ArticlePDF AvailableLiterature Review

COVID-19 and Neutrophils: The Relationship between Hyperinflammation and Neutrophil Extracellular Traps

Wiley
Mediators of Inflammation
Authors:

Abstract and Figures

Coronavirus disease 2019 (COVID-19) is a virus-induced respiratory disease that may progress to acute respiratory distress syndrome (ARDS) and is triggered by immunopathological mechanisms that cause excessive inflammation and leukocyte dysfunction. Neutrophils play a critical function in the clearance of bacteria with specific mechanisms to combat viruses. The aim of this review is to highlight the current advances in the pathways of neutrophilic inflammation against viral infection over the past ten years, focusing on the production of neutrophil extracellular traps (NETs) and its impact on severe lung diseases, such as COVID-19. We focused on studies regarding hyperinflammation, cytokine storms, neutrophil function, and viral infections. We discuss how the neutrophil's role could influence COVID-19 symptoms in the interaction between hyperinflammation (overproduction of NETs and cytokines) and the clearance function of neutrophils to eliminate the viral infection. We also propose a more in-depth investigation into the neutrophil response mechanism targeting NETosis in the different phases of COVID-19.
This content is subject to copyright. Terms and conditions apply.
Review Article
COVID-19 and Neutrophils: The Relationship between
Hyperinflammation and Neutrophil Extracellular Traps
Leandro Borges ,
1
Tania Cristina Pithon-Curi,
1
Rui Curi,
1,2
and Elaine Hatanaka
1
1
Instituto de Ciências da Atividade Física e Esportes (ICAFE), Universidade Cruzeiro do Sul, São Paulo, SP, Brazil
2
Instituto Butantan, São Paulo, SP, Brazil
Correspondence should be addressed to Leandro Borges; sbleandro@yahoo.com.br
Received 8 September 2020; Revised 15 November 2020; Accepted 25 November 2020; Published 4 December 2020
Academic Editor: Juliana Vago
Copyright © 2020 Leandro Borges et al. This is an open access article distributed under the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
Coronavirus disease 2019 (COVID-19) is a virus-induced respiratory disease that may progress to acute respiratory distress
syndrome (ARDS) and is triggered by immunopathological mechanisms that cause excessive inammation and leukocyte
dysfunction. Neutrophils play a critical function in the clearance of bacteria with specic mechanisms to combat viruses. The
aim of this review is to highlight the current advances in the pathways of neutrophilic inammation against viral infection over
the past ten years, focusing on the production of neutrophil extracellular traps (NETs) and its impact on severe lung diseases,
such as COVID-19. We focused on studies regarding hyperinammation, cytokine storms, neutrophil function, and viral
infections. We discuss how the neutrophils role could inuence COVID-19 symptoms in the interaction between
hyperinammation (overproduction of NETs and cytokines) and the clearance function of neutrophils to eliminate the viral
infection. We also propose a more in-depth investigation into the neutrophil response mechanism targeting NETosis in the
dierent phases of COVID-19.
1. Introduction
The World Health Organization (WHO) established the
coronavirus disease 2019 (COVID-19) as a pandemic on
March 11, 2020. Severe acute respiratory syndrome corona-
virus 2 (SARS-CoV-2) is a member of the coronavirus family,
a class of enveloped viruses with a positive-sense single-
stranded RNA genome. This virus can cross species barriers
and induce illnesses ranging from the usual cold to severe
interstitial pneumonia, respiratory failure, and septic shock
[1]. While there is a global eort in the development of
vaccines and improvement of diagnostic methods [2, 3] and
therapies that relieve the symptoms and prognosis of
COVID-19 patients under severe infection [4], there remain
gaps in our understanding of the pathophysiology of
COVID-19 related to innate immunity.
In a scenario where patients with severe COVID-19 could
develop dysfunction of the immune response that aggravates
the hyperinammation [5, 6], it is hypothesized that neutro-
phils can amplify pathological damage or control other cell
subsets depending on the infection features. Therefore, to
use the potential of NETs with minimal damage to the hosts,
there must be a right balance of NET formation and reduc-
tion of the amount of NETs that accumulate in tissues [7].
Notwithstanding the rapid progress in the eld, there are
many critical unknown features of neutrophils in ghting
viral infections. We highlighted the current progress in the
pathways of neutrophilic inammation in viral infection,
with a focus on the release of NETs and its inuence on lung
disease. The knowledge summarized in this study should
benet researchers in integrating neutrophil biology to
design new and more ecient virus-targeted interventions
concerning COVID-19.
2. Hyperinflammation
Although a well-regulated innate immune process is the rst
protection action against viral infections [8], in severe
COVID-19 condition occurs hyperinammation (cytokine
Hindawi
Mediators of Inflammation
Volume 2020, Article ID 8829674, 7 pages
https://doi.org/10.1155/2020/8829674
storm) that might lead to the acute respiratory distress
syndrome (ARDS) [6, 9].
Cytokines play a relevant function in immunopathology
during virus infections. The host-viral interactions are estab-
lished via host identication of pathogen-associated molecu-
lar patterns (PAMPs) of the virus [10]. This identication
occurs through host pattern recognition receptors (PRRs)
manifested on innate immune cells (e.g., neutrophils,
dendritic cells, epithelial cells, and macrophages) [11], and
the recognition of PAMPs and viral danger-associated
molecular patterns (DAMPs) by conserved PRRs marks the
rst line of defense against pathogens, involving toll-like
receptors (TLRs) [11].
TLR stimulation activates the nuclear factor-κB (NF-κB)
signaling cascade, causing the production of inammatory
markers from monocytes (interleukin- (IL-) 1, tumor necro-
sis factor-alpha (TNF-α), and IL-6) to control virus infec-
tions [8] by direct antiviral pathways and the recruitment
of other leukocytes [10]. Moreover, the exacerbated oxidative
stress induced by elevated concentrations of cytokines, along
with reduced concentrations of interferon αand interferon β
(IFN-α, IFN-β), inuences the severity of COVID-19 [12].
Several mediators control the release of chemoattractants
and neutrophil activity [10], and studies have demonstrated
that higher values of proinammatory markers are related
to extensive lung damage and pulmonary inammation in
MERS-CoV [13] and ARDS infection [14]. COVID-19 in
the severe state exhibits a cytokine storm with elevated
plasma levels of chemokine ligand 2 (CCL2), IFNγ, IFNγ-
inducible protein 10, G-CSF, chemokine C-C motif ligand 3
(CCL3), IL-1β, IL-2, IL-6, IL-7, IL-8, IL-10, IL-17, and
TNF-α[12, 15]. Nucleotide-binding oligomerization
domain- (NOD-) like receptor and increased plasma levels
of chemokines and cytokines in COVID-19 patients relate
to the severity of the disease rather than did those nonsevere
patients [5]. In this sense, Huang et al. [15] found that
patients in the intensive care unit (ICU) with laboratory-
conrmed COVID-19 infection had higher plasma levels of
IL-2, IL-7, IL-10, interferon-inducible protein 10, granulo-
cyte colony-stimulating factor, CCL2, CCL3, and TNF-α
when compared with non-ICU patients [15].
3. Neutrophils: The First Cell Recruitment
Neutrophils are innate immune cells with a brief lifespan
after leaving the bone marrow and exist in a quiescent,
primed, or active state. These leukocytes are the leading
players in innate immunity since they are among the rst
innate leukocytes recruited during infections [16]. The pri-
mary function of neutrophil is clearance of pathogens and
debris through phagocytosis [17]. They also have a distinct
array of other immune roles, such as the liberation of NETs
for viral infection inactivation [18] and cytokine production
to restrict virus replication [16].
The release of neutrophil-chemoattractive elements and
the resulting recruitment of neutrophils are a global host
response to viral infection [19]. In this scenario, the neutro-
phil cell membrane also expresses a complex array of recep-
tors and adhesion molecules for various ligands, including
immunoglobulins, membrane molecules on other cells, and
cytokines [20].
