ArticlePDF Available

Hyperglycemia and Loss of Redox Homeostasis in COVID-19 Patients

Authors:

Abstract

The infection with SARS-CoV-2 impairs the glucose–insulin axis and this contributes to oxidative (OS) and nitrosative (NSS) stress. Here, we evaluated changes in glucose metabolism that could promote the loss of redox homeostasis in COVID-19 patients. This was comparative cohort and analytical study that compared COVID-19 patients and healthy subjects. The study population consisted of 61 COVID-19 patients with and without comorbidities and 25 healthy subjects (HS). In all subjects the plasma glucose, insulin, 8-isoprostane, Vitamin D, H2S and 3-nitrotyrosine were determined by ELISA. The nitrites (NO2 􀀀), lipid-peroxidation (LPO), total-antioxidant-capacity (TAC), thiols, glutathione (GSH) and selenium (Se) were determined by spectrophotometry. The glucose, insulin and HOMA-IR (p < 0.001), 8-isoprostanes, 3-nitrotyrosine (p < 0.001) and LPO were increased (p = 0.02) while Vitamin D (p = 0.01), H2S, thiols, TAC, GSH and Se (p < 0.001) decreased in COVID-19 patients in comparison to HS. The SARS-CoV-2 infection resulted in alterations in the glucose–insulin axis that led to hyperglycemia, hyperinsulinemia and IR in patients with and without comorbidities. These alterations increase OS and NSS reflected in increases or decreases in some oxidative markers in plasma with major impact or fatal consequences in patients that course with metabolic syndrome. Moreover, subjects without comorbidities could have long-term alterations in the redox homeostasis after infection.


Citation: Soto, M.E.;
Guarner-Lans, V.; Díaz-Díaz, E.;
Manzano-Pech, L.;
Palacios-Chavarría, A.;
Valdez-Vázquez, R.R.;
Aisa-Álvarez, A.;
Saucedo-Orozco, H.; Pérez-Torres, I.
Hyperglycemia and Loss of Redox
Homeostasis in COVID-19 Patients.
Cells 2022,11, 932. https://doi.org/
10.3390/cells11060932
Academic Editor: Zhixiang Wang
Received: 10 February 2022
Accepted: 7 March 2022
Published: 9 March 2022
Publisher’s Note: MDPI stays neutral
with regard to jurisdictional claims in
published maps and institutional affil-
iations.
Copyright: © 2022 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/).
cells
Article
Hyperglycemia and Loss of Redox Homeostasis in
COVID-19 Patients
María Elena Soto 1, , Verónica Guarner-Lans 2,† , Eulises Díaz-Díaz 3, Linaloe Manzano-Pech 4,
Adrían Palacios-Chavarría5, Rafael Ricardo Valdez-Vázquez 5, Alfredo Aisa-Álvarez 1,
Huitzilihuitl Saucedo-Orozco 1and Israel Pérez-Torres 4,*
1
Department of Immunology, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano 1, Sección XVI,
Tlalpan, Mexico City 14080, Mexico; elena.soto@cardiologia.org.mx (M.E.S.);
alfredoaisaa@gmail.com (A.A.-Á.); huitzilihuitls@hotmail.com (H.S.-O.)
2Department of Physiology, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano 1, Sección XVI,
Tlalpan, Mexico City 14080, Mexico; veronica.guarner@cardiologia.org.mx
3Department of Reproductive Biology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán,
Vasco de Quiroga 15, Sección XVI, Tlalpan, Mexico City 14000, Mexico; eulisesd@yahoo.com
4Department of Cardiovascular Biomedicine, Instituto Nacional de Cardiología Ignacio Chávez,
Mexico City 14080, Mexico; loe_mana@hotmail.com
5Critical Care Unit of the Temporal COVID-19 Unit, Citibanamex Center, Mexico City 11200, Mexico;
a2novi@hotmail.com (A.P.-C.); rrvaldezvazquez@gmail.com (R.R.V.-V.)
*Correspondence: israel.perez@cardiologia.org.mx; Tel.: +52-5573-2911 (ext. 25203); Fax: +52-5573-0926
These authors contributed equally to this work.
