19. Cell Death Differ (2020) 27
(12):320925. doi: 10.1038/s41418-020-00633-7
99. Wang L, Astone M, Alam SK, Zhu Z, Pei W, Frank DA, et al. Suppressing
STAT3 Activity Protects the Endothelial Barrier From VEGF-Mediated
Vascular Permeability. bioRxiv (2020) 14(11):dmm049029. doi: 10.1101/
2020.10.27.358374
100. Barbosa LC, Goncalves TL, de Araujo LP, Rosario LVO, Ferrer VP.
Endothelial Cells and SARS-CoV-2: An Intimate Relationship. Vascul
Pharmacol (2021) 137:106829. doi: 10.1016/j.vph.2021.106829
Conict of Interest: The authors declare that the research was conducted in the
absence of any commercial or nancial relationships that could be construed as a
potential conict of interest.
Publishers Note: All claims expressed in this article are solely those of the authors
and do not necessarily represent those of their afliated organizations, or those of
the publisher, the editors and the reviewers. Any product that may be evaluated in
this article, or claim that may be made by its manufacturer, is not guaranteed or
endorsed by the publisher.
Copyright © 2022 Huang, Zuo, Li, Oduro, Tan, Wang, Liu, Li, Wang, Guo, Li and
Yang. This is an open-access article distributed under the terms of the Creative
Commons Attribution License (CC BY). The use, distribution or reproduction in other
forums is permitted, provided the original author(s) and the copyright owner(s) are
credited and that the original publication in this journal is cited, in accordance with
accepted academic practice. No use, distribution or reproduction is permitted which
does not comply with these terms.
Huang et al. COVID-19 Patients Suffer Vicious Cycle
Frontiers in Immunology | www.frontiersin.org June 2022 | Volume 13 | Article 9306739
... IL-6 [37,[49][50][51][52][53][54] and TNF-α [37,51,[53][54][55] are two critical components of cytokine storm. Since plasma levels of TNF-α [34,56,57] and IL-6 [33,58,59] are at least partly determined by variants of rs1800629 [34,56,57] and rs1800795 [33,58,59], it indicates that variants of rs1800629 and rs1800795 may impact COVID-19 outcomes by modulating cytokine storm. ...
... IL-6 [37,[49][50][51][52][53][54] and TNF-α [37,51,[53][54][55] are two critical components of cytokine storm. Since plasma levels of TNF-α [34,56,57] and IL-6 [33,58,59] are at least partly determined by variants of rs1800629 [34,56,57] and rs1800795 [33,58,59], it indicates that variants of rs1800629 and rs1800795 may impact COVID-19 outcomes by modulating cytokine storm. ...
... (1) By inducing a cytokine storm. IL-6 and TNF-α are critical components of cytokine storm [37,[49][50][51][52][53][54][55]. The elevated TNF-α and IL-6 levels associated with pro-inflammatory cytokines variants (Tables 1, 2) may be helpful to the formation of cytokine storm, thus deteriorating COVID-19 outcomes (Tables 1, 2). ...
Article
Full-text available
Background Cytokine storm is known to impact the prognosis of coronavirus disease 2019 (COVID-19), since pro-inflammatory cytokine variants are associated with cytokine storm. It is tempting to speculate that pro-inflammatory cytokines variants may impact COVID-19 outcomes by modulating cytokine storm. Here, we verified this hypothesis via a comprehensive analysis. Methods PubMed, Cochrane Library, Central, CINAHL, and ClinicalTrials.gov were searched until December 15, 2023. Case–control or cohort studies that investigated the impacts of rs1800795 or rs1800629 on COVID-19 susceptibility, severity, mortality, IL-6, TNF-α, or CRP levels were included after an anonymous review by two independent reviewers and consultations of disagreement by a third independent reviewer. Results 47 studies (8305 COVID-19 individuals and 17,846 non-COVID-19 individuals) were analyzed. The rs1800629 A allele (adenine at the −308 position of the promoter was encoded by the A allele) was associated with higher levels of tumor necrosis factor-α (TNF-α) and C-reactive protein (CRP). In contrast, the rs1800795 C allele (cytosine at the −174 position of the promoter was encoded by the C allele) was linked to higher levels of interleukin-6 (IL-6) and CRP. In addition, the A allele of rs1800629 increased the severity and mortality of COVID-19. However, the C allele of rs1800795 only increased COVID-19 susceptibility. Conclusions rs1800629 and rs1800795 variants of pro-inflammatory cytokines have significant impacts on systemic inflammatory profile and COVID-19 clinical outcomes. rs1800629 may serve as a genetic marker for severe COVID-19.