In addition to the tracking to infection places to phago-
cytize viruses, the neutrophils can initiate, enlarge, and/or
repress adaptive immune eector processes by promoting
bidirectional cross-talk with T cells [21, 22]. Following the
acute inammation arising from immunological processes,
such as viral infections, neutrophils with decreased expres-
sion of CD62L weaken T cell migration via the CXCL11 che-
mokine gradient by releasing H
2
O
2
into an immunological
synapse [23]. Thus, neutrophils that uncovered viral antigens
can home to draining lymph nodes, acting as antigen-
presenting cells (APC) [24]. Huord et al. [25] evidenced
that neutrophils expressing viral antigen as an outcome of
direct infection by inuenza A virus (IAV) display the most
potent APC activity and that viral antigen-presenting neutro-
phils inltrating the IAV-infected lungs act as APC for eec-
tor CD8(+) T lymphocytes in the infected lungs [25].
Neutrophils recruit the T cell molecular mechanism during
the inuenza virus infection and associate to CXCL12 reser-
voirs left behind. CD8+ T cells follow the chemoattractant
trail left behind by neutrophil uropods to the inuenza virus
infection site [26].
Decreased cell number or impaired leukocyte function
can play a part in advance of mild to severe clinical disease
conditions [16]. Regarding the new coronavirus, the
neutrophil-to-lymphocyte ratio (NLR), a well-known marker
of infection and systemic inammation, has evidenced an
enhanced inammatory response in COVID-19 patients
[5]. Since the ARDS is the primary cause of mortality in
patients with COVID-19, the elevated NLR values suggest a
poor prognosis in COVID-19 disease [27], especially severe
COVID-19 compared to mild patients. Sun et al. [28] studied
116 patients with COVID-19 and showed a higher NLR [28].
The authors compared severe COVID-19 patients admitted
to the ICU with others or severe patients not admitted to
the ICU. They reported that COVID-19 patients have the
lowest count of lymphocytes and the highest neutrophil
count and NLR [28]. Wang et al. [29] also showed that sev-
eral COVID-19 patients have a rising neutrophil count and
a falling lymphocyte count during the severe phase [29].
Similarly, Barnes et al. [30] found extensive neutrophil inl-
tration in pulmonary capillaries from a COVID-19 patient
[30]. Nevertheless, even though severe cases of COVID-19
appear to be related to increased NLR levels [5], whether
NLR could be an independent predictor of mortality in
COVID-19 patients still requires investigation.
4. Neutrophil Extracellular Traps (NETs) and
Viral Infection
Neutrophils can develop a sophisticated network of DNA
called NETs through NETosis, a liberation of web-like struc-
tures of nucleic acids wrapped with histones that detain viral
particles [31]. Upon discovery, the researchers believed that
the production of NETs defended only against fungi and bac-
teria [32]. However, the NETosis process plays an important
function in the response to viral diseases [33], thereby
2 Mediators of Inammation
protecting the host during the virus response by trapping and
eliminating distinct pathogens [31].
The formation of NETs is a controlled process, even
though the related signals remain unknown. NETosis is con-
ditional on the production of reactive oxygen species (ROS)
by nicotinamide adenine dinucleotide phosphate oxidase
(NADPH oxidase) [34]. There is evidence of NETosis pro-
duced in a ROS-independent mechanism [35]. In general,
the NETosis process includes the release of nuclear chroma-
tin lined with eector proteins and peptidyl arginine deimi-
nase type IV (PAD4) activation [36]. After stimulation, the
neutrophil nuclear envelope disintegrates to enable the
mixing of chromatin with granular proteins [37]. Myeloper-
oxidase (MPO) and neutrophil elastase (NE) stimulate
chromatin condensation and deteriorate histones [38]. In
the presence of histone hypercitrullination, PAD4 mediates
chromatin decondensation, and the DNA-protein complexes
are released extracellularly as NETs [37]. Therefore, dier-
ently from apoptosis or necrosis, both the granular mem-
brane and nuclear membrane deteriorate during NETosis,
whereas plasma membrane integrity remains [36].
The overproduction of NETs induces lung tissue damage
by NETosis-related enzymes such as NE and MPO [39].
Uncontrolled NET production correlates with disease gravity
and lung injury extension. For instance, NETosis markers
are related to bacterial burden and local inammation in the
lung [40] and patients with pneumonia-associated ARDShave
neutrophils in a primedcondition to generate NETs [41].
During chronic obstructive pulmonary disease aggrava-
tion, the production of NETs increases in people with acute
respiratory failure [39] and in ARDS patients [40, 42]. The
elevated NET production, as noted in patients with severe
IAV infection [43], increased injury to the pulmonary
endothelial and epithelial cells [44], directing to severe pneu-
monia. Zhu et al. [43] also noted that the production of NETs
positively correlates with multiple organ dysfunction
syndromes [43].
The inammatory process is a trigger for thrombotic
complications usually noted in COVID-19 patients, and the
immunothrombotic dysregulation seems to be an important
key marker for the disease severity [45]. Skendros et al. [46]
found that complement activation potentiates the platelet/-
NET/tissue factor/thrombin axis during SARS-CoV-2 infec-
tion [46]. In contrast, Nicolai et al. [47] noted that, in
COVID-19, inammatory microvascular thrombi are found
in the kidney, lung, and heart, containing NETs related to
the brin and platelets. In blood, Nicolai et al. also show that
COVID-19 patients have neutrophil-platelet aggregates and
adierent platelet and neutrophil activation pattern, which
alters with the disease severity [47]. Middleton et al. [48] also
found that plasma MPO-DNA complexes increased in
COVID-19 and that the elevated NET formation correlates
with COVID-19-related ARDS. Together, these ndings sug-
gest the timely application of therapeutic strategies that can
disrupt the vicious cycle of COVID-19 immunothrombosis/-
thromboinammation by targeting neutrophil activation and
NET formation.
In addition to the physical containment promoted by
NETosis [33], NETs contain DNA, modied extracellular
histones, proteases, and cytotoxic enzymes that allow neutro-
phils to centralize lethal proteins at infection sites [7]. The
mechanisms of NETsrelease in the viral response seem to
Recognition mechanism
PAMP/DAMP activation
Neutrophil inltration
IL-6
IL-8
Hyperinammation
TNF
IL-1
ROS
Cytokine storms
Lung thromboinammation
SARS-CoV-2
Viral infection
Alveolar
injury
SARS-CoV-2
NETosis
NET overproduction
MPO
Histones
NE
Figure 1: The interaction hypothesis between neutrophil and hyperinammation in COVID-19. After the host-viral interaction, the virus
signaling leads to a cascade of interactions between the virus recognition mechanism, neutrophil activation, and inammatory stimuli.
The NETosis process can protect the host during the virus response or exacerbate lung hyperinammation in COVID-19 patients. The
gure is made with BioRender (https://app.biorender.com/). Abbreviations: SARS-CoV-2: severe acute respiratory syndrome coronavirus
2; PAMP: pathogen-associated molecular pattern; DAMP: danger-associated molecular pattern; TNF: tumor necrosis factor; IL-6:
interleukin-6; IL-1: interleukin-1; IL-8: interleukin-8; ROS: reactive oxygen species; NE: neutrophil elastase; MPO: myeloperoxidase.
3Mediators of Inammation
involve neutrophil NE production attributed to the change of
macrophage role by the cleavage of TLRs [49]. A range of
stimuli, including toxic factors, viruses, and proinammatory
cytokines, such as TNF-αand IL-8, can lead neutrophils to
release NETs [7, 33]. Mechanisms that determine strain spec-
icity to induce NETosis formation during viral infection are
still unknown.
Lung inammation is the leading cause of the life-
threatening respiratory complication at the severe levels of
COVID-19 [50]. Veras et al. [51] investigated the potentially
detrimental function of NETs in the pathophysiology of 32
hospitalized severe COVID-19 patients and found that the
levels of NETs increase in tracheal aspirate and plasma from
patients with COVID-19 and their neutrophils naturally pro-
duced more signicant concentrations of NETs [51]. The
authors also reported NETs in the lung tissue specimens
from autopsies of COVID-19 patients. In vitro, they noted
that viable SARS-CoV-2 cause NET production by healthy
neutrophils through a PAD-4-dependent manner and that
NETs produced by SARS-CoV-2-activated neutrophils
instigated lung epithelial cell death [51]. Zuo et al. [52] also
investigated sera from COVID-19 patients and found higher
cell-free DNA, myeloperoxidase-DNA (MPO-DNA), and
citrullinated histone H3 (Cit-H3) [52]. In vitro, they also
noted that sera from COVID-19 patients trigger NET release
from control neutrophils [52].