Abstract:
The infection with SARS-CoV-2 impairs the glucose–insulin axis and this contributes to
oxidative (OS) and nitrosative (NSS) stress. Here, we evaluated changes in glucose metabolism that
could promote the loss of redox homeostasis in COVID-19 patients. This was comparative cohort
and analytical study that compared COVID-19 patients and healthy subjects. The study population
consisted of 61 COVID-19 patients with and without comorbidities and 25 healthy subjects (HS).
In all subjects the plasma glucose, insulin, 8-isoprostane, Vitamin D, H
2
S and 3-nitrotyrosine were
determined by ELISA. The nitrites (NO
2
), lipid-peroxidation (LPO), total-antioxidant-capacity
(TAC), thiols, glutathione (GSH) and selenium (Se) were determined by spectrophotometry. The
glucose, insulin and HOMA-IR (p< 0.001), 8-isoprostanes, 3-nitrotyrosine (p< 0.001) and LPO were
increased (p= 0.02) while Vitamin D (p= 0.01), H
2
S, thiols, TAC, GSH and Se (p< 0.001) decreased
in COVID-19 patients in comparison to HS. The SARS-CoV-2 infection resulted in alterations in the
glucose–insulin axis that led to hyperglycemia, hyperinsulinemia and IR in patients with and without
comorbidities. These alterations increase OS and NSS reflected in increases or decreases in some
oxidative markers in plasma with major impact or fatal consequences in patients that course with
metabolic syndrome. Moreover, subjects without comorbidities could have long-term alterations in
the redox homeostasis after infection.
Keywords:
SARS-CoV-2; homeostasis redox; hyperglycemia; lipid peroxidation; selenium; thiols;
nitrotyrosine; H2S
1. Introduction
Coronavirus disease 2019 (COVID-19) is caused by the type 2
β
-coronavirus and it
induces a severe acute respiratory syndrome (SARS-CoV-2). This disease causes multiple
organ failures that results from an exacerbated cytokine storm by the immune system.
The cytokine storm is initially aimed at bringing down the infection and may have as a
consequence, a possible fatal outcome for the patient [1].
COVID-19 impairs glucose homeostasis and metabolism in non- and diabetes mellitus
(DM) and metabolic syndrome (MS) patients due to the cytokine storm, inflammation,
angiotensin II-converting enzyme (ACE2) down-regulation and the direct injury to the
Cells 2022,11, 932. https://doi.org/10.3390/cells11060932 https://www.mdpi.com/journal/cells
Cells 2022,11, 932 2 of 23
pancreatic
β
-cells [
2
]. It also deteriorates the already impaired glucose homeostasis in
patients with DM and in patients with other comorbidities such as obesity, insulin resistance
(IR), hyperinsulinemia and hypertension in whom alterations in insulin secretion are
already present and contribute to hyperglycemia [
3
]. Furthermore, increased inflammation
and massive production of cytokines generate IR which adds to the deterioration of the
insulin secretion by β-cells [4].
In addition, high glucose levels also contribute to the virulence and replication of
SARS-CoV-2 since it is associated with a decrease in the number of phagocytic and nuclear
polymorphic leukocytes [
4
]. Moreover, the virus sequesters the host’s mitochondrial
function and shifts it from aerobic to anaerobic [
5
]. In this state, the pyruvate produced
from glucose during glycolysis is oxidized to lactate, thereby increasing glucose levels in
the cytosol and leading to the generation of limited amounts of adenosine-5-triphosphate
(ATP) [
5
,
6
]. The viral replication also consumes large amounts of ATP and therefore, its
concentration is depleted. In this condition, lactate is not metabolized by gluconeogenesis
and accumulates in the blood, leading to a loss of the balance in glucose metabolism [
7
].
The increase in hyperglycemia contributes a high production of inflammatory cytokines
and cellular mediators implied in pro-thrombotic process present in COVID-19 patients. [
8
].
All these alterations favor oxidative stress (OS) but also, the viral infection induces the
cytokine storm by the immune system that contribute to OS [
9
]. This alteration leads to
positive feedback systems for each element and/or metabolic pathway and may contribute
to severe pneumonia with possible multiple organ failure (MOF) in the COVID-19 patient.
On the other hand, an overproduction of reactive oxygen species (ROS) leads to a
deprivation of the antioxidant mechanisms which are crucial to decrease or abolish viral
replication and the level of infection present in the patient [
10
]. Therefore, SARS-CoV-2
favors an increased threshold in the production of ROS and reactive nitrogen species (RNS).
