ArticlePDF Available

Preliminary Findings on the Association of the Lipid Peroxidation Product 4-Hydroxynonenal with the Lethal Outcome of Aggressive COVID-19

Authors:

Abstract

Major findings of the pilot study involving 21 critically ill patients during the week after admission to the critical care unit specialized for COVID-19 are presented. Fourteen patients have recovered, while seven passed away. There were no differences between them in respect to clinical or laboratory parameters monitored. However, protein adducts of the lipid peroxidation product 4-hydroxynonenal (HNE) were higher in the plasma of the deceased patients, while total antioxidant capacity was below the detection limit for the majority of sera samples in both groups. Moreover, levels of the HNE-protein adducts were constant in the plasma of the deceased patients, while in survivors, they have shown prominent and dynamic variations, suggesting that survivors had active oxidative stress response mechanisms reacting to COVID-19 aggression, which were not efficient in patients who died. Immunohistochemistry revealed the abundant presence of HNE-protein adducts in the lungs of deceased patients indicating that HNE is associated with the lethal outcome. It seems that HNE was spreading from the blood vessels more than being a consequence of pneumonia. Due to the limitations of the relatively small number of patients involved in this study, further research on HNE and antioxidants is needed. This might allow a better understanding of COVID-19 and options for utilizing antioxidants by personalized, integrative biomedicine approach to prevent the onset of HNE-mediated vitious circle of lipid peroxidation in patients with aggressive inflammatory diseases.
antioxidants
Article
Preliminary Findings on the Association of the Lipid
Peroxidation Product 4-Hydroxynonenal with the Lethal
Outcome of Aggressive COVID-19
Neven Žarkovi´c 1,* , Biserka Orehovec 2, Lidija Milkovi´c 1, Bruno Barši´c 2, Franz Tatzber 3,
Willibald Wonisch 3, Marko Tarle 2, Marta Kmet 2, Ana Matai´c 4, Antonia Jakovˇcevi´c 4, Tea Vukovi´c 1,
Danijela Tali´c 1, Georg Waeg 5, Ivica Lukši´c 2,6, Elzbieta Skrzydlewska 7and Kamelija Žarkovi´c 4,6


Citation: Žarkovi´c, N.; Orehovec, B.;
Milkovi´c, L.; Barši´c, B.; Tatzber, F.;
Wonisch, W.; Tarle, M.; Kmet, M.;
Matai´c, A.; Jakovˇcevi´c, A.; et al.
Preliminary Findings on the
Association of the Lipid Peroxidation
Product 4-Hydroxynonenal with the
Lethal Outcome of Aggressive
COVID-19. Antioxidants 2021,10,
1341. https://doi.org/10.3390/
antiox10091341
Academic Editors:
Dimitrios Kouretas,
Konstantinos Poulas,
Konstantinos Farsalinos and
Joanna Floros
Received: 15 July 2021
Accepted: 24 August 2021
Published: 25 August 2021
Publisher’s Note: MDPI stays neutral
with regard to jurisdictional claims in
published maps and institutional affil-
iations.
Copyright: © 2021 by the authors.
Licensee MDPI, Basel, Switzerland.
This article is an open access article
distributed under the terms and
conditions of the Creative Commons
Attribution (CC BY) license (https://
creativecommons.org/licenses/by/
4.0/).
1Laboratory for Oxidative Stress (LabOS), Ru ¯
der Boškovi´c Institute, 10000 Zagreb, Croatia;
Lidija.Milkovic@irb.hr (L.M.); Tea.Vukovic@irb.hr (T.V.); Danijela.Talic@irb.hr (D.T.)
2Clinical Hospital Dubrava, 10000 Zagreb, Croatia; biserka.orehovec@gmail.com (B.O.);
barsicbruno@gmail.com (B.B.); tarlemarko1@gmail.com (M.T.); zmarta.km@gmail.com (M.K.);
luksic.ivica@gmail.com (I.L.)
3
Omnignostica Ltd., 3421 Höflein an der Donau, Austria; franz@tatzber.at (F.T.); willi.wonisch@aon.at (W.W.)
4Clinical Hospital Centre Zagreb, 10000 Zagreb, Croatia; ana.mataic@gmail.com (A.M.);
antonia.jakovcevic@gmail.com (A.J.); kamelijazarkovic@gmail.com (K.Ž.)
5Institute of Molecular Biosciences, Karl Franzens University, 8010 Graz, Austria; georg.waeg@uni-graz.at
6University of Zagreb School of Medicine, 10000 Zagreb, Croatia
7
Department of Inorganic and Analytical Chemistry, Medical University of Bialystok, 15-089 Bialystok, Poland;
elzbieta.skrzydlewska@umb.edu.pl
*Correspondence: zarkovic@irb.hr; Tel.: +385-1-4571212
Abstract:
Major findings of the pilot study involving 21 critically ill patients during the week after
admission to the critical care unit specialized for COVID-19 are presented. Fourteen patients have
recovered, while seven passed away. There were no differences between them in respect to clinical
or laboratory parameters monitored. However, protein adducts of the lipid peroxidation product
4-hydroxynonenal (HNE) were higher in the plasma of the deceased patients, while total antioxidant
capacity was below the detection limit for the majority of sera samples in both groups. Moreover,
levels of the HNE-protein adducts were constant in the plasma of the deceased patients, while in
survivors, they have shown prominent and dynamic variations, suggesting that survivors had active
oxidative stress response mechanisms reacting to COVID-19 aggression, which were not efficient in
patients who died. Immunohistochemistry revealed the abundant presence of HNE-protein adducts
in the lungs of deceased patients indicating that HNE is associated with the lethal outcome. It seems
that HNE was spreading from the blood vessels more than being a consequence of pneumonia. Due
to the limitations of the relatively small number of patients involved in this study, further research on
HNE and antioxidants is needed. This might allow a better understanding of COVID-19 and options
for utilizing antioxidants by personalized, integrative biomedicine approach to prevent the onset of
HNE-mediated vitious circle of lipid peroxidation in patients with aggressive inflammatory diseases.
Keywords:
COVID-19; oxidative stress; lipid peroxidation; 4-hydroxynonenal (HNE); inflamma-
tion; antioxidants; peroxides; free radicals; reactive oxygen species (ROS); blood vessels; lungs;
immunohistochemistry; HNE-ELISA
1. Introduction
First cases of coronavirus disease 2019 (COVID-19), caused by severe acute respiratory
syndrome coronavirus 2 (SARS-CoV-2) infection, were reported at the very end of the year
2019. In 2020, the virus has spread worldwide, causing pandemics as never seen before.
Since the COVID-19 outbreak, scientists are trying to understand the pathology of the
disease in order to improve the patient’s outcome. While only a few drugs were approved
Antioxidants 2021,10, 1341. https://doi.org/10.3390/antiox10091341 https://www.mdpi.com/journal/antioxidants
Antioxidants 2021,10, 1341 2 of 10
by the Food and Drug Administration as appropriate to treat patients with COVID-19, the
first vaccines are implemented worldwide according to the urgent criteria, occasionally
causing inexplicable side effects. Major complications of severe COVID-19 infection include
acute respiratory distress syndrome (ARDS), sepsis, and multiple organ dysfunction or
failure (MOF) [
1
]. Each of these severe disorders is associated with inflammation and
excessive generation of reactive oxygen species (ROS), affecting body redox balance, thus
causing oxidative damage of macromolecules under the pathophysiological process of
oxidative stress (OS). Therefore, the potential use of antioxidant treatments to prevent
COVID-19 progression have been already suggested [24].
One of the underlying mechanisms of organ damage in COVID-19 could be mitochon-
drial dysfunctions resulting in excessive production of ROS, triggered by inflammatory
response to SARS-CoV-2 infection, hence resembling other types of sepsis characterized by
severe inflammation [
5
]. There are various pathophysiological sources of OS, causing either
acute or chronic disbalance of pro- and anti-oxidants, many of which do not need to result
in severe health disorders unless they induce lipid peroxidation (LPO). Namely, under such
aggressive OS, polyunsaturated fatty acids (PUFAs, notably
ω
-6 fatty acids or n-6 fatty
acids, especially arachidonic and linoleic acid) in cellular biomembranes become targets for
ROS-induced damage, triggering a self-catalyzed, non-enzymatic chain reaction of LPO.
Thus, damaged cells in the affected tissues, together with erythrocytes in blood, generate
ROS, and release of free iron, which can consequently amplify LPO through Fenton reac-
tions and promote LPO and ferroptosis [
6
], resulting in membrane dysfunction/destruction
and MOF in COVID-19 patients [
7
]. Indeed, the autopsy findings in COVID-19 patients
revealed a ferroptosis signature in the epicardium and myocardium in the area of severe
SARS-CoV-2 myocarditis associated with generalized lipid peroxidation [
8
]. Similarly, a
pilot study on oxidative stress in COVID-19 patients hospitalized in an intensive care unit
(ICU) for severe pneumonia has indicated systemic OS in critically ill COVID-19 patients
manifested by increased LPO and deficits of antioxidants [9].