Although the literature does not report direct evidence
linking NETs and SARS-CoV2 clearance, virus entrapping
by NETs was already found in syncytial respiratory virus
infection [53] or inuenza [54]. Furthermore, in virus infec-
tion, NETs are ecient to block viruses at the infection site,
entrapping them in a DNA web [22]. Therefore, the NETosis
process induced by the virus could operate as a double-edged
sword: on the one hand, there are essential and ecient
mechanisms for trapping the virus [55], and on the other,
there are highly intense immunological and inammatory
processes triggered by NET release causing damage to the
organism [7]. These interactions could inuence the
COVID-19 symptoms in the relationship between hyperin-
ammation (overproduction of NETs and cytokine storm)
and the function of neutrophils to destroy the viral infection
(Figure 1).
5. Concluding Remarks and Future Directions
The exacerbated NET formation can drive to a cascade of
inammatory reactions that destroys surrounding tissues,
favors microthrombosis, contributes to the progress of can-
cer cell metastasis, and results in permanent damage to the
pulmonary, cardiovascular, and renal systems [56]. Whether
by coincidence or a cause-and-eect relationship, these
organs are aected in the severe state of the COVID-19
Table 1: Interventional studies registered at the ClinicalTrials.gov database relating the treatment of COVID-19 with NET inhibitors.
NCT identier Status Location Study type Condition
or disease
Intervention
and phase Primary outcome
Estimated
completion
date
NCT04409925 Not yet
recruiting Canada Nonrandomized
pilot study COVID-19 Dornase Alfa
Phase: 1 (1) Rate of all adverse events January 2021
NCT04359654 Not yet
recruiting
United
Kingdom
Randomized
clinical trial
COVID-19
Hypoxia
Drug:
Dornase Alfa
Phase: 2
(1) Change in inammation
(C-reactive protein)
November
2020
NCT04445285 Recruiting United
States
Randomized
clinical trial COVID-19 Dornase Alfa
Phase: 2
(1) All-cause mortality
(2) Systemic therapeutic response
February
2021
NCT04432987 Recruiting Turkey Randomized
clinical trial COVID-19 Dornase Alfa
Phase: 2
(1) Clinical improvement and
inammatory markers in blood
(2) Intubation or extubation
September
2020
NCT04402944 Not yet
recruiting
United
States
Randomized
clinical trial COVID-19 Dornase Alfa
Phase: 2 (1) Ventilator-free days December
2021
NCT04322565 Recruiting Italy Randomized
clinical trial
COVID-19
Pneumonia
Colchicine
Phase: 2
(1) Clinical improvement
(2) Hospital discharge
December
2020
NCT04326790 Recruiting Greece Randomized
clinical trial COVID-19 Colchicine
Phase: 2
(1) Time to clinical deterioration
(2) Concentration of cardiac
troponin
September
2020
NCT04402970 Recruiting United
States
Nonrandomized
clinical trial
COVID-19
ARDS
Dornase Alfa
Phase: 3
(1) Improvement in partial pressure
of O
2
to fraction of inspired O
2
ratio May 2022
NCT04355364 Recruiting France Randomized
clinical trial
COVID-19
ARDS
Dornase Alfa
Phase: 3
(1) Occurrence of at least one grade
improvement (ARDS scale severity) August 2020
NCT04322682 Recruiting United
States
Randomized
clinical trial COVID-19 Colchicine
Phase: 3
(1) Number of participants who die
or require hospitalization
December
2020
NCT04328480 Recruiting Argentina Randomized
clinical trial COVID-19 Colchicine
Phase: 3
(1) Number of participants who die
(all-cause mortality) August 2020
Retrieved October 30, 2020, from https://www.https://www.clinicaltrials.gov/ct2/home. Abbreviations: NCT: National Clinical Trial; O
2
: oxygen; ARDS: acute
respiratory distress syndrome.
4 Mediators of Inammation
disease [57, 58]. The uncontrolled and poorly acknowledged
host response regarding the cytokine storm is one of the
major causes of severe COVID-19 conditions [12]. In this
pandemic scenario, there is a compelling need to investigate
the mechanisms associated with hyperinammation process
and NET production in response to COVID-19.
The NLR is an independent risk factor for severe
COVID-19 [27], and neutrophilia forecasts poor outcomes
in COVID-19 patients [29]. In this sense, new frontiers in
NET assessment regarding COVID-19 may be expressed by
analyzing NETosis directly after sputum induction or after
bronchoscopy using the bronchial alveolar uid of COVID-
19 patients [42]. Since patient samples usually become acces-
sible at the hospital, it could investigate whether the existence
of NETs is associated with the severity of COVID-19.
Treatments using NET-targeting approaches, although
would not directly target the new coronavirus, could reduce
the damage caused by hyperinammation [59], thereby
decreasing the diseases severity and avoiding invasive
mechanical ventilation, consequently diminishing mortality.
Drugs that target NETs include inhibitors of the molecules
necessary for NET formation, such as gasdermin D [60],
PAD4 [61], and NE [62]. Studies on treatment of inamma-
tory state in COVID-19 patients with NET inhibitors are still
in development (please see Table 1).
Caution is needed to dene which people would advan-
tage from suppressing the neutrophil response and which
would help more from a strengthened neutrophil action dur-
ing viral infections. Despite prior studies linking pulmonary
diseases to aberrant NET formation
[
3, 4
]
, our understanding
of NETosis mechanisms in viral infection is still limited.
The hyperinammation is related to the severity of
COVID-19 by inuencing the pulmonary inammation
[12]. Neutrophils exhibit an intense response to virus infec-
tion, promoting bidirectional cross-talk with T cells [21].
Neutrophils also express a complex array of receptors and
adhesion molecules for various ligands, including immuno-
globulins and inammatory markers [20]. In this sense,
severe cases of COVID-19 appear to be related to increased
NLR levels [5], and treatments using NET-targeting
approaches have the potential to decrease the damage caused
by hyperinammation [40, 41]. The researchers should
consider hyperinammation in the dierent phases of
COVID-19, neutrophil response mechanisms, and NETosis.
Data Availability
The data supporting this narrative review are from previously
reported studies and datasets, which have been cited.
Conflicts of Interest
The authors declare that there is no conict of interest
regarding the publication of this paper.
Acknowledgments
This work was supported by the CAPES (Coordenação de
Aperfeiçoamento de Pessoal de Nível Superior) (grant
number 88882.314890/2013-01); FAPESP (Fundação de
Amparo a Pesquisa do Estado de São Paulo); and CNPq
(Conselho Nacional de Desenvolvimento Cientíco e
Tecnológico).
References
[1] G. Kutti-Sridharan, R. Vegunta, R. Vegunta, B. P. Mohan, and
V. R. P. Rokkam, SARS-CoV2 in dierent body uids, risks of
transmission, and preventing COVID-19: a comprehensive
evidence-based review,International Journal of Preventive
Medicine, vol. 11, p. 97, 2020.
[2] L. L. Fernandes, V. B. Pacheco, L. Borges et al., Saliva in the
diagnosis of COVID-19: a review and new research direc-
tions,Journal of Dental Research, vol. 99, no. 13, pp. 1435
1443, 2020.
[3] L. L. Fernandes, L. Borges, V. B. Pacheco et al., SARS-CoV-2 :
a promising path in salivary diagnosis,The Open Dentistry
Journal, vol. 14, no. 1, pp. 343-344, 2020.
[4] A. C. L. Fernandes, A. J. M. Vale, F. P. Guzen, F. I. Pinheiro,
R. N. Cobucci, and E. P. de Azevedo, Therapeutic options
against the new coronavirus: updated clinical and laboratory
evidences,Frontiers in Medicine, vol. 7, p. 546, 2020.