The overproduction of these molecules is associated with elevated activity and/or ex-
pression of the inducible nitric oxide synthase (iNOS), nicotinamide-adenine dinucleotide
phosphate (NADP) oxidases, cyclooxygenase 2, xanthine oxidase and with alterations in
the mitochondrial functions that activate transcription factors such as nuclear factor kappa
B subunit (NFkB) resulting in an exacerbated proinflammatory state and the production
of interleukins [
5
,
11
]. Therefore, Interleukin (IL)-6, IL-8 and tumor necrosis factor alpha
(TNF-
α
) are increased in bronchial epithelial cells and alveolar macrophages and these
cytokines can then activate macrophages and neutrophils, resulting in the destruction of the
alveolar wall, the collapse of small airways, hyper-permeability of pulmonary capillaries
and pulmonary edema, that result in the deterioration of pulmonary gas exchange [
10
].
These changes contribute to acute respiratory distress syndrome (ARDS), chronic obstruc-
tive pulmonary disease (COPD) and acute lung injury (ALI) [
1
]. However, the complex
combination of the alterations in glucose metabolism and the association with the loss of
redox homeostasis in COVID-19 patients have not been completely analyzed. Therefore, the
aim of this study was to evaluate changes in glucose metabolism that could contribute to the
loss of redox homeostasis in the plasma of patients with moderate and severe pneumonia
by SARS-CoV-2 with and without comorbidities. In this study, we also tested whether the
use of the presence of the anti-N protein and anti-S protein antibodies of the SARS-CoV-2
virus at the time of admission of the patients can be used as a diagnostic tool for COVID-19.
2. Materials and Methods
2.1. Study Type
This was a comparative cohort and analytical study that compared COVID-19 patients
and healthy subjects (HS). The study was run between 20 August and 20 September 2020.
The study population consisted of 61 patients over 18 years of age who were admitted to the
intensive care unit (ICU) of the CITIBANAMEX Center and that had not developed septic
shock, secondary to moderate or severe pneumonia due to COVID-19. Diagnostic criteria
for septic shock were based on the Sepsis-3 consensus [
12
]. Exclusion from this study
occurred when patients were younger than 18 years of age, when they were not able to
Cells 2022,11, 932 3 of 23
grant an informed consent or when they refused to be included. Patients were also excluded
if pregnant or breastfeeding or if they were under chronic use (last 6 months) or recent use
of steroids, statins or antioxidants. The hospitalized patients included were considered
to have moderate or severe symptoms considering their ventilatory status. Moderate
COVID-19 patients were classified according to the Horowitz index which is defined as the
ratio of partial pressure of oxygen (PaO
2
in mmHg) in blood to the fraction of oxygen in
inhaled air (FiO
2
), (PaO
2
/FiO
2
) with value index percentage of fraction
>200 mmHg [13]
.
Patients with the severe condition required invasive mechanical intubation according to
the criteria of Berlin for ARDS. The Berlin definition proposes 3 categories of ARDS based
on the severity of hypoxemia: mild (200 mmHg < PaO
2
/Fio2
300 mmHg), moderate
(100 mmHg < PaO
2
/FiO
2
200 mmHg) and severe (PaO
2
/FiO
2
100 mmHg), along with
explicit criteria related to timing of the syndrome’s onset, origin of edema and the chest
radiographic findings. The ARDS definition task force was considered [14].
Twenty-five HS were matched by age and gender. HS were negative for SARS-CoV-2.
The collection of peripheral blood samples was carried out by venipuncture. In these sub-
jects, there was no suspicion of inflammatory disease or presence of degenerative disorders
such as thyroid and autoimmune diseases, DM, dyslipidemia, arterial hypertension and MS.
The intake of some medications that could interfere with the results of the study such as an-
tioxidant drugs and non-steroidal anti-inflammatory drugs was considered, and the drugs
were suspended 48 h before the obtainment of the sample. Ethical approval to perform the
study was obtained from the local ethics committee on 19 August 2020 (Control-9867/2020,
register REG. CONBIOETICA-09-CEI-011-20160627). A written informed consent for en-
rollment or consent to use data from the patients was obtained directly from them or their
legal surrogates. The protocol was registered (TRIAL REGISTRATION: ClinicalTrials.gov
(accessed on 6 February 2022) Identifier: NCT 04570254). Results related to this study
were previously reported by Chavarría et al. during the pre-treatment and post-treatment
evaluation with antioxidants [
11
]. We observed that the glucose levels were increased since
the admission of the patients. It was therefore considered that, in addition to the main
objective, the evaluation if this increase needed to be explored since it could be related to
the oxidative background present in these patients.