... There is a growing body of evidence in support of the important role that T-cell responses in protection against COVID-19, as recently reviewed by Wherry and Barouch (11): (i) cross-reactive poly-antigenic CD4 + and CD8 + T-cell responses in COVID-19 patients appear to contribute to the resolution of SARS-CoV-2 infection and reduction in severe symptoms (12)(13)(14)(15)(16)(17)(18)(19)(20)(21)(22); (ii) SARS-CoV-2-specific CD4 + and CD8 + T-cell responses reduced viral loads in non-human primates (23,24); (iii) SARS-CoV-2-infected patients with agammaglobulinemia and B-cell depletion displayed only a small increase in COVID-19 symptoms, indicating that the cross-reactive T cells alone may have protected from severe disease (25)(26)(27)(28)(29)(30); and (iv) cancer patients with B-cell deficiencies experience milder COVID-19 disease that correlated with strong SARS-CoV-2specific CD8 + T-cell responses (22). Conversely, other reports have associated cross-reactive memory CD4 + and CD8 + T cells with poor COVID-19 disease outcomes (16,(31)(32)(33)(34)(35)(36). However, the antigen specificity, frequency, phenotype, and function of cross-reactive memory CD4 + and CD8 + T cells that protect against the severity of COVID-19 in unvaccinated asymptomatic patients remain to be determined. ...
Article
Full-text available
Background Cross-reactive SARS-CoV-2-specific memory CD4⁺ and CD8⁺ T cells are present in up to 50% of unexposed, pre-pandemic, healthy individuals (UPPHIs). However, the characteristics of cross-reactive memory CD4⁺ and CD8⁺ T cells associated with subsequent protection of asymptomatic coronavirus disease 2019 (COVID-19) patients (i.e., unvaccinated individuals who never develop any COVID-19 symptoms despite being infected with SARS-CoV-2) remains to be fully elucidated. Methods This study compares the antigen specificity, frequency, phenotype, and function of cross-reactive memory CD4⁺ and CD8⁺ T cells between common cold coronaviruses (CCCs) and SARS-CoV-2. T-cell responses against genome-wide conserved epitopes were studied early in the disease course in a cohort of 147 unvaccinated COVID-19 patients who were divided into six groups based on the severity of their symptoms. Results Compared to severely ill COVID-19 patients and patients with fatal COVID-19 outcomes, the asymptomatic COVID-19 patients displayed significantly: (i) higher rates of co-infection with the 229E alpha species of CCCs (α-CCC-229E); (ii) higher frequencies of cross-reactive functional CD134⁺CD137⁺CD4⁺ and CD134⁺CD137⁺CD8⁺ T cells that cross-recognized conserved epitopes from α-CCCs and SARS-CoV-2 structural, non-structural, and accessory proteins; and (iii) lower frequencies of CCCs/SARS-CoV-2 cross-reactive exhausted PD-1⁺TIM3⁺TIGIT⁺CTLA4⁺CD4⁺ and PD-1⁺TIM3⁺TIGIT⁺CTLA4⁺CD8⁺ T cells, detected both ex vivo and in vitro. Conclusions These findings (i) support a crucial role of functional, poly-antigenic α-CCCs/SARS-CoV-2 cross-reactive memory CD4⁺ and CD8⁺ T cells, induced following previous CCCs seasonal exposures, in protection against subsequent severe COVID-19 disease and (ii) provide critical insights into developing broadly protective, multi-antigen, CD4⁺, and CD8⁺ T-cell-based, universal pan-Coronavirus vaccines capable of conferring cross-species protection.
... Targeted inhibition of IL-6 and PAI-1 signal pathways shows promise in improving outcomes for critically ill patients facing cytokine storms and venous thrombosis. The overview emphasizes the interconnected relationship between inflammatory cytokines and thrombosis in COVID-19, aiming to enhance the understanding of this vicious cycle for the benefit of critically ill patients worldwide[3].Lab research revealed that the spike protein of SARS-CoV-2 has the ability to directly cause damage to cells and induce electrophysiological dysfunction in hiPSC-CMs. This finding suggests a potential intrinsic and mechanistic susceptibility, contributing to the heightened risk of sudden cardiac death (SCD) observed in COVID-19 patients. ...
Article
Full-text available
The COVID-19 pandemic, caused by the novel coronavirus SARS-CoV-2, has emerged as a global health crisis with far-reaching implications. Beyond the respiratory manifestations commonly associated with the virus, emerging evidence suggests a significant impact on the cardiovascular system. This comprehensive literature review aims to summarize and analyze current knowledge regarding the effects of SARS-CoV-2 on the cardiovascular system, encompassing various aspects from direct cardiac involvement to long-term cardiovascular consequences.
... Besides directly infected by SARS-CoV-2, the endothelial cells also undergo injury by systemic inflammation caused by over-activation of innate immune response, referring to "cytokine storm"(Hu et al., 2021;, 92 Huang et al., 2020). Severe COVID-19-induced cytokine storm (such as IL-6, IL-1β, TNF-α, MCP-1, etc) is a good predictor of the severity of COVID-19, which also aggravates multi-organ injury by propagating the vicious cycle of endothelial cells damage, inflammation and thrombosis(Huang et al., 2022).The current study reported increased cytokines in COVID-19 patients with TMA than in TMA negative COVID-19 patients. Varga et al., (2020) reported evidence of direct SARS-CoV-2 infection of endothelial cells in several organs and diffuse endothelial inflammation associated with apoptosis in COVID-19 patients in Switzerland. ...