[5] C. Qin, L. Zhou, Z. Hu et al., Dysregulation of immune
response in patients with coronavirus 2019 (COVID-19) in
Wuhan, China,Clinical Infectious Diseases, vol. 71, no. 15,
pp. 762768, 2020.
[6] Q. Ruan, K. Yang, W. Wang, L. Jiang, and J. Song, Clinical
predictors of mortality due to COVID-19 based on an analysis
of data of 150 patients from Wuhan, China,Intensive Care
Medicine, vol. 46, no. 5, pp. 846848, 2020.
[7] O. Z. Cheng and N. Palaniyar, NET balancing: a problem in
inammatory lung diseases,Frontiers in Immunology, vol. 4,
p. 1, 2013.
[8] G. Zhu, Y. Xu, X. Cen, K. S. Nandakumar, S. Liu, and
K. Cheng, Targeting pattern-recognition receptors to dis-
cover new small molecule immune modulators,European
Journal of Medicinal Chemistry, vol. 144, pp. 8292, 2018.
[9] C.-K. Min, S. Cheon, N.-Y. Ha et al., Comparative and kinetic
analysis of viral shedding and immunological responses in
MERS patients representing a broad spectrum of disease sever-
ity,Scientic Reports, vol. 6, no. 1, article 25359, 2016.
[10] R. Channappanavar and S. Perlman, Pathogenic human coro-
navirus infections: causes and consequences of cytokine storm
and immunopathology,Seminars in Immunopathology,
vol. 39, no. 5, pp. 529539, 2017.
[11] D. Kolli, T. S. Velayutham, and A. Casola, Host-viral interac-
tions: role of pattern recognition receptors (PRRs) in human
pneumovirus infections,Pathogens, vol. 2, no. 2, pp. 232
263, 2013.
[12] P. Mehta, D. F. McAuley, M. Brown, E. Sanchez, R. S. Tatter-
sall, and J. J. Manson, COVID-19: consider cytokine storm
syndromes and immunosuppression,The Lancet, vol. 395,
no. 10229, pp. 1033-1034, 2020.
[13] W. H. Mahallawi, O. F. Khabour, Q. Zhang, H. M. Makhdoum,
and B. A. Suliman, MERS-CoV infection in humans is associ-
ated with a pro-inammatory Th1 and Th17 cytokine prole,
Cytokine, vol. 104, pp. 813, 2018.
[14] M. Prete, E. Favoino, G. Catacchio, V. Racanelli, and F. Perosa,
SARS-CoV-2 inammatory syndrome. Clinical features and
5Mediators of Inammation
rationale for immunological treatment,International Journal
of Molecular Sciences, vol. 21, no. 9, article 3377, 2020.
[15] C. Huang, Y. Wang, X. Li et al., Clinical features of patients
infected with 2019 novel coronavirus in Wuhan, China,The
Lancet, vol. 395, no. 10223, pp. 497506, 2020.
[16] P. P. Lamichhane and A. E. Samarasinghe, The role of innate
leukocytes during inuenza virus infection,Journal of Immu-
nology Research, vol. 2019, Article ID 8028725, 17 pages, 2019.
[17] C. Rosales, Neutrophils at the crossroads of innate and adap-
tive immunity,Journal of Leukocyte Biology, vol. 108, no. 1,
pp. 377396, 2020.
[18] F. D. Barr, C. Ochsenbauer, C. R. Wira, and M. Rodriguez-
Garcia, Neutrophil extracellular traps prevent HIV infection
in the female genital tract,Mucosal Immunology, vol. 11,
no. 5, pp. 14201428, 2018.
[19] J. Bordon, S. Aliberti, R. Fernandez-Botran et al., Under-
standing the roles of cytokines and neutrophil activity and
neutrophil apoptosis in the protective versus deleterious
inammatory response in pneumonia,International Journal
of Infectious Diseases, vol. 17, no. 2, pp. e76e83, 2013.
[20] C. D. Russell, S. A. Unger, M. Walton, and J. Schwarze, The
human immune response to respiratory syncytial virus infec-
tion,Clinical Microbiology Reviews, vol. 30, no. 2, pp. 481
502, 2017.
[21] S. Costa, D. Bevilacqua, M. A. Cassatella, and P. Scapini, Recent
advances on the crosstalk between neutrophils and B or T lym-
phocytes,Immunology, vol. 156, no. 1, pp. 2332, 2019.
[22] V. D. Giacalone, C. Margaroli, M. A. Mall, and
R. Tirouvanziam, Neutrophil adaptations upon recruitment
to the lung: new concepts and implications for homeostasis
and disease,International Journal of Molecular Sciences,
vol. 21, no. 3, p. 851, 2020.
[23] J. Pillay, V. M. Kamp, E. van Hoen et al., A subset of neutro-
phils in human systemic inammation inhibits T cell
responses through Mac-1,The Journal of Clinical Investiga-
tion, vol. 122, no. 1, pp. 327336, 2012.
[24] M. V. Lukens, A. C. van de Pol, F. E. J. Coenjaerts et al., A sys-
temic neutrophil response precedes robust CD8
+
T-cell activa-
tion during natural respiratory syncytial virus infection in
infants,Journal of Virology, vol. 84, no. 5, pp. 23742383,
2010.
[25] M. M. Huord, G. Richardson, H. Zhou et al., Inuenza-
infected neutrophils within the infected lungs act as antigen
presenting cells for anti-viral CD8(+) T cells,PLoS One,
vol. 7, no. 10, article e46581, 2012.
[26] K. Lim, Y.-M. Hyun, K. Lambert-Emo et al., Neutrophil trails
guide inuenza-specic CD8
+
T cells in the airways,Science,
vol. 349, no. 6252, article aaa 4352, 2015.
[27] F. A. Lagunas-Rangel, Neutrophil-to-lymphocyte ratio and
lymphocyte-to-C-reactive protein ratio in patients with severe
coronavirus disease 2019 (COVID-19): a meta-analysis,Jour-
nal of Medical Virology, vol. 92, no. 10, pp. 1733-1734, 2020.
[28] S. Sun, X. Cai, H. Wang et al., Abnormalities of peripheral
blood system in patients with COVID-19 in Wenzhou,
China,Clinica Chimica Acta, vol. 507, pp. 174180, 2020.
[29] D. Wang, B. Hu, C. Hu et al., Clinical characteristics of 138
hospitalized patients with 2019 novel coronavirus-infected
pneumonia in Wuhan, China,JAMA, vol. 323, no. 11,
pp. 10611069, 2020.
[30] B. J. Barnes, J. M. Adrover, A. Baxter-Stoltzfus et al., Target-
ing potential drivers of COVID-19: neutrophil extracellular
traps,The Journal of Experimental Medicine, vol. 217, no. 6,
article e20200652, 2020.
[31] C. N. Jenne, C. H. Y. Wong, F. J. Zemp et al., Neutrophils
recruited to sites of infection protect from virus challenge by
releasing neutrophil extracellular traps,Cell Host & Microbe,
vol. 13, no. 2, pp. 169180, 2013.
[32] V. Brinkmann, U. Reichard, C. Goosmann et al., Neutrophil
extracellular traps kill bacteria,Science, vol. 303, no. 5663,
pp. 15321535, 2004.
[33] T. Saitoh, J. Komano, Y. Saitoh et al., Neutrophil extracellular
traps mediate a host defense response to human immunode-
ciency virus-1,Cell Host & Microbe, vol. 12, no. 1, pp. 109
116, 2012.
[34] V. Delgado-Rizo, M. A. Martínez-Guzmán, L. Iñiguez-Gutier-
rez, A. García-Orozco, A. Alvarado-Navarro, and M. Fafutis-
Morris, Neutrophil extracellular traps and its implications
in inammation: an overview,Frontiers in Immunology,
vol. 8, p. 81, 2017.
[35] O. Tatsiy and P. P. McDonald, Physiological stimuli induce
PAD4-dependent, ROS-independent NETosis, with early and
late events controlled by discrete signaling pathways,Fron-
tiers in Immunology, vol. 9, article 2036, 2018.