Final products of LPO are reactive aldehydes that have a longer lifetime (minutes
to hours) than is the lifetime of ROS (nano to milliseconds) and might accordingly rep-
resent better biomarkers of SARS-CoV-2 induced OS. Among the LPO-derived reactive
aldehydes, the most bioactive seems to be 4-hydroxynonenal (HNE). Namely, HNE acts
as “second messenger of free radicals”, spreading OS and LPO even in the absence of
ROS, accumulating in blood vessels in an age-dependent manner [
10
,
11
]. The affinity of
HNE to bind to proteins, modifying their structure and function and acting dynamically
in a concentration-dependent manner not only as a cytotoxic product of LPO, but also
as a signaling, regulatory molecule, makes HNE a valuable bioactive marker for various
diseases associated with OS [
12
,
13
] and their respective therapies [
14
], suggesting it could
also be a predictive biomarker of SARS-CoV-2 induced OS. In favor of our assumption
are recent autopsy findings of HNE in COVID-19 patient [
8
] and a positive correlation
of HNE expression with the functional receptor of SARS-CoV-2, angiotensin-converting
enzyme 2 (ACE2), also suggesting significant involvement of this particular LPO product
in COVID-19 pathogenesis [15].
Therefore, to test our hypothesis that the onset of the vicious circle of LPO mediated by
HNE might be relevant for the pathogenesis of COVID-19 and its outcome, we used genuine
enzyme-linked immunosorbent assay (ELISA) specific for the HNE-protein adducts in
the blood of critically ill patients who survived and those how were killed by aggressive
COVID-19. The same monoclonal antibody used for the HNE-ELISA was eventually used
for immunohistochemistry of lungs of some deceased COVID-19 patients.
2. Materials and Methods
2.1. Patients
This study was done upon the ethical approval 2020-1012-13 of the Clinical Hospital
Dubrava in Zagreb, serving as the national center for COVID-19, thus, providing medical
care for patients suffering from the most aggressive COVID-19. Only patients who signed
Antioxidants 2021,10, 1341 3 of 10
the informed written consent and who were hospitalized at the ICU for the period long
enough to collect three samples of blood starting on the first day after admission, followed
by the blood sampling every second day afterward.
Of the 21 patients involved (11 men and 10 women of average age 65.3
±
14.6 years),
fourteen patients have recovered, while seven died. Although survivors were on the
average ten years younger than patients who died (62.0
±
14.7 years vs. 71.9
±
12.4),
there was no significant difference between them in respect to age (p> 0.1), comorbidities
(3 patients had diabetes mellitus type 2, one passed away, while two survived), clinical
treatment or laboratory parameters monitored.
2.2. Laboratory Analysis
For the analysis of biochemical and immunochemical parameters, samples of blood
were centrifuged at 1370
×
gfor 10 min and sera were analysed immediately after centrifu-
gation or were stored at 80 C to be analysed afterwards.
Leukocytes were counted in EDTA-anticoagulated blood on the DxH 800 Hematology
Analyzer (Beckman Coulter, Tokyo, Japan). Concentrations of C-reactive protein (CRP)
and ferritin were measured using immunoturbidimetric assays on the AU 5800 analyzer
(Beckman Coulter, Tokyo, Japan). Lactate dehydrogenase, EC 1.1.1.27 (LDH) activity was
measured using a kinetic UV test on AU 5800 analyzer (Beckman Coulter, Tokyo, Japan).
Levels of interleukin 6 (IL-6) and Procalcitonin (PCT) were measured using chemilumines-
cent immunoassays on the UniCel DxI 800 Access Immunoassay System (Beckman Coulter,
Tokyo, Japan).
An enzymatic assay was used to test ROS scavenging of sera using uric acid standards
as described before [
16
]. In parallel, a complementary assay for total serum peroxides was
done [
16
] using hydrogen peroxide standards. However, only results for serum peroxide
levels were obtained, because for the majority of sera samples, total antioxidant capacity
was below the detection limit equivalent to 11.7
µ
M of the uric acid standard. Sera samples
were also used to determine the titer of the autoantibodies against oxidized low-density
lipoproteins (LDL) using the oLAB ELISA as described before [17].
Parallel to the sera sampling, the EDTA plasma samples were prepared for the HNE-
ELISA analysis done as described before [
18
,
19
] using a genuine monoclonal antibody
specific for the HNE-histidine (HNE-His) epitopes.
2.3. Immunohistochemistry
Finally, immunohistochemical analysis using the same monoclonal antibody, as in the
case of the HNE-ELISA, was done for the tissue specimens obtained by autopsy of patients
that eventually passed away at the University Hospital for Infectious Diseases, Zagreb.
Formalin-fixed, paraffin-embedded tissue specimens of lungs were analyzed by pathol-
ogists with expertise in the field, as described before [
13
,
20
]. The 3,3
0
-Diaminobenzidine
tetrahydrochloride (DAB) was used as chromophore giving brown colored reaction in case
of positive immunohistochemical reaction for the HNE-His, with blue-colored contrast
staining of hematoxylin.
2.4. Statistics
Comparison of the mean values of the HNE-protein adducts in plasma between
the two groups of patients was carried out using the t-test, the differences between the
incidence of patients with static levels and those with dynamic changes of the HNE levels
were compared between survivors and the deceased patients by chi-square test, while
correlations between biomarkers analysed in comparison to the HNE levels in plasma were
done by the Pearson Rtest.
3. Results
The analysis of HNE levels in the EDTA-plasma of patients analyzed by the gen-
uine HNE-ELISA using a non-commercial monoclonal antibody specific for the HNE-His
Antioxidants 2021,10, 1341 4 of 10
epitopes revealed significantly higher (p< 0.05) average plasma values of HNE-protein
adducts in the blood of the deceased patients than in the blood of survivors for the first
days (1–3) in hospital (Figure 1).
Antioxidants 2021, 10, x FOR PEER REVIEW 4 of 10
correlations between biomarkers analysed in comparison to the HNE levels in plasma
were done by the Pearson R test.
3. Results
The analysis of HNE levels in the EDTA-plasma of patients analyzed by the genuine
HNE-ELISA using a non-commercial monoclonal antibody specific for the HNE-His
epitopes revealed significantly higher (p < 0.05) average plasma values of HNE-protein
adducts in the blood of the deceased patients than in the blood of survivors for the first
days (1–3) in hospital (Figure 1).
Figure 1. Values of the HNE-protein adducts determined in the EDTA-plasma of COVID-19 patients by the genuine HNE-
ELISA specific for the HNE-His adducts. The amounts of the HNE are presented in pmol/mg protein values, while the
asterisk indicates a significant difference (p < 0.05) between the average values determined for the plasma of survivors and
for the deceased COVID-19 patients.
However, values of HNE-His detected in the plasma of survivors slightly increased
on the fifth day, reaching the levels that were not significantly different from the values
of the deceased patients indicating that the high levels of the HNE-protein adducts in
plasma were not the only HNE-related parameter predicting the lethal outcome of aggres-
sive COVID-19. Namely, the individual levels of HNE determined at different time points
in the blood of patients have shown that the levels of HNE were stable in the blood of
patients who died, but have shown dynamic variations in the blood of survivors revealing
significantly different patterns between the two groups (chi-square p < 0.01). Namely, 6/7
patients who passed away did not show prominent variations of the HNE levels in the
Figure 1.
Values of the HNE-protein adducts determined in the EDTA-plasma of COVID-19 patients by the genuine
HNE-ELISA specific for the HNE-His adducts. The amounts of the HNE are presented in pmol/mg protein values, while
the asterisk indicates a significant difference (p< 0.05) between the average values determined for the plasma of survivors
and for the deceased COVID-19 patients.
However, values of HNE-His detected in the plasma of survivors slightly increased
on the fifth day, reaching the levels that were not significantly different from the values of
the deceased patients indicating that the high levels of the HNE-protein adducts in plasma
were not the only HNE-related parameter predicting the lethal outcome of aggressive
COVID-19. Namely, the individual levels of HNE determined at different time points
in the blood of patients have shown that the levels of HNE were stable in the blood of
patients who died, but have shown dynamic variations in the blood of survivors revealing
significantly different patterns between the two groups (chi-square p< 0.01). Namely,
6/7 patients who passed away did not show prominent variations of the HNE levels in the
blood collected at different time points (as an exemption, we consider the 3rd patient from
the left side of the respective upper graph, who had a transient decrease of HNE levels in
the blood on the third day), while relatively stable, although gradually decreasing levels of
HNE were observed only in 1/14 survivors (the 5th patient from the left side on the bottom
graph). Hence, these findings suggest that survivors had active oxidative stress response
mechanisms reacting to the COVID-19 aggression, which were not efficient in the patients
who died.
Antioxidants 2021,10, 1341 5 of 10
The individual variations of the HNE levels in the blood were compared using Pearson
correlation with all available physical and laboratory parameters of the patients, but due
to the relatively small number of patients per group, the R values were significant for the
deceased patients only for HNE and IL-6 (R-0.83, p< 0.05) on the first day after admission to
the ICU. Although trends of differentially positive vs. negative correlations were observed
at different time points for other parameters between survivors and the deceased patients,
due to the lack of statistical significance, these trends are not presented.
However, because of possible high importance, we wish to say that the persistent
negative correlation trend between the HNE levels in the blood and the levels of the au-
toantibodies against oxidized LDL (oLAB) was observed only in the blood of the deceased
patients, while for survivors significantly positive correlation (R 0.54, p< 0.05) was ob-
served on the last day of evaluation, followed by the recovery from COVID-9. Similarly,
a trend of negative correlation was also observed between the HNE levels and the total
peroxide levels in sera of the patients who died but were gradually lost in the terminal
stage (day 5), while such a trend was not observed for survivors.
It should be mentioned here that the complementary assay for the total antioxidant
capacity was also carried out, but the levels of antioxidants were too low to be determined
(bellow detection limit) for the majority of samples in both groups, suggesting again that
OS is one of the major pathogenic factors in aggressive COVID-19, for which patients
needed additional oxygen support at ICU.
Microphotographs presented in Figure 2show the immunohistochemical appearance
of HNE-His adducts in the lungs of the deceased COVID-19 patient.