Article
Full-text available
Severe COVID-19 can result in multiorgan dysfunction, with the lungs being the most commonly affected and prominent organ. Recent studies suggest that an exaggerated immune response characterized by a cytokine storm may play a crucial role in the extensive organ damage observed in this disease. Additionally, COVID-19 patients often exhibit hypercoagulability, with a high incidence of thrombosis and a higher-than-expected failure rate of anticoagulation therapy. While macrovascular thrombosis is frequently observed, the presence of extensive microvascular thromboses, as reported in several case series and studies, raises the possibility of Thrombotic Microangiopathy (TMA) contributing to the thrombotic and multiorgan complications associated with COVID-19. Identifying TMA promptly and addressing the underlying pathophysiology may potentially improve outcomes for critically ill patients.
... negative in patients at this time, the virus continues to replicate in vivo (29). Among them, in severe COVID-19 patients requiring intensive care unit (ICU) admission, the condition may be further exacerbated by massive inflammatory cell infiltration and high levels of proinflammatory cytokines and chemokines (38, 39), which in severe cases can progress to acute lung injury, acute respiratory distress syndrome (ARDS), or multiple organ failure (40). Respiratory failure due to ARDS is the leading cause of patient mortality (41). ...
Article
Full-text available
The corona virus disease 2019 (COVID-19) global pandemic has had an unprecedented and persistent impact on oncological practice, especially for patients with lung cancer, who are more vulnerable to the virus than the normal population. Indeed, the onset, progression, and prognosis of the two diseases may in some cases influence each other, and inflammation is an important link between them. The original chronic inflammatory environment of lung cancer patients may increase the risk of infection with COVID-19 and exacerbate secondary damage. Meanwhile, the acute inflammation caused by COVID-19 may induce tumour progression or cause immune activation. In this article, from the perspective of the immune microenvironment, the pathophysiological changes in the lungs and whole body of these special patients will be summarised and analysed to explore the possible immunological storm, immunosuppression, and immune escape phenomenon caused by chronic inflammation complicated by acute inflammation. The effects of COVID-19 on immune cells, inflammatory factors, chemokines, and related target proteins in the immune microenvironment of tumours are also discussed, as well as the potential role of the COVID-19 vaccine and immune checkpoint inhibitors in this setting. Finally, we provide recommendations for the treatment of lung cancer combined with COVID-19 in this special group.
... PAI-1 is a driver of hypofibrinolysis in COVID-19 (42). Interestingly, PAI-1 has been suggested to regulate and be regulated by inflammatory biomarkers, namely IL-6, thus linking thrombotic and inflammatory processes (43,44). A large reservoir of PAI-1 resides in platelets themselves, and active PAI-1 is displayed on the platelet membrane on activation (45)(46)(47). ...
Article
Full-text available
Introduction Thromboinflammatory complications are well described sequalae of Coronavirus Disease 2019 (COVID-19), and there is evidence of both hyperreactive platelet and inflammatory neutrophil biology that contributes to the thromoinflammatory milieu. It has been demonstrated in other thromboinflammatory diseases that the circulating environment may affect cellular behavior, but what role this environment exerts on platelets and neutrophils in COVID-19 remains unknown. We tested the hypotheses that 1) plasma from COVID-19 patients can induce a prothrombotic platelet functional phenotype, and 2) contents released from platelets (platelet releasate) from COVID-19 patients can induce a proinflammatory neutrophil phenotype. Methods We treated platelets with COVID-19 patient and disease control plasma, and measured their aggregation response to collagen and adhesion in a microfluidic parallel plate flow chamber coated with collagen and thromboplastin. We exposed healthy neutrophils to platelet releasate from COVID-19 patients and disease controls and measured neutrophil extracellular trap formation and performed RNA sequencing. Results We found that COVID-19 patient plasma promoted auto-aggregation, thereby reducing response to further stimulation ex-vivo. Neither disease condition increased the number of platelets adhered to a collagen and thromboplastin coated parallel plate flow chamber, but both markedly reduced platelet size. COVID-19 patient platelet releasate increased myeloperoxidasedeoxyribonucleic acid complexes and induced changes to neutrophil gene expression. Discussion Together these results suggest aspects of the soluble environment circulating platelets, and that the contents released from those neutrophil behavior independent of direct cellular contact.
... Cardiovascular diseases [86][87][88] Metabolic disorders [89,90] Fibrosis [91,92] Cancer [93][94][95] Inflammation and infectious diseases [96][97][98] ...
Article
Full-text available
Stressful events trigger a set of complex biological responses which follow a bell-shaped pattern. Low-stress conditions have been shown to elicit beneficial effects, notably on synaptic plasticity together with an increase in cognitive processes. In contrast, overly intense stress can have deleterious behavioral effects leading to several stress-related pathologies such as anxiety, depression, substance use, obsessive-compulsive and stressor- and trauma-related disorders (e.g., post-traumatic stress disorder or PTSD in the case of traumatic events). Over a number of years, we have demonstrated that in response to stress, glucocorticoid hormones (GCs) in the hippocampus mediate a molecular shift in the balance between the expression of the tissue plasminogen activator (tPA) and its own inhibitor plasminogen activator inhibitor-1 (PAI-1) proteins. Interestingly, a shift in favor of PAI-1 was responsible for PTSD-like memory induction. In this review, after describing the biological system involving GCs, we highlight the key role of tPA/PAI-1 imbalance observed in preclinical and clinical studies associated with the emergence of stress-related pathological conditions. Thus, tPA/PAI-1 protein levels could be predictive biomarkers of the subsequent onset of stress-related disorders, and pharmacological modulation of their activity could be a potential new therapeutic approach for these debilitating conditions.