[36] S. Youse, D. Stojkov, N. Germic et al., Untangling NETosis
from NETs,European Journal of Immunology, vol. 49, no. 2,
pp. 221227, 2019.
[37] T. A. Fuchs, U. Abed, C. Goosmann et al., Novel cell death
program leads to neutrophil extracellular traps,The Journal
of Cell Biology, vol. 176, no. 2, pp. 231241, 2007.
[38] V. Papayannopoulos, K. D. Metzler, A. Hakkim, and
A. Zychlinsky, Neutrophil elastase and myeloperoxidase
regulate the formation of neutrophil extracellular traps,
The Journal of Cell Biology, vol. 191, no. 3, pp. 677691,
2010.
[39] F. Grabcanovic-Musija, A. Obermayer, W. Stoiber et al.,
Neutrophil extracellular trap (NET) formation charac-
terises stable and exacerbated COPD and correlates with
airow limitation,Respiratory Research, vol. 16, no. 1,
p. 59, 2015.
[40] C. Mikacenic, R. Moore, V. Dmyterko et al., Neutrophil extra-
cellular traps (NETs) are increased in the alveolar spaces of
patients with ventilator-associated pneumonia,Critical Care,
vol. 22, no. 1, p. 358, 2018.
[41] I. Bendib, L. de Chaisemartin, V. Granger et al., Neutrophil
extracellular traps are elevated in patients with pneumonia-
related acute respiratory distress syndrome,Anesthesiology,
vol. 130, no. 4, pp. 581591, 2019.
[42] J. J. M. Wong, J. Y. Leong, J. H. Lee, S. Albani, and J. G. Yeo,
Insights into the immuno-pathogenesis of acute respiratory
distress syndrome,Annals of Translational Medicine, vol. 7,
no. 19, p. 504, 2019.
[43] L. Zhu, L. Liu, Y. Zhang et al., High level of neutrophil extra-
cellular traps correlates with poor prognosis of severe inu-
enza A Infection,The Journal of Infectious Diseases,
vol. 217, no. 3, pp. 428437, 2018.
[44] T. Narasaraju, E. Yang, R. P. Samy et al., Excessive neutro-
phils and neutrophil extracellular traps contribute to acute
lung injury of inuenza pneumonitis,The American Journal
of Pathology, vol. 179, no. 1, pp. 199210, 2011.
[45] Y. Zuo, M. Zuo, S. Yalavarthi et al., Neutrophil extracellular
traps and thrombosis in COVID-19,Journal of Thrombosis
and Thrombolysis, vol. 1-8, 2020.
6 Mediators of Inammation
[46] P. Skendros, A. Mitsios, A. Chrysanthopoulou et al., Comple-
ment and tissue factor-enriched neutrophil extracellular traps
are key drivers in COVID-19 immunothrombosis,The Jour-
nal of Clinical Investigation, vol. 130, no. 11, pp. 61516157,
2020.
[47] L. Nicolai, A. Leunig, S. Brambs et al., Immunothrombotic
dysregulation in COVID-19 pneumonia is associated with
respiratory failure and coagulopathy,Circulation, vol. 142,
no. 12, pp. 11761189, 2020.
[48] E. A. Middleton, X.-Y. He, F. Denorme et al., Neutrophil
extracellular traps contribute to immunothrombosis in
COVID-19 acute respiratory distress syndrome,Blood,
vol. 136, no. 10, pp. 11691179, 2020.
[49] H. Domon, K. Nagai, T. Maekawa et al., Neutrophil elastase
subverts the immune response by cleaving toll-like receptors
and cytokines in pneumococcal pneumonia,Frontiers in
Immunology, vol. 9, p. 732, 2018.
[50] W.-J. Guan, Z.-Y. Ni, Y. Hu et al., Clinical characteristics of
coronavirus disease 2019 in China,The New England Journal
of Medicine, vol. 382, no. 18, pp. 17081720, 2020.
[51] F. P. Veras, M. Pontelli, C. Silva et al., SARS-CoV-2triggered
neutrophil extracellular traps mediate COVID-19 pathology,
The Journal of Experimental Medicine, vol. 217, no. 12, article
e20201129, 2020.
[52] Y. Zuo, S. Yalavarthi, H. Shi et al., Neutrophil extracellular
traps in COVID-19,JCI Insight, vol. 5, no. 11, article
e138999, 2020.
[53] G. A. Funchal, N. Jaeger, R. S. Czepielewski et al., Respira-
tory syncytial virus fusion protein promotes TLR-4-
dependent neutrophil extracellular trap formation by human
neutrophils,PLoS One, vol. 10, no. 4, article e0124082,
2015.
[54] L. A. Perrone, J. K. Plowden, A. García-Sastre, J. M. Katz, and
T. M. Tumpey, H5N1 and 1918 pandemic inuenza virus
infection results in early and excessive inltration of macro-
phages and neutrophils in the lungs of mice,PLoS Pathogens,
vol. 4, no. 8, article e1000115, 2008.
[55] S. P. Muraro, G. F. De Souza, S. W. Gallo et al., Respiratory
syncytial virus induces the classical ROS-dependent NETosis
through PAD-4 and necroptosis pathways activation,Scien-
tic Reports,vol. 8, no. 1, article 14166, 2018.
[56] S. K. Jorch and P. Kubes, An emerging role for neutrophil
extracellular traps in noninfectious disease,Nature Medicine,
vol. 23, no. 3, pp. 279287, 2017.
[57] R. O. Bonow, G. C. Fonarow, P. T. OGara, and C. W. Yancy,
Association of coronavirus disease 2019 (COVID-19) with
myocardial injury and mortality,JAMA Cardiology, vol. 5,
no. 7, pp. 751753, 2020.
[58] N. Chen, M. Zhou, X. Dong et al., Epidemiological and clini-
cal characteristics of 99 cases of 2019 novel coronavirus pneu-
monia in Wuhan, China: a descriptive study,The Lancet,
vol. 395, no. 10223, pp. 507513, 2020.
[59] S. Cicco, G. Cicco, V. Racanelli, and A. Vacca, Neutrophil
extracellular traps (NETs) and damage-associated molecular
patterns (DAMPs): two potential targets for COVID-19 treat-
ment,Mediators of Inammation, vol. 2020, Article ID
7527953, 25 pages, 2020.
[60] J. J. Hu, X. Liu, S. Xia et al., FDA-approved disulram
inhibits pyroptosis by blocking gasdermin D pore forma-
tion,Nature Immunology, vol. 21, no. 7, pp. 736745,
2020.
[61] C. C. Yost, H. Schwertz, M. J. Cody et al., Neonatal NET-
inhibitory factor and related peptides inhibit neutrophil extra-
cellular trap formation,The Journal of Clinical Investigation,
vol. 126, no. 10, pp. 37833798, 2016.
[62] T. Tagami, R. Tosa, M. Omura et al., Eect of a selective neu-
trophil elastase inhibitor on mortality and ventilator-free days
in patients with increased extravascular lung water: a post hoc
analysis of the PiCCO Pulmonary Edema Study,Journal of
Intensive Care, vol. 2, no. 1, p. 67, 2014.
7Mediators of Inammation
... Inhibiting or enhancing chemokines and chemokine receptor expression or activity are important approaches for studying and finding new treatment options for inflammatory diseases mainly by controlling cell migration (39)(40)(41). The massive accumulation of activated leukocytes, including neutrophils, in the lungs signifies a deterioration in the clinical prognosis of individuals affected by COVID-19 (42). Currently, different studies have demonstrated that targeting the chemokine system and neutrophil migration and activation in models of betacoronavirus infection reduces the overall inflammatory response (43). ...