Antioxidants 2021, 10, x FOR PEER REVIEW 5 of 10
blood collected at different time points (as an exemption, we consider the 3rd patient from
the left side of the respective upper graph, who had a transient decrease of HNE levels in
the blood on the third day), while relatively stable, although gradually decreasing levels
of HNE were observed only in 1/14 survivors (the 5th patient from the left side on the
bottom graph). Hence, these findings suggest that survivors had active oxidative stress
response mechanisms reacting to the COVID-19 aggression, which were not efficient in
the patients who died.
The individual variations of the HNE levels in the blood were compared using Pear-
son correlation with all available physical and laboratory parameters of the patients, but
due to the relatively small number of patients per group, the R values were significant for
the deceased patients only for HNE and IL-6 (R-0.83, p < 0.05) on the first day after admis-
sion to the ICU. Although trends of differentially positive vs. negative correlations were
observed at different time points for other parameters between survivors and the de-
ceased patients, due to the lack of statistical significance, these trends are not presented.
However, because of possible high importance, we wish to say that the persistent
negative correlation trend between the HNE levels in the blood and the levels of the au-
toantibodies against oxidized LDL (oLAB) was observed only in the blood of the deceased
patients, while for survivors significantly positive correlation (R 0.54, p < 0.05) was ob-
served on the last day of evaluation, followed by the recovery from COVID-9. Similarly,
a trend of negative correlation was also observed between the HNE levels and the total
peroxide levels in sera of the patients who died but were gradually lost in the terminal
stage (day 5), while such a trend was not observed for survivors.
It should be mentioned here that the complementary assay for the total antioxidant
capacity was also carried out, but the levels of antioxidants were too low to be determined
(bellow detection limit) for the majority of samples in both groups, suggesting again that
OS is one of the major pathogenic factors in aggressive COVID-19, for which patients
needed additional oxygen support at ICU.
Microphotographs presented in Figure 2 show the immunohistochemical appearance
of HNE-His adducts in the lungs of the deceased COVID-19 patient.
Figure 2. The immunohistochemical appearance of HNE in the lungs of the deceased COVID-19
patient. Eosinophilic (HE) liquid of edema in the alveoli (asterisks) shows abundant HNE content
(HNE-His giving brown-colored immunopositive reaction of DAB staining). However, inflamma-
tory mononuclear cells in the alveoli and in their septa (black arrows) are mostly negative for HNE
(only blue-colored hemotoxylin contrast staining is present), with some macrophages are positive
for HNE (yellow arrow). On the contrary, blood vessels (both their walls and the blood content,
indicated by the black stars) are HNE-positive, as are the hyaline membranes on the surface of the
alveolar septa (red arrows). Conclusion—HNE might be responsible for the fatal outcome, but more
Figure 2.
The immunohistochemical appearance of HNE in the lungs of the deceased COVID-19 patient. Eosinophilic (HE)
liquid of edema in the alveoli (asterisks) shows abundant HNE content (HNE-His giving brown-colored immunopositive
reaction of DAB staining). However, inflammatory mononuclear cells in the alveoli and in their septa (black arrows) are
mostly negative for HNE (only blue-colored hemotoxylin contrast staining is present), with some macrophages are positive
for HNE (yellow arrow). On the contrary, blood vessels (both their walls and the blood content, indicated by the black stars)
are HNE-positive, as are the hyaline membranes on the surface of the alveolar septa (red arrows). Conclusion—HNE might
be responsible for the fatal outcome, but more likely due to the systemic, vascular oxidative stress (sepsis-like, originating
from the blood), not due to the pulmonary inflammatory cells, which are mostly negative for HNE.
Antioxidants 2021,10, 1341 6 of 10
Immunohistochemistry was done by the same monoclonal antibody used for the
HNE-His ELISA thus detecting the same epitopes on the proteins modified by HNE due to
the onset of OS in the blood and in the lungs. As can be seen, the cell-free inflammatory
edema in the lungs contained abundant amounts of HNE-protein adducts. Similarly, blood
vessels were loaded with HNE, as were the hyaline membranes suggesting that abundant
HNE might be responsible for the vicious circle of OS in COVID-19, leading to the fatal
outcome. However, pulmonary OS was probably not caused by an oxidative burst of
inflammatory cells in the lungs, which were mostly negative for HNE, but was a crucial
component of systemic OS spreading HNE by blood through the entire organism.
4. Discussion
While excessive inflammation plays an important role in aggressive COVID-19, the
relevance of this generated oxidative stress is uncertain, mostly due to the short lifetime of
free radicals and the complexity of the disease itself [
21
23
]. On the other hand, reactive
aldehyde HNE, the particular LPO product of PUFAs, which regulates the sensitivity of cells
to OS and their adaptation acting as the second messenger of free radicals and a signaling
molecule, has a longer lifetime mostly because of high affinity for binding to proteins [
24
,
25
].
This pilot study revealed the abundant presence of the HNE-protein adducts in the lungs
of patients who died from aggressive COVID-19. The HNE-protein adducts were also
higher in the blood of these patients than in the blood of the equally-treated critically
ill patients who survived COVID-19 even five days before the death/recovery and were
correlated with the levels of the oxygen saturation, inflammatory parameters, and the
other parameters of oxidative stress analyzed. Due to the relatively small size of the group
of deceased patients (n= 7), the only significant correlation in the group of the deceased
patients was observed between HNE and IL-6 levels upon admission to the ICU and was
strongly negative (R-0.83). This is not surprising because HNE can act as a signaling
molecule suppressing IL-6 production [
26
], but it is worth mentioning that it was only
transient and was only observed for the patients who eventually died from COVID-19.
In spite of high levels of R, which were mostly not significant due to the relatively small
number of patients in the pilot study, obvious trends were observed showing a lot of
differences between patients who survived and patients who died, which deserve further
studies involving more patients.
Among other aspects of COVID-19 related OS and LPO further, studies should evalu-
ate the immune response to oxidatively modified proteins, as we have observed a trend of
negative correlation between HNE-protein adducts and the titers of autoimmune antibod-
ies generated against oxidized LDL, only in the patients who died. Although HNE-ELISA
determines mostly HNE bound to histidine residues of serum albumin, the assay also
detects HNE-His present on the other sera proteins [
19
,
27
]. Since HNE-His is also a major
epitope of the oxidized LDL [
27
], our results suggest the involvement of the immune sys-
tem in the fight with toxic mediators of OS, notably HNE in COVID-19 patients. Moreover,
the observed constantly negative trend of correlation between HNE-ELISA and oLAB
assays in non-survivors might be interpreted as exhaustion of the immune system, which
cannot produce specific anti-OS-IgG in sufficient amounts and should be further studied.
On the other hand, HNE is also binding to the histidine moiety of angiotensin [
27
], which,
together with the fact that HNE interferes with bioactivities of angiotensin [
28
,
29
] and is
positively correlated with the ACE2 [
15
], increase the need for further research on HNE in
the pathogenesis of COVID-19.
Immunohistochemistry revealed abundant HNE-protein adducts, especially in the
blood vessels and inflammatory edema of the lungs affected by COVID-19. Surprisingly,
inflammatory, mononuclear cells in the alveoli and in their septa were HNE-negative
in spite of the fact that for inflammatory cells, HNE acts as a signaling molecule and
pleiotropic regulatory factor, while macrophages, which were only exceptionally HNE-
positive, are otherwise known to generate ROS and abundant HNE [
30
,
31
]. These findings
suggest systemic, vascular oxidative stress in COVID-19 patients and the spread of HNE in
Antioxidants 2021,10, 1341 7 of 10
the form of protein adducts by blood. That is in agreement with findings of toxic HNE-
protein adducts accumulation in atherosclerotic blood vessels, proportional to the age, but
in potentially reversible form of histidine adducts [
11
,
32
,
33
]. Although in our pilot study
there was no significant difference between the age or patients who survived and those
who died, it should be mentioned that survivors were, on average, ten years younger. Our
previous research on the atherosclerotic aorta revealed that the accumulation of the HNE-
protein adducts reaches its maximum around the age of 65 [
11
], which was the average
age of our patients taken together. Hence, we can hypothesize that the age of patients
might play an important role in the survival of COVID-19 also due to the age-dependent
accumulation of HNE in the wall of blood vessels, while the possible release of toxic HNE
from the blood vessels into other tissues should be further studied, especially because
the similar accumulation of the blood-originating HNE was also revealed in abdominal
adipose tissue of obese people with metabolic syndrome [
34
]. This assumption deserves
further research, which could increase our knowledge on the oxidative stress response not
only to COVID-19 aggression but also about other stress- and age-associated diseases for
which HNE is known to be an important factor of pathogenesis or hormesis [3538].
Finally, it should be mentioned that trend of negative correlation of HNE in the
blood and the total serum peroxides observed in our study associated with undetectable
serum antioxidant capacity (according to the TAC assay) of COVID-19 patients resembles
findings of other studies and points to the high relevance of antioxidants in defense against
aggressive COVID-19. Among potential antioxidants present in tissues, including blood,
uric acid is often neglected, although it contributes a lot to the TAC analysis of sera, for
which we even used uric acid as standard. Although we did not evaluate urate levels
in the blood of our patients, further studies should do that with respect to the possible
onset of ischemic/reperfusion injury that might cause systemic oxidative stress generating
uric acid.