... This is chiefly attributed to the fast-evolving nature of the virus. 1 Until recently, five major variants (Alpha, Beta, Gamma, Delta, and Omicron) have been identified, 2 and more are expected in the upcoming years. There is always tremendous need to identify clinical and biochemical factors related to covid-19 severity and prognosis. ...
Article
Full-text available
Background and Aim Vitamin D is a hormone with essential roles in both cellular metabolism and immunity. It controls calcium homeostasis and modulates innate and adaptive immune system responses. Many studies suggested an association between vitamin D deficiency and clinical outcomes of covid-19 infection, while others failed to document such a relation. The present study aimed to evaluate the clinical and prognostic significance of baseline vitamin D levels in hospitalized Egyptian covid-19 patients. Patients and Methods The present retrospective study included 300 hospitalized covid-19 patients. Patients were submitted to standard clinical, laboratory, and radiological assessment. According to vitamin D levels, patients were classified to have normal levels (≥30), insufficient levels (20–29) or deficient levels (<20). Results According to their vitamin D levels, patients were classified into those with normal vitamin D (n=135), others with vitamin D insufficiency (n=114), and a third group with vitamin D deficiency (n=51). Patients with normal vitamin D levels and vitamin D insufficiency are significantly younger [median (IQR): 49.0 (39.0–57.0) versus 51.0 (40.0–61.0) and 55.0 (43.0–62.0) years, respectively, p=0.012] and had less frequency of severe disease (24.4% versus 40.4% and 51.0%, respectively) when compared with those with vitamin D deficiency. Moreover, they had significantly lower levels of D dimer [median (IQR): 1.5 (0.9–2.5) versus 1.8 (0.9–3.1) and 2.0 (1.0–3.2)], CRP [median (IQR): 58.0 (30.0–120.0) versus 76.0 (42.5–160.0) and 105.0 (74.0–208.0), respectively, p<0.001], ferritin [median (IQR): 458.0 (240.0–759.0) versus 606.0 (433.8–897.8) and 820.0 (552.0–1087.0), respectively, p<0.001], and procalcitonin [median (IQR): 290.0 (152.0–394.0) versus 372.5 (227.0–530.5) and 443.0 (272.0–575.0), respectively, p<0.001]. Only lower vitamin D levels were significant predictors of mortality in multivariate analysis [OR (95% CI): 0.88 (0.84–0.92), p<0.001]. Conclusion Low vitamin D levels are related to exaggerated inflammatory response, disease severity, and poor clinical outcome in hospitalized covid-19 patients.
Article
Inflammation contributes to the development of thrombosis, but the mechanistic basis for this association remains poorly understood. Innate immune responses and coagulation pathways are activated in parallel following infection or injury, and represent an important host defense mechanism to limit pathogen spread in the bloodstream. However, dysregulated proinflammatory activity is implicated in the progression of venous thromboembolism and arterial thrombosis. In this review, we focus on the role of myeloid cells in propagating thromboinflammation in acute inflammatory conditions, such as sepsis and coronavirus disease 2019 (COVID-19), and chronic inflammatory conditions, such as obesity, atherosclerosis, and inflammatory bowel disease. Myeloid cells are considered key drivers of thromboinflammation via upregulated tissue factor activity, formation of neutrophil extracellular traps (NETs), contact pathway activation, and aberrant coagulation factor–mediated protease-activated receptor (PAR) signaling. We discuss how strategies to target the intersection between myeloid cell–mediated inflammation and activation of blood coagulation represent an exciting new approach to combat immunothrombosis. Specifically, repurposed anti-inflammatory drugs, immunometabolic regulators, and NETosis inhibitors present opportunities that have the potential to dampen immunothrombotic activity without interfering with hemostasis. Such therapies could have far-reaching benefits for patient care across many thromboinflammatory conditions.
Article
Full-text available
(1) Background: The COVID-19 pandemic left many intriguing mysteries. Retrospective vulnerability trends tie as strongly to odd demographics as to exposure profiles, genetics, health, or prior medical history. This article documents the importance of nasal microbiome profiles in distinguishing infection rate trends among differentially affected subgroups. (2) Hypothesis: From a detailed literature survey, microbiome profiling experiments, bioinformatics, and molecular simulations, we propose that specific commensal bacterial species in the Pseudomonadales genus confer protection against SARS-CoV-2 infections by expressing proteases that may interfere with the proteolytic priming of the Spike protein. (3) Evidence: Various reports have found elevated Moraxella fractions in the nasal microbiomes of subpopulations with higher resistance to COVID-19 (e.g., adolescents, COVID-19-resistant children, people with strong dietary diversity, and omnivorous canines) and less abundant ones in vulnerable subsets (the elderly, people with narrower diets, carnivorous cats and foxes), along with bioinformatic evidence that Moraxella bacteria express proteases with notable homology to human TMPRSS2. Simulations suggest that these proteases may proteolyze the SARS-CoV-2 spike protein in a manner that interferes with TMPRSS2 priming.