Article
Full-text available
Introduction Pulmonary diseases represent a significant burden to patients and the healthcare system and are one of the leading causes of mortality worldwide. Particularly, the COVID-19 pandemic has had a profound global impact, affecting public health, economies, and daily life. While the peak of the crisis has subsided, the global number of reported COVID-19 cases remains significantly high, according to medical agencies around the world. Furthermore, despite the success of vaccines in reducing the number of deaths caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), there remains a gap in the treatment of the disease, especially in addressing uncontrolled inflammation. The massive recruitment of leukocytes to lung tissue and alveoli is a hallmark factor in COVID-19, being essential for effectively responding to the pulmonary insult but also linked to inflammation and lung damage. In this context, mice models are a crucial tool, offering valuable insights into both the pathogenesis of the disease and potential therapeutic approaches. Methods Here, we investigated the anti-inflammatory effect of the glycosaminoglycan (GAG)-binding chemokine fragment CXCL9(74-103), a molecule that potentially decreases neutrophil transmigration by competing with chemokines for GAG-binding sites, in two models of pneumonia caused by coronavirus infection. Results In a murine model of betacoronavirus MHV-3 infection, the treatment with CXCL9(74-103) decreased the accumulation of total leukocytes, mainly neutrophils, to the alveolar space and improved several parameters of lung dysfunction 3 days after infection. Additionally, this treatment also reduced the lung damage. In the SARS-CoV-2 model in K18-hACE2-mice, CXCL9(74-103) significantly improved the clinical manifestations of the disease, reducing pulmonary damage and decreasing viral titers in the lungs. Discussion These findings indicate that CXCL9(74-103) resulted in highly favorable outcomes in controlling pneumonia caused by coronavirus, as it effectively diminishes the clinical consequences of the infections and reduces both local and systemic inflammation.
... The host response to SARS-CoV-2 infection, involving both innate and adaptive immune system cells, mainly changes the production of lymphocytes, with more apoptosis, reflecting lymphopenia in most patients, as these types of cells are primarily affected 25 . ...
... By examining total cells, neutrophils, and neutrophil elastase activity in BALF, it was initially confirmed that mPEG-SPA-MDSPI16 could reduce the number of neutrophils at the site of inflammation by inhibiting the release of elastase, which indicates that mPEG-SPA-MDSPI16 can inhibit the migration of neutrophils to lung tissue and reduce inflammatory damage in lung tissue. Furthermore, studies have confirmed that excessive release of cytokines by activated neutrophils can cause tissue damage and that protease inhibitors can reduce the inflammatory response of cells by inhibiting the release of cytokines [19]. For example, urinary trypsin inhibitors exert clinical effects on ARDS patients by inhibiting the release of cytokines, such as IL-8 and TNF-α [20]. ...
Article
Full-text available
Anti-inflammatory drugs have become the second-largest class of common drugs after anti-infective drugs in animal clinical care worldwide and are often combined with other drugs to treat fever and viral diseases caused by various factors. In our previous study, a novel serine protease inhibitor-encoding gene (MDSPI16) with improved anti-inflammatory activity was selected from a constructed suppressive subducted hybridization library of housefly larvae. This protein could easily induce an immune response in animals and had a short half-life, which limited its wide application in the clinic. Thus, in this study, mPEG-succinimidyl propionate (mPEG-SPA, Mw = 5 kDa) was used to molecularly modify the MDSPI16 protein, and the modified product mPEG-SPA-MDSPI16, which strongly inhibited elastase production, was purified. It had good stability and safety, low immunogenicity, and a long half-life, and the IC50 for elastase was 86 nM. mPEG-SPA-MDSPI16 effectively inhibited the expression of neutrophil elastase and decreased ROS levels. Moreover, mPEG-SPA-MDSPI16 exerted anti-inflammatory effects by inhibiting activation of the NF-κB signaling pathway and the MAPK signaling pathway in neutrophils. It also exerted therapeutic effects on a lipopolysaccharide (LPS)-induced acute lung injury (ALI) mouse model. In summary, mPEG-SPA-MDSPI16 is a novel anti-inflammatory protein modified with PEG that has the advantages of safety, nontoxicity, improved stability, and strong anti-inflammatory activity in vivo and in vitro and is expected to become an effective anti-inflammatory drug.
... Neutrophils, as the main players in innate immunity, are recruited first during viral infection to engulf and clear pathogens. It also produces neutrophil extracellular traps and cytokines that recruit other immune cells to participate in adaptive immune processes while limiting or inactivating the virus 25 . The study of COVID-19 patients by Barnes et al. also showed high infiltration of neutrophils 26 . ...
Article
Full-text available
The death of coronavirus disease 2019 (COVID-19) is primarily due to from critically ill patients, especially from ARDS complications caused by SARS-CoV-2. Therefore, it is essential to contribute an in-depth understanding of the pathogenesis of the disease and to identify biomarkers for predicting critically ill patients at the molecular level. Immunogenic cell death (ICD), as a specific variant of regulatory cell death driven by stress, can induce adaptive immune responses against cell death antigens in the host. Studies have confirmed that both innate and adaptive immune pathways are involved in the pathogenesis of SARS-CoV-2 infection. However, the role of ICD in the pathogenesis of severe COVID-19 has rarely been explored. In this study, we systematically evaluated the role of ICD-related genes in COVID-19. We conducted consensus clustering, immune infiltration analysis, and functional enrichment analysis based on ICD differentially expressed genes. The results showed that immune infiltration characteristics were altered in severe and non-severe COVID-19. In addition, we used multiple machine learning methods to screen for five risk genes (KLF5, NSUN7, APH1B, GRB10 and CD4), which are used to predict COVID-19 severity. Finally, we constructed a nomogram to predict the risk of severe COVID-19 based on the classification and recognition model, and validated the model with external data sets. This study provides a valuable direction for the exploration of the pathogenesis and progress of COVID-19, and helps in the early identification of severe cases of COVID-19 to reduce mortality.
... In our case, antibiotics were used based on clinical appearance and the severity of the disease, the values of the inflammation biomarkers and according to the changes the doctors noticed based on the radiologic images. Considering the role of white blood cells in inflammation and infection, especially that of the neutrophiles and also the significant increase of CRP [14][15][16] , it was the most common laboratory exam performed on patients with COVID-19. Indeed, in our case, 99% of the patients had high number of neutrophiles and 56% of the data collected included high levels of CRP. ...
Article
Full-text available
Introduction: Throughout the COVID-19 pandemic, there has been a notable escalation in the administration of antibiotics among hospitalized individuals on a global scale. This trend holds the potential to exacerbate the prevailing issue of antibiotic resistance on a worldwide level. Purpose: The overarching purpose of this paper was to analyze the use of antibiotics in patients hospitalized with COVID-19 at a local hospital in a city of Kosovo during the period October 2020 to January 2021. Methodology: The work retrospectively analyzes the data of patients treated with COVID-19 at a Local Hospital in a city of Kosovo in the internal ward and included 300 patients, where the health records of hospitalized patients were used. Results: Based on the collected data, it appears that the most used empiric antibiotic in the ward was Levofloxacin and Ceftriaxone, the duration of the use of antibiotics per day was different in patients hospitalized with COVID-19, starting with Levofloxacin that was given every 24 hours, Imipenem every 12 hours and Ceftriaxone every 8 hours. Reasons for the use of antibiotics were: Disease burden, markers of inflammation, and radiological changes. According to the age group, the most infected group included the age between 51-70 years old with 44% and the smallest percentage was of the age group between 91-100 years old with 2%. Conclusions: The escalation in antibiotic usage among hospitalized patients during the pandemic has also impacted our nation, Kosovo, where such medications were consistently prescribed since the onset of the pandemic. Our observations reveal that Imipenem, Levofloxacin, and Ceftriaxone emerged as the most frequently utilized antibiotics.
... (13) This exaggerated immune response also leads to an increase in neutrophil production in COVID-19 infection, especially in more severe cases complicated with a secondary bacterial infection. (14,15) Thrombocytopenia is another important laboratory feature in COVID-19 patients, encountered in 20%-55% of cases and related to the severity of the disease. (16) The low platelet count is explained by different mechanisms, such as a decrease in platelet production either from SARS-CoV-2 infection myelosuppression or a strong inflammatory response, (17,18) platelet consumption in microthrombi induced from endothelial damage due to the potent COVID-19 inflammatory response, (19) or direct destruction of platelets by the immune system, as in an immune thrombocytopenic purpura-like state. ...