5. Conclusions
If further studies confirm our preliminary findings, novel options for the introduction
of an integrative biomedicine approach to COVID-19 and other severe inflammatory
diseases could be developed based on personalized medicine and the careful use of the most
promising antioxidants that could regulate HNE metabolism and its bioactivities. It is likely
that such options will include the use of lipid-soluble antioxidants to prevent the onset
of LPO, such as tocopherol and lycopene, but together with water-soluble antioxidants,
such as vitamin C, micronutrients needed to maintain physiological functions of enzymatic
antioxidants (such as zinc and selenium) and eventually HNE-scavengers resembling GSH—
the dominant physiological scavenger of HNE, such as those based on N-acetylcysteine,
together with other credible natural or synthetic regulators of OS and LPO in particular.
Author Contributions:
Conceptualization, N.Ž., K.Ž., B.O. and B.B.; methodology, L.M., T.V., D.T.,
M.K., M.T., G.W., A.J. and A.M.; software, L.M.; validation, F.T., W.W. and L.M.; formal analysis, N.Ž.,
E.S., I.L. and K.Ž.; investigation, B.B., M.T., B.O., M.K. and L.M.; resources, E.S., I.L., N.Ž., F.T. and
W.W.; data curation, N.Ž., B.O. and L.M.; writing—original draft preparation, N.Ž.; writing—review
and editing, N.Ž. L.M., F.T., B.B., B.O., W.W. and K.Ž.; visualization, N.Ž., K.Ž. and L.M.; supervision,
N.Ž. and K.Ž.; project administration, N.Ž., E.S., I.L. and K.Ž.; funding acquisition, E.S., N.Ž., I.L., F.T.
and W.W. All authors have read and agreed to the published version of the manuscript.
Funding:
This research was supported by the Polish National Agency for Academic Exchange NAWA
as a part of the International Academic Partnerships (PPI/APM/2018/00015/U/001) and by core
funding of the Rudjer Boskovic Institute.
Institutional Review Board Statement:
The study was conducted according to the guidelines of the
Declaration of Helsinki and approved by the Institutional Review Board of the Clinical Hospital
Dubrava in Zagreb by ethical approval 2020-1012-13.
Informed Consent Statement:
Informed consent was obtained from all subjects involved in the study.
Data Availability Statement: Data is contained within the article.
Antioxidants 2021,10, 1341 8 of 10
Antioxidants 2021,10, 1341 9 of 10
Acknowledgments:
Kind support in materials by Omnignostica is acknowledged. The panelist of
the HrZZ call IP-CORONA-2020-12 denied financial support for this research imposing its realization
by the pilot study presented. The authors are grateful to the reviewers for supportive, constructive,
and encouraging comments on this paper.
Conflicts of Interest: The authors declare no conflict of interest.
Abbreviations
Coronavirus disease 2019 (COVID-19); 4-Hydroxynonenal (HNE); severe acute respi-
ratory syndrome coronavirus 2 (SARS-CoV-2); acute respiratory distress syndrome (ARDS);
multiple organ dysfunction or failure (MOF); oxidative stress (OS); reactive oxygen species
(ROS); lipid peroxidation LPO); polyunsaturated fatty acids (PUFAs); low-density lipopro-
teins (LDL); enzyme-linked immunosorbent assay (ELISA); angiotensin-converting en-
zyme 2 (ACE2); HNE-histidine (HNE-His); intensive care unit (ICU); interleukin 6 (IL-6);
immunoglobulin G (IgG), hematoxylin/eosin staining (HE); 3,3
0
-Diaminobenzidine stain-
ing (DAB).
References
1.
Chen, N.; Zhou, M.; Dong, X.; Qu, J.; Gong, F.; Han, Y.; Qiu, Y.; Wang, J.; Liu, Y.; Wei, Y.; et al. Epidemiological and clinical
characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: A descriptive study. Lancet
2020
,395, 507–513.
[CrossRef]
2.
Beltrán-García, J.; Osca-Verdegal, R.; Pallardó, F.V.; Ferreres, J.; Rodríguez, M.; Mulet, S.; Carbonell, F.; García-Giménez, J.L.
Oxidative stress and inflammation in covid-19-associated sepsis: The potential role of anti-oxidant therapy in avoiding disease
progression. Antioxidants 2020,9, 936. [CrossRef]
3.
Moskowitz, D.W.; Sanchez-Gonzalez, M.; Marinelli, E.; Day, K. Quercetin for COVID19? Diabetes Complicat.
2020
,4, 1–2.
[CrossRef]
4.
Nasi, A.; McArdle, S.; Gaudernack, G.; Westmand, G.; Meliefe, C.; Rockberg, J.; Arens, R.; Kouretas, D.; Sjölin, J.; Mangsbo,
S. Reactive oxygen species as an initiator of toxic innate immune responses in retort to SARS-CoV-2 in an ageing population,
consider N-acetylcysteine as early therapeutic intervention. Toxicol. Rep. 2020,7, 768–771. [CrossRef] [PubMed]
5.
Nagar, H.; Piao, S.; Kim, C.-S. Role of mitochondrial oxidative stress in sepsis. Acute Crit. Care
2018
,33, 65–72. [CrossRef]
[PubMed]
6.
Cecchini, R.; Cecchini, A.L. SARS-CoV-2 infection pathogenesis is related to oxidative stress as a response to aggression. Med.
Hypotheses 2020,143, 110102. [CrossRef]
7.
Vlahakos, V.D.; Marathias, K.P.; Arkadopoulos, N.; Vlahakos, D.V. Hyperferritinemia in patients with COVID-19: An opportunity
for iron chelation? Artif. Organs 2020,45, 163–167. [CrossRef]
8.
Jacobs, W.; Lammens, M.; Kerckhofs, A.; Voets, E.; Van San, E.; Van Coillie, S.; Peleman, C.; Mergeay, M.; Sirimsi, S.; Matheeussen,
V.; et al. Fatal lymphocytic cardiac damage in coronavirus disease 2019 (COVID-19): Autopsy reveals a ferroptosis signature. ESC
Hear. Fail. 2020. [CrossRef]
9.
Pincemail, J.; Cavalier, E.; Charlier, C.; Cheramy–Bien, J.-P.; Brevers, E.; Courtois, A.; Fadeur, M.; Meziane, S.; Goff, C.; Misset, B.;
et al. Oxidative Stress Status in COVID-19 Patients Hospitalized in Intensive Care Unit for Severe Pneumonia. A Pilot Study.
Antioxidants 2021,10, 257. [CrossRef]
10.
Žarkovi´c, N. 4-Hydroxynonenal as a bioactive marker of pathophysiological processes. Mol. Asp. Med.
2003
,24, 281–291.
[CrossRef]
11.
Zarkovic, K.; Larroque-Cardoso, P.; Pucelle, M.; Salvayre, R.; Waeg, G.; Negre-Salvayre, A.; Zarkovic, N. Elastin aging and lipid
oxidation products in human aorta. Redox Biol. 2015,4, 109–117. [CrossRef] [PubMed]
12.
Frijhoff, J.; Winyard, P.G.; Zarkovic, N.; Davies, S.; Stocker, R.; Cheng, D.; Knight, A.R.; Taylor, E.L.; Oettrich, J.; Ruskovska, T.;
et al. Clinical Relevance of Biomarkers of Oxidative Stress. Antioxid. Redox Signal. 2015,23, 1144–1170. [CrossRef]
13.
Zarkovic, K.; Jakovcevic, A.; Zarkovic, N. Contribution of the HNE-Immunohistochemistry to Modern Pathological Concepts of
Major Human Diseases. Free Radic. Biol. Med. 2017,111, 110–125. [CrossRef]
14.
Jaganjac, M.; Milkovic, L.; Gegotek, A.; Cindric, M.; Zarkovic, K.; Skrzydlewska, E.; Zarkovic, N. The relevance of pathophysio-
logical alterations in redox signaling of 4-hydroxynonenal for pharmacological therapies of major stress-associated diseases. Free
Radic. Biol. Med. 2020,157, 128–153. [CrossRef]
15.
Liu, A.; Zhang, X.; Li, R.; Zheng, M.; Yang, S.; Dai, L.; Wu, A.; Hu, C.; Huang, Y.; Xie, M.; et al. Overexpression of the SARS-CoV-2
receptor ACE2 is induced by cigarette smoke in bronchial and alveolar epithelia. J. Pathol. 2020,253, 17–30. [CrossRef]
16.
Gveric-Ahmetasevic, S.; Sunjic, S.B.; Skala, H.; Andrisic, L.; Stroser, M.; Zarkovic, K.; Skrablin, S.; Tatzber, F.; Cipak, A.; Jaganjac,
M.; et al. Oxidative stress in small-for-gestational age (SGA) term newborns and their mothers. Free Radic. Res.
2009
,43, 376–384.
[CrossRef]
Antioxidants 2021,10, 1341 10 of 10
17.
Nikolic-Heitzler, V.; Rabuzin, F.; Tatzber, F.; Vrkic, N.; Bulj, N.; Borovic, S.; Wonisch, W.; Sunko, B.M.; Zarkovic, N. Persistent
oxidative stress after myocardial infarction treated by percutaneous coronary intervention. Tohoku J. Exp. Med.
2006
,210, 247–255.
[CrossRef]
18.
Borovic, S.; Rabuzin, F.; Waeg, G.; Zarkovic, N. Enzyme-linked immunosorbent assay for 4-hydroxynonenal-histidine conjugates.
Free Radic. Res. 2006,40, 809–820. [CrossRef]
19.
Weber, D.; Milkovic, L.; Bennett, S.J.; Griffiths, H.R.; Zarkovic, N.; Grune, T. Measurement of HNE protein adducts in human
plasma and serum by ELISA—Comparison of two primary antibodies. Redox Biol. 2013,1, 226–233. [CrossRef] [PubMed]
20.