Article
Full-text available
Vascular permeability triggered by inflammation or ischemia promotes edema, exacerbates disease progression and impairs tissue recovery. Vascular endothelial growth factor (VEGF) is a potent inducer of vascular permeability. VEGF plays an integral role in regulating vascular barrier function physiologically and in pathologies, including cancer, stroke, cardiovascular disease, retinal conditions and COVID-19-associated pulmonary edema, sepsis and acute lung injury. Understanding temporal molecular regulation of VEGF-induced vascular permeability will facilitate developing therapeutics to inhibit vascular permeability, while preserving tissue-restorative angiogenesis. Here, we demonstrate that VEGF signals through signal transducer and activator of transcription 3 (STAT3) to promote vascular permeability. We show that genetic STAT3 ablation reduces vascular permeability in STAT3-deficient endothelium of mice and VEGF-inducible zebrafish crossed with CRISPR/Cas9-generated Stat3 knockout zebrafish. Intercellular adhesion molecule 1 (ICAM-1) expression is transcriptionally regulated by STAT3, and VEGF-dependent STAT3 activation is regulated by JAK2. Pyrimethamine, an FDA-approved antimicrobial agent that inhibits STAT3-dependent transcription, substantially reduces VEGF-induced vascular permeability in zebrafish, mouse and human endothelium. Collectively, our findings suggest that VEGF/VEGFR-2/JAK2/STAT3 signaling regulates vascular barrier integrity, and inhibition of STAT3-dependent activity reduces VEGF-induced vascular permeability. This article has an associated First Person interview with the first author of the paper.
Article
Full-text available
Endothelial dysfunction is implicated in the thrombotic events reported in COVID-19 patients, but underlying molecular mechanisms are unknown. Circulating levels of the coagulation cascade activator PAI-1 are substantially higher in COVID-19 patients with severe respiratory dysfunction than in patients with bacterial-sepsis and ARDS. Indeed, the elevation of PAI-1 is recognized as an early marker of endothelial dysfunction. Here, we report that recombinant SARS-CoV-2-S1 stimulated robust production of PAI-1 by HPMEC. We examined the role of protein degradation in this SARS-CoV-2-S1 induction of PAI-1 and found that the proteasomal degradation inhibitor bortezomib inhibited SARS-CoV-2-S1 mediated changes in PAI-1. Our data further show that bortezomib upregulated KLF2, a shear-stress-regulated transcription factor that suppresses PAI-1 expression. Aging and metabolic disorders are known to increase the mortality and morbidity in COVID-19 patients. We therefore examined the role of ZMPSTE24, a metalloprotease with a demonstrated role in host defense against RNA viruses that is decreased in the elderly and in metabolic syndrome, in the induction of PAI-1 in HPMEC by SARS-CoV-2-S1. Indeed, overexpression of ZMPSTE24 blunted enhancement of PAI-1 production in spike protein-exposed HPMEC. Additionally, we found that membrane expression of the SARS-CoV-2 entry receptor ACE2 was reduced by ZMPSTE24-mediated cleavage and shedding of the ACE2 ectodomain, leading to accumulation of ACE2 decoy fragments that may bind SARS-CoV-2. These data indicate that decreases in ZMPSTE24 with age and comorbidities may increase vulnerability to vascular endothelial injury by SARS-CoV-2 viruses and that enhanced production of endothelial PAI-1 might play role in prothrombotic events in COVID-19 patients. This article is open access and distributed under the terms of the Creative Commons Attribution Non-Commercial No Derivatives License 4.0 (http://creativecommons.org/licenses/by-nc-nd/4.0/).