Article
COVID-19 is known to cause many hematological abnormalities, such as thrombocytopenia, leucopenia, and lymphopenia. Pancytopenia, a decrease in all peripheral blood cell lines, is a rare complication not commonly seen in patients with COVID-19. We report a case of a patient who experienced COVID-19 infection with mild clinical symptoms like fever, fatigue, and muscle and bone aches. The laboratory examinations revealed pancytopenia, mainly neutropenia, thrombocytopenia, mild anemia, and relative lymphocytosis, which persisted after infection resolution. The splenomegaly in abdominal echography and the characteristics of lymphocyte elements in peripheral blood smear examination raised suspicion of the presence of hairy cell leukemia (HCL); therefore, the patient was further examined with a bone marrow biopsy, which confirmed the diagnosis. The persistence of pancytopenia after recovery from COVID-19 infection, especially in patients with splenomegaly, should raise suspicion of another hematological coexistence diagnosis like HCL.
... Similarly, the level of NLR was not correlated with the risk of respiratory failure and cardiovascular death, indicating that when patients with the co-presentation of CAD and COVID-19, the NLR's assessment of the risk of all-cause death, including cardiovascular death and respiratory failure death, was not as strong as when CAD or COVID-19 exist alone. Part of the explanation may be attributed to the decreasing trend in lymphocyte count in COVID-19 infected patients, 22,23 and the recent high attention to the neutrophil extracellular traps hypothesis also suggested that high expression of neutrophils was a common phenomenon in COVID-19 infection, [24][25][26] where the NLR is elevated in all COVID-19 patients. Therefore, the generalization of high inflammatory status in these patients masks the risk of lowgrade inflammatory changes brought about by coronary artery disease. ...
Article
Full-text available
Aim The purpose of this study was to investigate the role of neutrophil-lymphocyte ratio (NLR), C-reactive protein-albumin ratio (CAR), and platelet-lymphocyte ratio (PLR) in the prognosis of patients with coronary artery disease (CAD) complicated with coronavirus disease 2019 (COVID-19). Methods This study included 265 patients. A receiver operating characteristic (ROC) curve analysis was performed to preliminarily evaluate the predictive ability of NLR, CAR, and PLR for all-cause death. The primary outcome was all-cause death during hospitalization, while the secondary outcomes were cardiovascular death and respiratory failure death. The Cox proportional hazard model with adjusted covariates was used to analyze the cumulative risk of outcomes. We also conducted subgroup analyses based on the acute and chronic characteristics of CAD. Propensity score matching (PSM) was used to further evaluate the robustness of the primary outcome. Results The ROC curve analysis results showed that the area under curve (AUC) values were 0.686 (95% CI 0.592–0.781, P<0.001) for NLR, 0.749 (95% CI 0.667–0.832, P<0.001) for CAR, and 0.571 (95% CI 0.455–0.687, P=0.232) for PLR. The Cox proportional hazard model showed that trends in NLR and PLR did not affect the risk of all-cause death (P=0.096 and P=0.544 for trend, respectively), but a higher CAR level corresponded to a higher risk of all-cause death (P<0.001 for trend). Similarly, The trends of NLR and PLR did not affect the risk of cardiovascular death and respiratory failure death, while a higher CAR level corresponded to a higher risk of cardiovascular death and respiratory failure death. The results of subgroup analyses and PSM were consistent with the total cohort. Conclusion In patients with CAD complicated with COVID-19, a higher CAR level corresponded to a higher risk of all-cause death, cardiovascular death, and respiratory failure death, while trends in NLR and PLR did not.
Article
Full-text available
Protein therapeutics offer high therapeutic potency and specificity; the broader adoptions and development of protein therapeutics, however, have been constricted by their intrinsic limitations such as inadequate stability, immunogenicity, suboptimal pharmacokinetics and biodistribution, and off‐target effects. This review describes a platform technology that formulates individual protein molecules with a thin formulation layer of crosslinked polymers, which confers the protein therapeutics with high activity, enhanced stability, controlled release capability, reduced immunogenicity, improved pharmacokinetics and biodistribution, and ability to cross the blood brain barriers. Based on currently approved protein therapeutics, this formulating platform affords the development of a vast family of superior protein therapeutics with improved efficacy and broadened indications at significantly reduced cost.
Article
Full-text available
COVID-19 has proven to be a disease that affects not only the respiratory tract but also leads to a state of generalized systemic hyperinflammation and overall immune dysregulation. An important role in its pathogenesis is the disturbance of many cytokines – a condition which, in its most pronounced form, is also called a „cytokine storm“. Objective To evaluate the serum cytokine levels during COVID-19 infection as potential biomarkers for the severity and course of infection. Materials and methods By design, the study is a retrospective cross-sectional, in which the serum concentrations of 10 pro- and anti-inflammatory cytokines (IFN-γ, IL-1β, IL-2, IL-4, IL-5, IL-6, IL-8, IL-10, TNF-α, and GM-CSF) were investigated in 36 individuals (divided into 3 groups by severity – with a mild form of the infection/presymptomatic, moderately severe and severe/critical) within two periods – before and after the second week from the onset of symptoms of the disease. Results In the period up to the 2nd week, the serum concentrations of IFN-γ (p = 0.029), IL-1β (p = 0.017), and IL-5 (p = 0.014) showed a statistically significant correlation with the disease severity, however in the later stage of the disease the cytokine levels did not show any clinical value. Conclusion Cytokine testing could be used to predict the severity of COVID-19 infection which could support individual therapeutic decisions. Analysis of a larger group of patients is needed to unfold the full potential of such testing.
Article
Full-text available
Studies of patients with COVID-19 have demonstrated markedly dysregulated coagulation and a high risk of morbid arterial and venous thrombotic events. Elevated levels of blood neutrophils and neutrophil extracellular traps (NETs) have recently been described in patients with COVID-19. However, their potential role in COVID-19-associated thrombosis remains incompletely understood. In order to elucidate the potential role of hyperactive neutrophils and NET release in COVID-19-associated thrombosis, we conducted a case–control study of patients hospitalized with COVID-19 who developed thrombosis, as compared with gender- and age-matched COVID-19 patients without clinical thrombosis. We found that remnants of NETs (cell-free DNA, myeloperoxidase-DNA complexes, and citrullinated histone H3) and neutrophil-derived S100A8/A9 (calprotectin) in patient sera were associated with higher risk of morbid thrombotic events in spite of prophylactic anticoagulation. These observations underscore the need for urgent investigation into the potential relationship between NETs and unrelenting thrombosis in COVID-19, as well as novel approaches for thrombosis prevention.
Article
Full-text available
The pandemic caused by the new coronavirus (SARS-Cov-2) has encouraged numerous in vitro studies and clinical trials around the world, with research groups testing existing drugs, novel drug candidates and vaccines that can prevent or treat infection caused by this virus. The urgency for an effective therapy is justified by the easy and fast viral transmission and the high number of patients with severe respiratory distress syndrome who have increasingly occupied intensive care hospital beds, leading to a collapse in health systems in several countries. However, to date, there is no sufficient evidence of the effectiveness of any researched therapy. The off-label or compassionate use of some drugs by health professionals is a reality in all continents, whose permission by regulatory agencies has been based on the results of some clinical trials. In order to guide decision-making for the treatment of COVID-19, this review aims to present studies and guidelines on the main therapies that have been and are currently being tested against SARS-CoV-2 and to critically analyze the reported evidences.