Jakovˇcevi´c, A.; Žarkovi´c, K.; Jakovˇcevi´c, D.; Rakuši´c, Z.; Prgomet, D.; Waeg, G.; Šunji´c, S.B.; Žarkovi´c, N. The Appearance of
4-Hydroxy-2-Nonenal (HNE) in Squamous Cell Carcinoma of the Oropharynx. Molecules 2020,25, 868. [CrossRef] [PubMed]
21.
LaForge, M.; Elbim, C.; Frère, C.; Hémadi, M.; Massaad, C.; Nuss, P.; Benoliel, J.-J.; Becker, C. Tissue damage from neutrophil-
induced oxidative stress in COVID-19. Nat. Rev. Immunol. 2020,20, 515–516. [CrossRef]
22.
Muhammad, Y.; Kani, Y.A.; Iliya, S.; Muhammad, J.B.; Binji, A.; Ahmad, A.E.-F.; Kabir, M.B.; Bindawa, K.U.; Ahmed, A. Deficiency
of antioxidants and increased oxidative stress in COVID-19 patients: A cross-sectional comparative study in Jigawa, Northwestern
Nigeria. SAGE Open Med. 2021,9, 1–8. [CrossRef] [PubMed]
23.
Gadotti, A.C.; Lipinski, A.L.; Vasconcellos, F.T.; Marqueze, L.F.; Cunha, E.B.; Campos, A.C.; Oliveira, C.F.; Amaral, A.N.; Baena,
C.P.; Telles, J.P.; et al. Susceptibility of the patients infected with Sars-Cov2 to oxidative stress and possible interplay with severity
of the disease. Free Radic. Biol. Med. 2021,165, 184–190. [CrossRef]
24.
Sovic, A.; Borovic, S.; Loncaric, I.; Kreuzer, T.; Zarkovic, K.; Vukovic, T.; Wäg, G.; Hrascan, R.; Wintersteiger, R.; Klinger, R.; et al.
The carcinostatic and proapoptotic potential of 4-Hydroxynonenal in HeLa cells is associated with its conjugation to cellular
proteins. Anticancer Res. 2001,21, 1997–2004. [PubMed]
25.
ˇ
Cipak, A.; Jaganjac, M.; Tehlivets, O.; Kohlwein, S.D.; Zarkovic, N. Adaptation to oxidative stress induced by polyunsaturated
fatty acids in yeast. Biochim. Biophys. Acta 2008,178, 283–287. [CrossRef]
26.
Luckey, S.W.; Taylor, M.; Sampey, B.; Scheinman, R.I.; Petersen, D.R.; Kang, J.L.; Lee, H.S.; Jung, H.J.; Kim, H.J.; Hah, J.S.; et al.
4-hydroxynonenal decreases interleukin-6 expression and protein production in primary rat Kupffer cells by inhibiting nuclear
factor-kappaB activation. J. Pharmacol. Exp. Ther. 2002,302, 296–303. [CrossRef]
27.
Wakita, C.; Honda, K.; Shibata, T.; Akagawa, M.; Uchida, K. A method for detection of 4-hydroxy-2-nonenal adducts in proteins.
Free Radic. Biol. Med. 2011,51, 1–4. [CrossRef] [PubMed]
28.
Rudic, M.; Nguyen, C.; Nguyen, Y.; Milkovi´c, L.; Zarkovic, N.; Sterkers, O.; Ferrary, E.; Grayeli, A.B. Effect of Angiotensin II on
Inflammation Pathways in Human Primary Bone Cell Cultures in Otosclerosis. Audiol. Neurootol.
2012
,17, 169–178. [CrossRef]
[PubMed]
29.
Rudi´c, M.; Milkovi´c, L.; Žarkovi´c, K.; Borovi´c-Šunji´c, S.; Sterkers, O.; Waeg, G.; Ferrary, E.; Grayeli, A.B.; Žarkovi´c, N. The effects
of angiotenzin II and oxidative stress mediator 4-hydroxynonenal on the human osteoblast-like cell growth: Possible relevance
for otosclerosis. Free Radic. Biol. Med. 2013,57, 22–28. [CrossRef]
30.
Jaganjac, M.; Cipak, A.; Schaur, R.J.; Zarkovic, N. Pathophysiology of Neutrophil-mediated Extracellular Redox Reactions. Front.
Biosci. Landmark 2015,29, 839–855. [CrossRef]
31.
Chacko, B.K.; Wall, S.B.; Kramer, P.A.; Ravi, S.; Mitchell, T.; Johnson, M.S.; Wilson, L.; Barnes, S.; Landar, A.; Darley-Usmar, V.M.
Pleiotropic effects of 4-hydroxynonenal on oxidative burst and phagocytosis in neutrophils. Redox Biol.
2016
,9, 57–66. [CrossRef]
32.
Riahi, Y.; Kaiser, N.; Cohen, G.; Abd-Elrahman, I.; Blum, G.; Shapira, O.M.; Koler, T.; Simionescu, M.; Sima, A.V.; Zarkovic, N.;
et al. Foam cell-derived 4-hydroxynonenal induces endothelial cell senescence in a TXNIP-dependent manner. J. Cell Mol. Med.
2015,18, 1887–1899. [CrossRef]
33.
Casós, K.; Zaragozá, M.C.; Zarkovic, N.; Zarkovic, K.; Andrisic, L.; Portero-Otín, M.; Cacabelos, D.; Mitjavila, M.T. A fish oil-rich
diet reduces vascular oxidative stress in apoE-/- mice. Free Radic. Res. 2010,44, 821–829. [CrossRef]
34.
Jaganjac, M.; Almuraikhy, S.; Al-Khelaifi, F.; Aljaber, M.; Bashah, M.; Mazloum, N.A.; Zarkovic, K.; Zarkovic, N.; Waeg, G.;
Kafienah, W.; et al. Combined metformin and insulin treatment reverses metabolically impaired omental adipogenesis and
accumulation of 4-hydroxynonenal in obese diabetic patients. Redox Biol. 2017,12, 483–490. [CrossRef]
35.
Silvagno, F.; Vernone, A.; Pescarmona, G.P. The Role of Glutathione in Protecting against the Severe Inflammatory Response
Triggered by COVID-19. Antioxidants 2020,9, 624. [CrossRef] [PubMed]
36.
Inchingolo, A.; Dipalma, G.; Inchingolo, A.; Malcangi, G.; Santacroce, L.; D’Oria, M.; Isacco, C.; Bordea, I.; Candrea, S.; Scarano,
A.; et al. The 15-Months Clinical Experience of SARS-CoV-2: A Literature Review of Therapies and Adjuvants. Antioxidants
2021
,
10, 881. [CrossRef]
37.
Wong, K.K.; Lee, S.W.H.; Kua, K.P. N-Acetylcysteine as Adjuvant Therapy for COVID-19—A Perspective on the Current State of
the Evidence. J. Inflamm. Res. 2021,14, 2993–3013. [CrossRef] [PubMed]
38.
Mohanty, R.R.; Padhy, P.M.; Das, S.; Meher, B.R. Therapeutic potential of N-acetyl cysteine (NAC) in preventing cytokine storm in
COVID-19: Review of current evidence. Eur. Rev. Med. Pharmacol. Sci. 2021,25, 2802–2807. [CrossRef] [PubMed]
... The outbreak of the COVID-19 pandemic has changed our lives and prompted many researchers to investigate the pathogenesis of this and related diseases (Table 1, Ref. [21][22][23][24][25][26][27][28][29][30]). The occurrence of oxidative stress and its consequences in viral infections such as SARS-CoV-2 infection have been extensively studied and reviewed [19]. ...
... Preliminary findings of the study comparing the levels of 4-HNE-protein adducts in the plasma of survivors and non-survivors during a five-day hospitalization in the intensive care unit, as determined by the 4-HNE-ELISA specific for the 4-HNE-histidine adducts, revealed higher levels of the 4-HNE-modified proteins in the plasma of deceased patients at admission and two days later, although no clinical differences were observed between these two groups at that time [22]. On the fifth day, which was critical for the patients' recovery or lethal outcome, levels of 4-HNE-protein adducts increased slightly in the plasma of survivors, whereas they decreased in the plasma of patients who later passed away. ...
... Myocardial and renal tissue Accumulation of 4-HNE IHC -- [21] Lungs, brain, heart, liver, kidneys --Accumulation of 4-HNE-protein adducts IHC [23] Lungs and plasma Accumulation of 4-HNE in the lungs IHC Dynamic changes in survivors: ab. 12 in the deceased patients. An autopsy of one patient revealed the abundant presence of 4-HNE-protein adducts in the lungs, according to immunohistochemical analysis performed with the same monoclonal antibody specific for the 4-HNE-histidine adducts that was used in the 4-HNE-ELISA [22]. ...
Article
Oxidative stress often affects the structure and metabolism of lipids, which in the case of polyunsaturated free fatty acids (PUFAs) leads to a self-catalysed chain reaction of lipid peroxidation (LPO). The LPO of PUFAs leads to the formation of various aldehydes, such as malondialdehyde, 4-hydroxynonenal (4-HNE), 4-hydroxyhexenal, and 4-oxo-2-nonenal. Among the reactive aldehydes, 4-HNE is the major bioactive product of LPO, which has a high affinity for binding to proteins. This review briefly discusses the available information on the applicability of assessment options for 4-HNE and its protein adducts determined by immunosorbent assay (the 4-HNE-ELISA) in patients with various diseases known to be associated with oxidative stress, LPO, and 4-HNE. Despite the differences in the protocols applied and the antibodies used, all studies confirmed the usefulness of the 4-HNE-ELISA for research purposes. Since different protocols and the antibodies used could give different values when applied to the same samples, the 4-HNE-ELISA should be combined with other complementary analytical methods to allow comparisons between the values obtained in patients and in healthy individuals. Despite large variations, the studies reviewed in this paper have mostly shown significantly increased levels of 4-HNE-protein adducts in the samples obtained from patients when compared to healthy individuals. As with any other biomarker studied in patients, it is preferred to perform not only a single-time analysis but measurements at multiple time points to monitor the dynamics of the occurrence of oxidative stress and the systemic response to the disease causing it. This is especially important for acute diseases, as individual levels of 4-HNE-protein adducts in blood can fluctuate more than threefold within a few days depending on the state of health, as was shown for the COVID-19 patients.