Article
Full-text available
We have read with great interest the two editorials by Burdorf et al: “The COVID-19 pandemic: one year later – an occupational perspective” (1) and “The COVID-19 (Coronavirus) pandemic: consequences for occupational health” (2). The authors highlight the importance of the societal consequences of the outbreak and changes in the world of work to manage occupational health. The key points identified – such as individual socio­economic factors, psychological effects and occupations with highest risk of contamination – modify return-to-work approaches. It is estimated that around 800 million people of working age worldwide were living with disabilities before the SARS-CoV-2 pandemic. In early January 2021, the cumulative COVID-19 hospitalisation rate reached 207.4/100 000 (18–49-year-olds) and 505.7/100 000 (50–64-year-olds), respectively, in the United States (3). In France, the hospitalisation rate was 411.5/100 000 across all ages (4). A recent cohort study of working-age men who were hospitalised for COVID-19 highlighted the long-term health consequences of such a disease (5). The SARS-CoV-2 pandemic creates new challenges for occupational health, shifting attention away from return-to-work after health problems to resuming work during an outbreak, dealing with lockdown, and taking special account of workers with vulnerabilities (6, 7). We recommend considering three different aspects of occupational medicine during a pandemic. Firstly, for most workers at high-risk of severe COVID-19, the issues of work disability and resuming work had never occurred before the epidemic. Recommendations such as physical and social distancing and wearing a facemask are highly advisable to protect against infection but may not be enough to enable some individuals to resume work. Therefore, decision-making requires individual comprehensive assessments of the underlying medical condition, the SARS-CoV-2 contamination risk associated with either regular work or teleworking, and vaccination opportunities. The second situation concerns workers who have suffered from COVID-19. Preliminary studies suggest that long recovery duration is related to high severity (7), but this is still a matter of debate for patients suffering from “long COVID-19” (5, 8, 9), a condition for which the long-term effects remain unknown. Any long-running recovery must be considered to be a potential sign of long COVID-19. These long-lasting syndromes occur among patients with severe symptoms but have also been reported independently of acute phase severity, hospitalisation and receiving medical oxygen (8, 9). Researchers worldwide are currently investigating such syndromes. Strategies promoting return to work for these workers will need to be implemented and could be similar to programmes developed for other chronic conditions. Moreover, numerous more serious sequelae following critical illness suggest the need for enhanced support by rehabilitation and occupational health specialists. Finally, the consequences of the epidemic must be evaluated over time for people who suffered from functional limitations before COVID-19 as their physical and mental condition may be modified by the epidemic and, specifically, the consequences of lockdown (10). In all of these situations, medical, social, financial and working contexts are key elements. In addition to a medical assessment, the use of scales such as the Work Ability Index (WAI) (11) or the Work Productivity and Activity Impairment (WPAI) (12) can help perform long-term follow-up and provide information about work capacity and workload. It also gives a “back to basics” perspective, urging politicians to move towards a `decent-work-for-all` policy, as advocated by the United Nation`s Sustainable Development Goal (SDG) 8, which the WHO has endorsed (13). References 1. Burdorf A, Porru F, Rugulies R. The COVID-19 pandemic: one year later – an occupational perspective. Scand J Work Environ Health – online first. https://doi.org/10.5271/sjweh.3956 2. Burdorf A, Porru F, Rugulies R. The COVID-19 (Coronavirus) pandemic: consequences for occupational health. Scand J Work Environ Health. 2020;46(3):229–230. https://doi:org/10.5271/sjweh.3893. 3. COVID-19 Hospitalizations [Internet]. Available from: https://gis.cdc.gov/grasp/COVIDNet/COVID19_3.html 4. COVID-19 in France, vaccine and allergy management in occupational setting. Descatha A et al. Arch Mal Prof Environ 2021. Accepted for publication. 5. Huang C, Huang L, Wang Y, Li X, Ren L, Gu X, et al. 6-month consequences of COVID-19 in patients discharged from hospital: a cohort study. Lancet 2021;397(10270):220-32 https://doi.org/10.1016/S0140-6736(20)32656-8 6. Shaw WS, Main CJ, Findley PA, Collie A, Kristman VL, Gross DP. Opening the Workplace After COVID-19: What Lessons Can be Learned from Return-to-Work Research? J Occup Rehabil. 2020;30(3):299–302. https://doi.org/10.1007/s10926-020-09908-9 7. Taylor T, Das R, Mueller K, Pransky G, Christian J, Orford R, et al. Safely Returning America to Work: Part I: General Guidance for Employers. J Occup Environ Med. 2020;62(9):771–9. https://doi.org/10.1097/JOM.0000000000001984 8. Carfì A, Bernabei R, Landi F, Gemelli Against COVID-19 Post-Acute Care Study Group. Persistent Symptoms in Patients After Acute COVID-19. JAMA. 2020;324(6):603–5. https://doi.org/10.1001/jama.2020.12603 9. Tenforde MW, Kim SS, Lindsell CJ, Billig Rose E, Shapiro NI, Files DC, et al. Symptom Duration and Risk Factors for Delayed Return to Usual Health Among Outpatients with COVID-19 in a Multistate Health Care Systems Network - United States, March-June 2020. MMWR Morb Mortal Wkly. 2020;69(30):993–8. https://doi.org/10.15585/mmwr.mm6930e1 10. Chudasama YV, Gillies CL, Zaccardi F, Coles B, Davies MJ, Seidu S, et al. Impact of COVID-19 on routine care for chronic diseases: A global survey of views from healthcare professionals. Diabetes Metab Syndr. 2020;14(5):965–7. https://doi.org/10.1016/j.dsx.2020.06.042 11. Tuomi K. Eleven-year follow-up of aging workers. Scand J Work Environ Health. 1997;23(1):1–71. 12. Reilly MC, Zbrozek AS, Dukes EM. The validity and reproducibility of a work productivity and activity impairment instrument. PharmacoEconomics. 1993;4(5):353–65. https://doi.org/10.2165/00019053-199304050-00006 13. Organization WH. Health in the 2030 agenda for sustainable development. Sixty-Ninth World Health Assembly. Document A. 2016, p69.