Article
Full-text available
Severe COVID-19 patients develop acute respiratory distress syndrome that may progress to cytokine storm syndrome, organ dysfunction, and death. Considering that neutrophil extracellular traps (NETs) have been described as important mediators of tissue damage in inflammatory diseases, we investigated whether NETs would be involved in COVID-19 pathophysiology. A cohort of 32 hospitalized patients with a confirmed diagnosis of COVID-19 and healthy controls were enrolled. The concentration of NETs was augmented in plasma, tracheal aspirate, and lung autopsies tissues from COVID-19 patients, and their neutrophils released higher levels of NETs. Notably, we found that viable SARS-CoV-2 can directly induce the release of NETs by healthy neutrophils. Mechanistically, NETs triggered by SARS-CoV-2 depend on angiotensin-converting enzyme 2, serine protease, virus replication, and PAD-4. Finally, NETs released by SARS-CoV-2-activated neutrophils promote lung epithelial cell death in vitro. These results unravel a possible detrimental role of NETs in the pathophysiology of COVID-19. Therefore, the inhibition of NETs represents a potential therapeutic target for COVID-19. © 2020 Veras et al. This article is distributed under the terms of an Attribution-Noncommercial-Share Alike-No Mirror Sites license for the first six months after the publication date (see http://www.rupress.org/terms/). After six months it is available under a Creative Commons License (Attribution-Noncommercial-Share Alike 4.0 International license, as described at https://creativecommons.org/licenses/by-nc-sa/4.0/).
Article
Full-text available
Emerging data indicate that complement and neutrophils contribute to the maladaptive immune response that fuels hyper-inflammation and thrombotic microangiopathy, thereby increasing COVID-19 mortality. Here, we investigated how complement interacts with the platelet/neutrophil extracellular traps (NETs)/thrombin axis, using COVID-19 specimens, cell-based inhibition studies and NETs/human aortic endothelial cell (HAEC) co-cultures. Increased plasma levels of NETs, tissue factor (TF) activity and sC5b-9 were detected in patients. Neutrophils of patients yielded high TF expression and released NETs carrying active TF. Treatment of control neutrophils with COVID-19 platelet-rich plasma generated TF-bearing NETs that induced thrombotic activity of HAEC. Thrombin or NETosis inhibition or C5aR1 blockade attenuated platelet-mediated NET-driven thrombogenicity. COVID-19 serum induced complement activation in vitro, consistent with high complement activity in clinical samples. Complement C3 inhibition with compstatin Cp40 disrupted TF expression in neutrophils. In conclusion, we provide a mechanistic basis for a pivotal role of complement and NETs in COVID-19 immunothrombosis. This study supports strategies against SARS-CoV-2 that exploit complement or NETosis inhibition.
Article
Full-text available
Background: SARS-CoV-2 infection causes severe pneumonia (COVID-19), but the mechanisms of subsequent respiratory failure and complicating renal and myocardial involvement are poorly understood. In addition, a systemic prothrombotic phenotype has been reported in COVID-19 patients. Methods: A total of 62 subjects were included in our study (n=38 patients with RT-PCR confirmed COVID-19 and n=24 non-COVID-19 controls). We performed histopathological assessment of autopsy cases, surface-marker based phenotyping of neutrophils and platelets, and functional assays for platelet, neutrophil functions as well as coagulation tests. Results: We provide evidence that organ involvement and prothrombotic features in COVID-19 are linked by immunothrombosis. We show that in COVID-19 inflammatory microvascular thrombi are present in the lung, kidney, and heart, containing neutrophil extracellular traps associated with platelets and fibrin. COVID-19 patients also present with neutrophil-platelet aggregates and a distinct neutrophil and platelet activation pattern in blood, which changes with disease severity. Whereas cases of intermediate severity show an exhausted platelet and hyporeactive neutrophil phenotype, severely affected COVID-19 patients are characterized by excessive platelet and neutrophil activation compared to healthy controls and non-COVID-19 pneumonia. Dysregulated immunothrombosis in SARS-CoV-2 pneumonia is linked to both ARDS and systemic hypercoagulability. Conclusions: Taken together, our data point to immunothrombotic dysregulation as a key marker of disease severity in COVID-19. Further work is necessary to determine the role of immunothrombosis in COVID-19.
Article
Full-text available
COVID-19 is a pandemic disease caused by the new coronavirus SARS-CoV-2 that mostly affects the respiratory system. The consequent inflammation is not able to clear viruses. The persistent excessive inflammatory response can build up a clinical picture that is very difficult to manage and potentially fatal. Modulating the immune response plays a key role in fighting the disease. One of the main defence systems is the activation of neutrophils that release neutrophil extracellular traps (NETs) under the stimulus of autophagy. Various molecules can induce NETosis and autophagy; some potent activators are damage-associated molecular patterns (DAMPs) and, in particular, the high-mobility group box 1 (HMGB1). This molecule is released by damaged lung cells and can induce a robust innate immunity response. The increase in HMGB1 and NETosis could lead to sustained inflammation due to SARS-CoV-2 infection. Therefore, blocking these molecules might be useful in COVID-19 treatment and should be further studied in the context of targeted therapy.
Article
Full-text available
COVID-19 affects millions of patients worldwide with clinical presentation ranging from isolated thrombosis to acute respiratory distress syndrome (ARDS) requiring ventilator support. Neutrophil extracellular traps (NETs) originate from decondensed chromatin released to immobilize pathogens and can trigger immunothrombosis. We studied the connection between NETs and COVID-19 severity and progression. We conducted a prospective cohort study of COVID-19 patients (n=33) with age- and sex-matched controls (n=17). We measured plasma myeloperoxidase (MPO)-DNA complexes (NETs), Platelet Factor 4, RANTES, and selected cytokines. Three COVID-19 lung autopsies were examined for NETs and platelet involvement. We assessed NET formation ex vivo in COVID-19 neutrophils and in healthy neutrophils incubated with COVID-19 plasma. We also tested the ability of neonatal NET-Inhibitory Factor (nNIF) to block NET formation induced by COVID-19 plasma. Plasma MPO-DNA complexes increased in COVID-19 with intubation (P<0.0001) and death as outcome (P<0.0005). Illness severity correlated directly with plasma MPO-DNA complexes (P=0.0360), while PaO2/FiO2 correlated inversely(P=0.0340). Soluble and cellular factors triggering NETs were significantly increased in COVID-19 and pulmonary autopsies confirmed NET-containing microthrombi with neutrophil-platelet infiltration. Finally, COVID-19 neutrophils ex vivo displayed excessive NETs at baseline and COVID-19 plasma triggered NET formation which was blocked by nNIF. Thus, NETs triggering immunothrombosis may, in part, explain the prothrombotic clinical presentations in COVID-19 and NETs may represent targets for therapeutic intervention.
Article
This review presents literature that highlights saliva’s utility as a biofluid in the diagnosis and monitoring of COVID-19. A systematic search was performed in 5 electronic databases (PubMed, Embase, LILACS, Scopus, and Web of Science). Studies were eligible for inclusion if they assessed the potential diagnostic value and/or other discriminatory properties of biological markers in the saliva of patients with COVID-19. As of July 22, 2020, a total of 28 studies have investigated the presence of SARS-CoV-2 RNA in saliva. Several of those studies confirmed reliable detection of SARS-CoV-2 in the saliva of patients with COVID-19. Saliva offered sensitivity and specificity for SARS-CoV-2 detection comparable to that of the current standard of nasopharyngeal and throat swabs. However, the utility of saliva in diagnosing COVID-19 infection remains understudied. Clinical studies with larger patient populations that measure recordings at different stages during the disease are still necessary to confirm the accuracy of COVID-19 diagnosis with saliva. Nevertheless, the utility of saliva as a diagnostic tool opens the possibility of using rapid and less invasive diagnostic strategies by targeting bioanalytes rather than the pathogen.
Article
The world is combating a common and invisible enemy severe acute respiratory syndrome coronavirus 2 (SARS-CoV2), a highly transmissible virus responsible for serious respiratory illness coronavirus disease-2019 (COVID-19). As with all respiratory viruses, public health measures are focused on contact tracing, isolation, and treatment of affected individuals, who have respiratory symptoms. However, it is spreading efficiently, and it can be explained from its stealth transmission from presymptomatic and asymptomatic individuals. Droplet and contact precautions are followed universally. Healthcare workers are at higher risk of acquiring infection and they are additionally required to follow airborne and eye protection. Recent studies indicate viral particles can be isolated from many body fluids including feces, saliva, semen, and tears, suggesting transmission could be possibly occurring through some of these routes as well. We have done an evidence-based review of all potential modes of transmission and discussed preventive measures to stop the spread. There is an urgent need for educating the healthcare professionals, governments, and public regarding other potential modes of transmission. Strict preventive measures need to be used to stop the spread.