... Previously, it has been described that the development of COVID-19 is also a panied with a significant disturbance of redox homeostasis resulting in a high le oxidative stress that is related to the severity of the damage caused by the infection The decrease in the activity of antioxidant enzymes (superoxide dismutases 1 and 2 the decrease in the amount of low-molecular-weight antioxidants (glutathione, vit A and E) observed in the patient's plasma [4] also favors pro-oxidant metabolism patient's body. As a consequence of COVID-19, increased lipid metabolism has bee served, due to both oxidative stress and increased activity of phospholipase A2 (P lipoxygenases (LOXs), and cyclooxygenases (COXs) [5,6], manifested by an incr level of lipid metabolism products, including reactive aldehydes, such as 4-hydrox enal (4-HNE) and malondialdehyde (MDA), and cyclization products like neuro tanes/isoprostanes and prostaglandins [4,5,7]. ...
... To date, however, little is known about the modification o teins by lipid peroxidation products that have occurred in the bodies of COVID-1 tients. Of particular importance seem to be data correlating the degree of protein m cation by 4-HNE with the severity and impact of SARS-CoV-2 infection, which may lead to death [7]. MDA adducts with the innate immune system protein (protein D also equally important for neutralizing the SARS-CoV-2 virus [13]. ...
... The decrease in the activity of antioxidant enzymes (superoxide dismutases 1 and 2) and the decrease in the amount of low-molecular-weight antioxidants (glutathione, vitamins A and E) observed in the patient's plasma [4] also favors pro-oxidant metabolism in the patient's body. As a consequence of COVID-19, increased lipid metabolism has been observed, due to both oxidative stress and increased activity of phospholipase A2 (PLA2), lipoxygenases (LOXs), and cyclooxygenases (COXs) [5,6], manifested by an increased level of lipid metabolism products, including reactive aldehydes, such as 4-hydroxynonenal (4-HNE) and malondialdehyde (MDA), and cyclization products like neuroprostanes/isoprostanes and prostaglandins [4,5,7]. ...
Article
Full-text available
Although the COVID-19 pandemic has ended, it is important to understand the pathology of severe SARS-CoV-2 infection associated with respiratory failure and high mortality. The plasma proteome, including protein modification by lipid peroxidation products in COVID-19 survivors (COVID-19; n = 10) and deceased individuals (CovDeath; n = 10) was compared in samples collected upon admission to the hospital, when there was no difference in their status, with that of healthy individuals (Ctr; n = 10). The obtained results show that COVID-19 development strongly alters the expression of proteins involved in the regulation of exocytosis and platelet degranulation (top 20 altered proteins indicated by analysis of variance; p-value (False Discovery Rate) cutoff at 5%). These changes were most pronounced in the CovDeath group. In addition, the levels of 4-hydroxynonenal (4-HNE) adducts increased 2- and 3-fold, whereas malondialdehyde (MDA) adducts increased 7- and 2.5-fold, respectively, in COVID-19 and CovDeath groups. Kinases and proinflammatory proteins were particularly affected by these modifications. Protein adducts with 15-deoxy-12,14-prostaglandin J2 (15d-PGJ2) were increased 2.5-fold in COVID-19 patients, including modifications of proteins such as p53 and STAT3, whereas CovDeath showed a decrease of approximately 60% compared with Ctr. This study for the first time demonstrates the formation of lipid metabolism products—protein adducts in plasma from survived and deceased COVID-19 patients, significantly distinguishing them, which may be a predictor of the course of SARS-CoV-2 infection.
... Elevated levels of lipid peroxidation products in patients infected with the SARS-CoV-2 were also reported in other studies. 6,63,64 Some researchers found correlations between the concentrations of lipid peroxidation products and disease severity, 65 and between 4-HNE levels and the risk of mortality in COVID-19 patients. 6 The peroxidation of lipids, mainly unsaturated fatty acids, leads to the production of peroxides and, in subsequent reactions, aldehydes such as MDA and HNE 66,67 which were analyzed in this study. ...
Article
Full-text available
Aim COVID-19 triggers the overproduction of reactive oxygen species (ROS) which, in combination with a weakened antioxidant barrier, can lead to protein oxidation and lipid peroxidation. The aim of this study was to evaluate enzymatic and non-enzymatic antioxidants, the overall redox potential, and protein and lipid peroxidation products in COVID-19 patients, convalescents, and healthy subjects, and to the determine the diagnostic applicability of these parameters in COVID-19 patients. Materials and Methods The study involved 218 patients with COVID-19, 69 convalescents, and 48 healthy subjects who were selected for the research based on age and sex. The study was conducted between 20 February 2021 and 20 November 2021 in Białystok, Poland. The antioxidant barrier, redox status, and oxidative damage products were assessed in serum/plasma samples with the use of colorimetric and spectrophotometric assays. Results Glutathione reductase (GR) activity was higher, whereas total antioxidant capacity (TAC) was lower in COVID-19 patients than in convalescents (p<0.0001) and the control group (p<0.0001). The concentrations of advanced glycation end products (AGEs), advanced oxidation protein products (AOPP), 4-hydroxynonenal (4-HNE), and malondialdehyde (MDA) were higher in COVID-19 patients (p<0.0001) and convalescents (p<0.0001) than in the control group. AGEs were the most effective diagnostic biomarker for differentiating COVID-19 patients from the control group (AUC=0.9971) and convalescents from the control group (AUC=1.000). Conclusion An infection with the SARS-CoV-2 disrupts the redox balance and increases protein oxidation and lipid peroxidation. AGEs fulfill the criteria for a potential diagnostic biomarker in COVID-19 patients and convalescents.
... Another important modification of proteins that may alter mFLT is related to the creation of malondialdehyde (MDA)-protein adducts known as advanced lipoxidation end-products (ALEs). It has been revealed that higher lipid peroxidation and aldehydes levels are associated with increased severity of disease in COVID-19 patients [43,44]. MDA is a highly reactive compound that can readily react with amino groups of proteins such as HSA, leading to the formation of MDA-HSA adducts. ...
Article
Full-text available
Fluorescence lifetime measurements of blood or plasma offer valuable insights into the microenvironment and molecular interactions of fluorophores, particularly concerning albumin. Neutrophil- and hypoxia-induced oxidative stress in COVID-19 pneumonia patients leads to hyperinflammation, various oxidative modifications of blood proteins, and potential alterations in the fluorescence lifetime of tryptophan-containing proteins, especially albumin. The objective of this study was to investigate the efficacy of time-resolved fluorescence spectroscopy of blood and plasma as a prompt diagnostic tool for the early diagnosis and severity assessment of COVID-19-associated pneumonia. This study examined a cohort of sixty COVID-19 patients with respiratory symptoms. To investigate whether oxidative stress is the underlying cause of the change in fluorescence lifetime, human serum albumin was treated with chloramine T. The time-resolved spectrometer Life Spec II (Edinburgh Instruments Ltd., Livingston, UK), equipped with a sub-nanosecond pulsed 280 nm diode, was used to measure the fluorescence lifetime of blood and plasma. The findings revealed a significant reduction in the fluorescence lifetime of blood (diluted 200 times) and plasma (diluted 20 times) at 360 nm in COVID-19 pneumonia patients compared with their respective values recorded six months post-infection and those of healthy individuals. Significant negative correlations were observed between the mean fluorescence lifetime of blood and plasma at 360 nm and several severity biomarkers and advanced oxidation protein products, while a positive correlation was found with albumin and the albumin–globulin ratio. The time-resolved fluorescence spectroscopy method demonstrates the potential to be used as a preliminary screening technique for identifying patients who are at risk of developing severe complications. Furthermore, the small amount of blood required for the measurements has the potential to enable a rapid fingerstick blood test.
... One of the NFκB-IκB complex inhibitors, i.e., 4-HNE, is the main bioactive product of arachidonic acid peroxidation, the level of which increases oxidative stress under the influence of COVID-19 infection [41,42], which may be associated with systemic vascular stress in patients, especially those who died from COVID-19 [27,43]. It is known, however, that NFκB is necessary to maintain immune homeostasis and prevent autoimmunity caused by SARS-CoV-2 [39]. ...
Article
Full-text available
The aim of this study was to evaluate selected parameters of redox signaling and inflammation in the granulocytes of COVID-19 patients who recovered and those who died. Upon admission, the patients did not differ in terms of any relevant clinical parameter apart from the percentage of granulocytes, which was 6% higher on average in those patients who died. Granulocytes were isolated from the blood of 15 healthy people and survivors and 15 patients who died within a week, and who were selected post hoc for analysis according to their matching gender and age. They differed only in the lethal outcome, which could not be predicted upon arrival at the hospital. The proteins level (respective ELISA), antioxidant activity (spectrophotometry), and lipid mediators (UPUPLC–MS) were measured in the peripheral blood granulocytes obtained via gradient centrifugation. The levels of Nrf2, HO-1, NFκB, and IL-6 were higher in the granulocytes of COVID-19 patients who died within a week, while the activity of cytoplasmic Cu,Zn-SOD and mitochondrial Mn-SOD and IL-2/IL-10 were lower in comparison to the levels observed in survivors. Furthermore, in the granulocytes of those patients who died, an increase in pro-inflammatory eicosanoids (PGE2 and TXB2), together with elevated cannabinoid receptors 1 and 2 (associated with a decrease in the anti-inflammatory 15d-PGJ2), were found. Hence, this study suggests that by triggering transcription factors, granulocytes activate inflammatory and redox signaling, leading to the production of pro-inflammatory eicosanoids while reducing cellular antioxidant capacity through SOD, thus expressing an altered response to COVID-19, which may result in the onset of systemic oxidative stress, ARDS, and the death of the patient.