Article
Full-text available
Among COVID-19 hospitalized patients, high incidence of alterations in inflammatory and coagulation biomarkers correlates with a poor prognosis. Comorbidities such as chronic degenerative diseases are frequently associated with complications in COVID-19 patients. The aim of this study was to evaluate inflammatory and procoagulant biomarkers in COVID-19 patients from a public hospital in Mexico. Blood was sampled within the first 48 h after admission in 119 confirmed COVID-19 patients that were classified in 3 groups according to oxygen demand, evolution and the severity of the disease as follows: 1) Non severe: nasal cannula or oxygen mask; 2) Severe: high flow nasal cannula and 3) Death: mechanical ventilation eventually leading to fatal outcome. Blood samples from 20 healthy donors were included as a Control Group. Analysis of inflammatory and coagulation biomarkers including D-dimer, interleukin 6, interleukin 8, PAI-1, P-selectin and VWF was performed in plasma. Routine laboratory and clinical biomarkers were also included and compared among groups. Concentrations of D-dimer (14.5 ± 13.8 µg/ml) and PAI-1 (1223 ± 889.6 ng/ml) were significantly elevated in severe COVID-19 patients (P < 0.0001). A significant difference was found in interleukin-6, PAI-1 and P-selectin in non-severe and healthy donors when compared to Severe COVID-19 and deceased patients (P < 0.001). VWF levels were also significantly different between severe patients (153.5 ± 24.3 UI/dl) and non-severe ones (133.9 ± 20.2 UI/dl) (P < 0.0001). WBC and glucose levels were also significantly elevated in patients with Severe COVID-19. Plasma concentrations of all prothrombotic biomarkers were significantly higher in patients with a fatal outcome.
Article
Full-text available
Background: Accumulating evidence indicates towards an association between SARS-CoV-2 infection and procoagulatory state in blood. Thromboelastographic investigations are useful point-of-care devices to assess coagulation and fibrinolysis. Objectives: We investigated the hypothesis that the procoagulatory state in COVID-19 patients is caused by impaired fibrinolysis system. Methods: COVID-19 patients admitted to normal wards or to the intensive care unit (ICU) were included in the study. Whole blood samples were investigated by thromboelastography. Additionally, global parameters of coagulation and factors of fibrinolysis as plasminogen activator inhibitor-1 (PAI-1), tissue plasminogen activator (tPA), plasminogen activity and alpha 2-antiplasmin (A2AP) were determined. Results and conclusion: A significantly increased lysis-resistance and a significantly longer time of lysis after adding tissue plasminogen activator was observed in blood samples from ICU COVID-19 patients compared to controls (maximal lysis: 3.25% ± 0.56 vs. 6.20% ± 0.89, p=0.0127; lysis time: 365.7s ± 44.6 vs. 193.2s ± 16.3, p=0.0014). PAI-1 activity was significantly higher in plasma samples of ICU COVID-19 patients (PAI-1: 4.92U/ml ± 0.91 vs. 1.28U/ml ± 0.33, p=0.001). Interestingly, a positively correlation between the activity of PAI-1 and the lysis time of the formed clot (r=0.7015, p=0.0006) was observed. Our data suggest that severe SARS-CoV-2 infection is associated with impaired fibrinolytic activity in blood, where fibrinolytic inhibitors are elevated leading to an increased resistance to clot lysis. Future clinical studies should address the contribution of the fibrinolysis system to the procoagulatory state in blood of COVID-19 patients and investigate potential therapeutic targets in this system.
Article
Coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been led to a pandemic emergency. So far, different pathological pathways for SARS-CoV-2 infection have been introduced in which the excess release of pro-inflammatory cytokines (such as interleukin 1 β [IL-1β], IL-6, and tumor necrosis factor α [TNFα]) has earned most of the attentions. However, recent studies have identified new pathways with at least the same level of importance as cytokine storm in which endothelial cell (EC) dysfunction is one of them. In COVID-19, two main pathologic phenomena have been seen as a result of EC dysfunction: hyper-coagulation state and pathologic angiogenesis. The EC dysfunction-induced hypercoagulation state seems to be caused by alteration in the levels of different factors such as plasminogen activator inhibitor 1 (PAI-1), von Willebrand factor (vWF) antigen, soluble thrombomodulin, and tissue factor pathway inhibitor (TFPI). As data have shown, these thromboembolic events are associated with severity of disease severity or even death in COVID-19 patients. Other than thromboembolic events, pathologic angiogenesis is among the recent findings. Furthermore, over-expression/higher levels of different proangiogenic factors such as vascular endothelial growth factor (VEGF), hypoxia-inducible factor 1 α (HIF-1α), IL-6, TNF receptor super family 1A and 12, and angiotensin-converting enzyme 2 (ACE2) have been found in the lung biopsies/sera of both survived and non-survived COVID-19 patients. Also, there are some hypotheses regarding the role of nitric oxide in EC dysfunction and acute respiratory distress syndrome (ARDS) in SARS-CoV-2 infection. It has been demonstrated that different pathways involved in inflammation are generally common with EC dysfunction and angiogenesis. Altogether, considering the common possible upstream pathways in cytokine storm, pathologic angiogenesis, and EC dysfunction, it seems that targeting these molecules (such as nuclear factor κB) could be more effective in the management of patients with COVID-19.
Article
Numerous studies have established that acute or chronic exposure to environmental pollutants like particulate matter (PM) leads to the development of accelerated aging related pathologies including pulmonary and cardiovascular diseases, and thus air pollution is one of the major global threats to human health. Air pollutant particulate matter 2.5 (PM2.5)-induced cellular dysfunction impairs tissue homeostasis and causes vascular and cardiopulmonary damage. To test a hypothesis that elevated plasminogen activator inhibitor-1 (PAI-1) levels play a pivotal role in air pollutant-induced cardiopulmonary pathologies, we examined the efficacy of a drug-like novel inhibitor of PAI-1, TM5614, in treating PM2.5-induced vascular and cardiopulmonary pathologies. Results from biochemical, histological, and immunohisto-chemical studies revealed that PM2.5 increases the circulating levels of PAI-1 and thrombin and that TM5614 treatment completely abrogates these effects in plasma. PM2.5 significantly augments the levels of pro-inflammatory cytokine interleukin-6 (IL-6) in bronchoalveolar lavage fluid (BALF), and this also can be reversed by TM5614, indicating its efficacy in amelioration of PM2.5-induced increases in inflammatory and pro-thrombotic factors. TM5614 reduces PM2.5-induced increased levels of inflammatory markers cluster of differentiation 107b (Mac3) and signal transducer and activator of transcription-3 (pSTAT3), adhesion molecule vascular cell adhesion molecule 1 (VCAM1), and apoptotic marker cleaved caspase 3. Longer exposure to PM2.5 induces pulmonary and cardiac thrombosis, but TM5614 significantly ameliorates PM2.5-induced vascular thrombosis. TM5614 also reduces PM2.5-induced increased blood pressure and heart weight. In vitro cell culture studies revealed that PM2.5 induces the levels of PAI-1, type I collagen, fibronectin, and sterol regulatory element binding protein-1 and 2 (SREBP-1 and SREBP-2), transcription factors that mediate profibrogenic signaling, in cardiac fibroblasts. TM5614 abrogated that stimulation, indicating that it may block PM2.5-induced PAI-1 and profibrogenic signaling through suppression of SREBP-1 and 2. Furthermore, TM5614 blocked PM2.5-mediated suppression of nuclear factor erythroid related factor 2 (Nrf2), a major antioxidant regulator, in cardiac fibroblasts. Pharmacological inhibition of PAI-1 with TM5614 is a promising therapeutic approach to control air pollutant PM2.5-induced cardiopulmonary and vascular pathologies.
Article
Background: Acute cerebral infarction is a clinically common and critical disease which seriously endangers the life and safety of elderly patients. Evidence-based nursing is an effective way of nursing and has great significance in improving the neurological function and quality of life of patients. In China, evidence-based nursing has been highlighted and highly developed in recent decades. Objectives: This research aimed to investigate the effect of evidence-based nursing on the recovery of neurological function and serum inflammatory cytokines in patients with acute cerebral infarction. Methods: A total of 116 patients with acute cerebral infarction were randomly divided into two groups: the control group patients (n = 58) received conventional nursing, while the intervention group patients (n = 58) received evidence-based nursing intervention. National Institutes of Health Stroke Scale (NIHSS), Fugl-Meyer assessment (FMA) and activities of daily living (ADL) scores, as well as serum TNF-α and IL-6 levels were evaluated and compared between the two groups. Results: NIHSS scores in the intervention group were significantly lower than the control group. FMA and ADL scores in the intervention group were significantly higher than the control group. TNF-α and IL-6 levels in the serum of the intervention group were significantly lower than the control group. Conclusions: In conclusion, evidence-based nursing has a positive effect on the treatment of patients with acute cerebral infarction, which decreases the level of serum inflammatory cytokines and contributes to the recovery of neurological function, motor function and activities of daily living.
Article
Circulating concentrations of the pleiotropic cytokine interleukin-6 (IL-6) are known to be increased in pro-inflammatory critical care syndromes, such as sepsis and acute respiratory distress syndrome. Elevations in serum IL-6 concentrations in patients with severe COVID-19 have led to renewed interest in the cytokine as a therapeutic target. However, although the pro-inflammatory properties of IL-6 are widely known, the cytokine also has a series of important physiological and anti-inflammatory functions. An adequate understanding of the complex processes by which IL-6 signalling occurs is crucial for the correct interpretation of IL-6 concentrations in the blood or lung, the use of IL-6 as a critical care biomarker, or the design of effective anti-IL-6 strategies. Here, we outline the role of IL-6 in health and disease, explain the different types of IL-6 signalling and their contribution to the net biological effect of the cytokine, describe the approaches to IL-6 inhibition that are currently available, and discuss implications for the future use of treatments such as tocilizumab in the critical care setting.
Article
Latest research show that SERPINE1 overexpression plays an important role in COVID-19-associated coagulopathy leading to acute respiratory distress syndrome (ARDS). Ways to target this protein remain elusive. In this forum, we discuss recent evidence linking SERPINE1 with COVID-19 related ARDS and summarize the available data on inhibitors of this target.