... This is supported by the current knowledge in the scientific literature related to similar respiratory infectious diseases and, sometimes, SARS-CoV-2 infection itself. Thus, the following sections address the existing link between immunity, oxidative stress, and COVID-19 [9], together with a compilation of the scientific insights into the relationships between several minerals and this disease. The review focuses on the trace elements zinc, selenium, copper, and iron, and other minerals such as magnesium, as they exert the most relevant oxidative and immune-related effects, having been the most studied by the scientific community within the context of COVID-19 and other respiratory infections. ...
Article
Full-text available
Since the coronavirus disease 2019 (COVID-19) pandemic appeared, both governments and the scientific community have focused their efforts on the search for prophylactic and therapeutic alternatives in order to reduce its effects. Vaccines against SARS-CoV-2 have been approved and administered, playing a key role in the overcoming of this situation. However, they have not reached the whole world population, and several doses will be needed in the future in order to successfully protect individuals. The disease is still here, so other strategies should be explored with the aim of supporting the immune system before and during the infection. An adequate diet is certainly associated with an optimal inflammatory and oxidative stress status, as poor levels of different nutrients could be related to altered immune responses and, consequently, an augmented susceptibility to infections and severe outcomes derived from them. Minerals exert a wide range of immune-modulatory, anti-inflammatory, antimicrobial, and antioxidant activities, which may be useful for fighting this illness. Although they cannot be considered as a definitive therapeutic solution, the available evidence to date, obtained from studies on similar respiratory diseases, might reflect the rationality of deeper investigations of the use of minerals during this pandemic.
... In addition to the reduced endogenous CoQ 10 biosynthesis, decreased antioxidant capacity in patients with COVID-19, as alterations in the activity of glutathione peroxidase, total antioxidant capacity was observed [65,66]. In another study, the opposite results were found. ...
Article
Full-text available
Background: Mitochondrial dysfunction and redox cellular imbalance indicate crucial function in COVID-19 pathogenesis. Since 11 March 2020, a global pandemic, health crisis and economic disruption has been caused by SARS-CoV-2 virus. Vaccination is considered one of the most effective strategies for preventing viral infection. We tested the hypothesis that preventive vaccination affects the reduced bioenergetics of platelet mitochondria and the biosynthesis of endogenous coenzyme Q10 (CoQ10) in patients with post-acute COVID-19. Material and methods: 10 vaccinated patients with post-acute COVID-19 (V + PAC19) and 10 unvaccinated patients with post-acute COVID-19 (PAC19) were included in the study. The control group (C) consisted of 16 healthy volunteers. Platelet mitochondrial bioenergy function was determined with HRR method. CoQ10, γ-tocopherol, α-tocopherol and β-carotene were determined by HPLC, TBARS (thiobarbituric acid reactive substances) were determined spectrophotometrically. Results: Vaccination protected platelet mitochondrial bioenergy function but not endogenous CoQ10 levels, in patients with post-acute COVID-19. Conclusions: Vaccination against SARS-CoV-2 virus infection prevented the reduction of platelet mitochondrial respiration and energy production. The mechanism of suppression of CoQ10 levels by SARS-CoV-2 virus is not fully known. Methods for the determination of CoQ10 and HRR can be used for monitoring of mitochondrial bioenergetics and targeted therapy of patients with post-acute COVID-19.
Article
Full-text available
The COVID‐19 pandemic has had a devastating impact on global health, highlighting the need to understand how the SARS‐CoV‐2 virus damages the lungs in order to develop effective treatments. Recent research has shown that patients with COVID‐19 experience severe oxidative damage to various biomolecules. We propose that the overproduction of reactive oxygen species (ROS) in SARS‐CoV‐2 infection involves an interaction between copper ions and the virus‘s spike protein. We tested two peptide fragments, Ac‐ELDKYFKNH‐NH2 (L1) and Ac‐WSHPQFEK‐NH2 (L2), derived from the spike protein of the Wuhan strain and the β variant, respectively, and found that they bind Cu(II) ions and form a three‐nitrogen complexes at lung pH. Our research demonstrates that these complexes trigger the overproduction of ROS, which can break both DNA strands and transform DNA into its linear form. Using A549 cells, we demonstrated that ROS overproduction occurs in the mitochondria, not in the cytoplasm. Our findings highlight the importance of the interaction between copper ions and the virus‘s spike protein in the development of lung damage and may aid in the development of therapeutic procedures.
Article
Full-text available
Introduction The COVID-19 is a pandemic caused by SARS-CoV-2 which has infected over 74 million people, killing more than 1,600,000 million people around the world as of 17th December 2020. Accumulation of free radicals coupled by weakened antioxidant system leads to oxidative stress, which will further worsen respiratory diseases, COVID-19 inclusive. This study aimed to examine the levels of some antioxidants and oxidative stress markers in COVID-19 patients. Methods This was a cross-sectional comparative study in which 50 COVID-19 symptomatic patients who were on admission at the COVID-19 isolation center in Jigawa, Northwestern Nigeria, were recruited. Twenty one (21) apparently healthy individuals were included as controls. Levels of antioxidant trace elements (Se, Zn, Mg, Cu and Cr), 8-isoprostaglandin F2 alpha and malondialdehyde in the plasma and erythrocytes activity of glutathione, glutathione peroxidase, superoxide dismutase and catalase were determined. Results The plasma concentrations of vitamins A, C and E were significantly lower (p < 0.001) in COVID-19 patients than controls. Activities of glutathione, glutathione peroxidase, catalase and superoxide dismutase were lower in COVID-19 subjects than controls (p < 0.001). The concentrations of Se, Zn, Mg and Cu were significantly lower (p < 0.001; p = 0.039; p < 0.001; and p < 0.001), respectively, in COVID-19 patients than controls, while chromium showed no significant difference (p = 0.605). Oxidative stress marker, 8-isoprostaglandin F2 alpha, was significantly higher (p = 0.049), while malondialdehyde was lower (p < 0.001) in COVID-19 patients than controls. Conclusion In conclusion, COVID-19 patients are prone to depleted levels of antioxidant substances due to their increase utilization in counterbalancing the negative effect of free radicals. Furthermore, COVID-19 infection with other comorbidities, such as malaria, hypertension and diabetes, are at higher risk of developing oxidative stress.
Article
Full-text available
The looming severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused a long-lasting pandemic of coronavirus disease 2019 (COVID-19) around the globe with substantial morbidity and mortality. N-acetylcysteine, being a nutraceutical precursor of an important antioxidant glutathione, can perform several biological functions in mammals and microbes. It has consequently garnered a growing interest as a potential adjunctive therapy for coronavirus disease. Here, we review evidence concerning the effects of N-acetylcysteine in respiratory viral infections based on currently available in vitro, in vivo, and human clinical investigations. The repurposing of a known drug such as N-acetylcysteine may significantly hasten the deployment of a novel approach for COVID-19. Since the drug candidate has already been translated into the clinic for several decades, its established pharmacological properties and safety and side-effect profiles expedite preclinical and clinical assessment for the treatment of COVID-19. In vitro data have depicted that N-acetylcysteine increases antioxidant capacity, interferes with virus replication, and suppresses expression of pro-inflammatory cytokines in cells infected with influenza viruses or respiratory syncytial virus. Furthermore, findings from in vivo studies have displayed that, by virtue of immune modulation and anti-inflammatory mechanism, N-acetylcysteine reduces the mortality rate in influenza-infected mice animal models. The promising in vitro and in vivo results have prompted the initiation of human subject research for the treatment of COVID-19, including severe pneumonia and acute respiratory distress syndrome. Albeit some evidence of benefits has been observed in clinical outcomes of patients, precision nanoparticle design of N-acetylcysteine may allow for greater therapeutic efficacy.
Article
Full-text available
Background: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the virus responsible for the coronavirus disease of 2019 (COVID-19) that emerged in December 2019 in Wuhan, China, and rapidly spread worldwide, with a daily increase in confirmed cases and infection-related deaths. The World Health Organization declared a pandemic on the 11th of March 2020. COVID-19 presents flu-like symptoms that become severe in high-risk medically compromised subjects. The aim of this study was to perform an updated overview of the treatments and adjuvant protocols for COVID-19. Methods: A systematic literature search of databases was performed (MEDLINE PubMed, Google Scholar, UpToDate, Embase, and Web of Science) using the keywords: "COVID-19", "2019-nCoV", "coronavirus" and "SARS-CoV-2" (date range: 1 January 2019 to 31st October 2020), focused on clinical features and treatments. Results: The main treatments retrieved were antivirals, antimalarials, convalescent plasma, immunomodulators, corticosteroids, anticoagulants, and mesenchymal stem cells. Most of the described treatments may provide benefits to COVID-19 subjects, but no one protocol has definitively proven its efficacy. Conclusions: While many efforts are being spent worldwide in research aimed at identifying early diagnostic methods and evidence-based effective treatments, mass vaccination is thought to be the best option against this disease in the near future.
Article
Full-text available
Since November 2019, SARS Coronavirus 2 disease (COVID-19) pandemic has spread through more than 195 nations worldwide. Though the coronavirus infection affects all age and sex groups, the mortality is skewed towards the elderly population and the cause of death is mostly acute respiratory distress syndrome (ARDS). There are data suggesting the role of excessive immune activation and cytokine storm as the cause of lung injury in COVID-19. The excessive immune activation and cytokine storm usually occurs due to an imbalance in redox homeostasis of the individuals. Considering the antioxidant and free radical scavenging action of N acetyl cysteine (NAC), its use might be useful in COVID-19 patients by decreasing the cytokine storm consequently decreasing the disease severity. Therefore, we reviewed all the available resources pertaining to the role of reactive oxygen species (ROS) in cytokine storm and the mechanism of action of NAC in preventing ROS. We also reviewed the use of NAC in COVID-19.
Article
Full-text available
Background: A key role of oxidative stress has been highlighted in the pathogenesis of COVID-19. However, little has been said about oxidative stress status (OSS) of COVID-19 patients hospitalized in intensive care unit (ICU). Material and methods: Biomarkers of the systemic OSS included antioxidants (9 assays), trace elements (3 assays), inflammation markers (4 assays) and oxidative damage to lipids (3 assays). Results: Blood samples were drawn after 9 (7-11) and 41 (39-43) days of ICU stay, respectively in 3 and 6 patients. Vitamin C, thiol proteins, reduced glutathione, γ-tocopherol, β-carotene and PAOT® score were significantly decreased compared to laboratory reference values. Selenium concentration was at the limit of the lower reference value. By contrast, the copper/zinc ratio (as a source of oxidative stress) was higher than reference values in 55% of patients while copper was significantly correlated with lipid peroxides (r = 0.95, p < 0.001). Inflammatory biomarkers (C-reactive protein and myeloperoxidase) were significantly increased when compared to normals. Conclusions: The systemic OSS was strongly altered in critically ill COVID-19 patients as evidenced by increased lipid peroxidation but also by deficits in some antioxidants (vitamin C, glutathione, thiol proteins) and trace elements (selenium).
Article
Full-text available
Since the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) outbreak emerged, countless efforts are being made worldwide to understand the molecular mechanisms underlying the coronavirus disease 2019 (COVID-19) in an attempt to identify the specific clinical characteristics of critically ill COVID-19 patients involved in its pathogenesis and provide therapeutic alternatives to minimize COVID-19 severity. Recently, COVID-19 has been closely related to sepsis, which suggests that most deceases in intensive care units (ICU) may be a direct consequence of SARS-CoV-2 infection-induced sepsis. Understanding oxidative stress and the molecular inflammation mechanisms contributing to COVID-19 progression to severe phenotypes such as sepsis is a current clinical need in the effort to improve therapies in SARS-CoV-2 infected patients. This article aims to review the molecular pathogenesis of SARS-CoV-2 and its relationship with oxidative stress and inflammation, which can contribute to sepsis progression. We also provide an overview of potential antioxidant therapies and active clinical trials that might prevent disease progression or reduce its severity.
Article
Full-text available
Aims: Cardiovascular complications, including myocarditis, are observed in coronavirus disease 2019 (COVID-19). Major cardiac involvement is a potentially lethal feature in severe cases. We sought to describe the underlying pathophysiological mechanism in COVID-19 lethal cardiogenic shock. Methods and results: We report on a 48-year-old male COVID-19 patient with cardiogenic shock; despite extracorporeal life support, dialysis, and massive pharmacological support, this rescue therapy was not successful. Severe acute respiratory syndrome coronavirus 2 RNA was detected at autopsy in the lungs and myocardium. Histopathological examination revealed diffuse alveolar damage, proliferation of type II pneumocytes, lymphocytes in the lung interstitium, and pulmonary microemboli. Moreover, patchy muscular, sometimes perivascular, interstitial mononuclear inflammatory infiltrates, dominated by lymphocytes, were seen in the cardiac tissue. The lymphocytes 'interlocked' the myocytes, resulting in myocyte degeneration and necrosis. Predominantly, T-cell lymphocytes with a CD4:CD8 ratio of 1.7 infiltrated the interstitial myocardium, reflecting true myocarditis. The myocardial tissue was examined for markers of ferroptosis, an iron-catalysed form of regulated cell death that occurs through excessive peroxidation of polyunsaturated fatty acids. Immunohistochemical staining with E06, a monoclonal antibody binding to oxidized phosphatidylcholine (reflecting lipid peroxidation during ferroptosis), was positive in morphologically degenerating and necrotic cardiomyocytes adjacent to the infiltrate of lymphocytes, near arteries, in the epicardium and myocardium. A similar ferroptosis signature was present in the myocardium of a COVID-19 subject without myocarditis. In a case of sudden death due to viral myocarditis of unknown aetiology, however, immunohistochemical staining with E06 was negative. The renal proximal tubuli stained positively for E06 and also hydroxynonenal (4-HNE), a reactive breakdown product of the lipid peroxides that execute ferroptosis. In the case of myocarditis of other aetiology, the renal tissue displayed no positivity for E06 or 4-HNE. Conclusions: The findings in this case are unique as this is the first report on accumulated oxidized phospholipids (or their breakdown products) in myocardial and renal tissue in COVID-19. This highlights ferroptosis, proposed to detrimentally contribute to some forms of ischaemia-reperfusion injury, as a detrimental factor in COVID-19 cardiac damage and multiple organ failure.
Article
Several recent reviews have suggested a role of oxidative stress in the pathophysiology of COVID-19, but its interplay with disease severity has not been revealed yet. In the present study, we aimed to investigate the association between the severity of COVID‐19 and oxidative stress parameters. Clinical data of 77 patients with COVID‐19 admitted to the hospital were analyzed and divided into moderate (n = 44) and severe (n = 33) groups based on their clinical condition. Production of oxidant (hydrogen peroxide) and defense antioxidants (total antioxidant capacity, reduced and oxidized glutathione, glutathione s-transferase), and oxidative damage (malondialdehyde, carbonyl, and sulfhydryl) were assessed using the serum samples. The results revealed that severe patients who presented high serum leukocyte count and CRP level stayed for a longer period in the hospital. However, there was no correlation observed between the oxidative stress parameters and degree of COVID-19 severity in the present study. In conclusion, these results indicate that the disease severity may not be a detrimental factor contributing to the changes in the redox profile of hospitalized patients with COVID-19.
Article
Angiotensin‐converting enzyme 2 (ACE2) has been identified as the functional receptor of severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2) and a target for disease prevention. However, the relationship between ACE2 expression and its clinical implications in SARS‐CoV‐2 pathogenesis remains unknown. Here, we explored the location and expression of ACE2, and its correlation with gender, age, and cigarette smoke (CS), in a CS‐exposed mouse model and 224 non‐malignant lung tissues (125 non‐smokers, 81 current smokers, and 18 ex‐smokers) by immunohistochemistry. Moreover, the correlations of ACE2 with CS‐induced oxidative stress‐related markers, hypoxia‐inducible factor‐1α (HIF‐1α), inducible nitric oxide synthase (iNOS), and 4‐hydroxynonenal (4‐HNE) were investigated. Chromatin immunoprecipitation and luciferase reporter assays identified the cause of ACE2 overexpression in human primary lung epithelial cells. We demonstrated that ACE2 was predominantly overexpressed on the apical surface of bronchial epithelium, while reduced in alveolar epithelium, owing to the dramatically decreased abundance of alveolar type II pneumocytes in CS‐exposed mouse lungs. Consistent with this, ACE2 was primarily significantly overexpressed in human bronchial and alveolar epithelial cells in smokers regardless of age or gender. Decreased ACE2 expression was observed in bronchial epithelial cells from ex‐smokers compared with current smokers, especially in those who had ceased smoking for more than 10 years. Moreover, ACE2 expression was positively correlated with the levels of HIF‐1α, iNOS, and 4‐HNE in both mouse and human bronchioles. The results were further validated using a publicly available dataset from The Cancer Genome Atlas (TCGA) and our previous integrated data from Affymetrix U133 Plus 2.0 microarray (AE‐meta). Finally, our results showed that HIF‐1α transcriptionally upregulates ACE2 expression. Our results indicate that smoking‐induced ACE2 overexpression in the apical surface of bronchial epithelial cells provides a route by which SARS‐CoV‐2 enters host cells, which supports clinical relevance in attenuating the potential transmission risk of COVID‐19 in smoking populations by smoking cessation. © 2020 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Article
Studies from China on Covid‐19 revealed that non‐survivors had cytokine storm with high IL‐6 and hyperferritinemia. Iron liberated from necrotic cells may catalyze free radical production and amplify lipid peroxidation causing membrane dysfunction and multiorgan failure. Consequently, iron chelators have been successfully utilized in various experimental and clinical models of cytokine storm and multiorgan damage, such as in ischemia–reperfusion injury, sepsis and infections. Since viral replication may be influenced by iron accumulation, iron chelation has been proven beneficial in a variety of viral infections, such as HIV‐1, hepatitis B virus, Mengo virus, Marburg hemorrhagic fever, Enterovirus 71 and West Nile virus. In this commentary we elaborate on the idea of considering iron chelation, as therapeutic modality in patients with severe COVID‐19 infection. For critically ill patients in ICU, intravenous deferoxamine would provide sufficient and rapid iron chelation to ameliorate cytokine storm, whereas in less severe cases an oral chelator could prevent the development of excessive inflammatory response.