The collection of peripheral blood samples was carried out by venipuncture when
patients entered the ICU and they tested positive by the qRT-PCR test. In total, 34 moderate
and 27 severe COVID-19 patients were included and 25 HS. The blood samples were
centrifuged for 20 min at 936 g and 4
C. The plasma of the samples was placed in 3 or
4 aliquots and stored at 30 C.
2.2. Collection of Samples to Verify Infection by SARS-CoV-2 upon Admission to the Hospital
Paired saliva and nasopharyngeal swab samples were collected from all patients who
were suspected to be infected by SARS-CoV-2. Samples were classified as positive for SARS-
CoV-2 when both the N1 and N2 primer-probe sets were detected. The presence of the SARS-
CoV-2 virus was evaluated using specific probes for the detection of the virus in conjunction
with the real-time reverse transcriptase polymerase chain reaction technique (qRT-PCR).
To evaluate organ dysfunction, the SOFA score (neurologic, respiratory, hemodynamic,
hepatic and hematologic) was calculated at admission [12].
2.3. Detection in Plasma of the N and S Protein Antibodies of the SARS-CoV-2 Virus
The anti-N and anti-S protein antibodies of the SARS-CoV-2 virus were detected using
two ELISA-type immunoassays. The plates were coated with 100
µ
L/well of 5
µ
g/mL of
recombinant SARS-CoV-2 nucleocapsid protein derived from E. coli (CODE: 230-01104)
or 2.5
µ
g/mL of recombinant SARS-CoV-2 Spike protein, S1 subunit derived from E. coli
(CODE: 230-01101), supplied by RayBiotech Company (RayBiotech Inc., Georgia, GA,
USA) and 100
µ
L per well of diluted plasma of the patients and added to the plates. The
absorbance in the plates was measured on an ELISA reader Chromate 4300 (Awareness
Technology, Inc., Palm City, FL, USA), at 450 nm. The percentages of positivity of each
Cells 2022,11, 932 4 of 23
patient, for each type of anti-viral protein antibodies were evaluated and calculated using
the following mathematical model: [(Absorbance at 450 nm of the patient/absorbance
at 450 nm of the negative control)
1]
×
100, for both anti-N protein and anti-S protein
antibodies [15].
2.4. Laboratory Tests
Laboratory tests were made for the COVID-19 patients to determine acute-phase reac-
tants, hemoglobin, leukocytes, lymphocytes, platelets, creatinine, urea nitrogen, glucose,
C-reactive protein (CRP), albumin, D-dimer, ferritin, IL-6 and oxygen saturation. Data from
the patient’s medical history including demographic data, prior illnesses to infection by
SARS-CoV-2, test result for COVID-19 and whether mechanical ventilation were used for
the analysis of the results.
2.5. Glucose, Insulin, and HOMA-IR Concentrations
Glucose concentrations were determined using the enzymatic commercial kit from
Pointe Scientific (Pointe Scientific Inc., Michigan, MO, USA), and insulin was determined
by radioimmunoassay as previously described by Jiménez et al. [
12
]. (The HOMA index of
IR was calculated by HOMAIR = (insulin µU/mL ×glucose mmol/L)/22.5 [16].
2.6. 8-Isoprostane, Vitamin D, H2S, and 3-Nitrotyrosine Concentrations
The kits for the determination of 8-isoprostane and Vitamin D were provided by
Cayman Chemical Company, Michigan MO, USA. (8-isoprostane ELISA kit Item No. 516351
and vitamin D ELISA kit Item No. 501050). This assay is based on the competition between
8-isoprostane and an 8-isoprostane–acetycholinesterase conjugate for a limited number
of 8-isoprostane-specific rabbit antiserum binding sites. The product of this enzymatic
reaction has a distinct yellow color and adsorbs strongly at 412 nm. The assay for Vitamin
D is based on the competition between vitamin D and a conjugated vitamin D-acetyl-
cholinesterase. The product of this enzymatic reaction has a distinctive yellow color and
absorbs strongly at 412 nm. Hydrogen sulfide (H
2
S) was quantified by a commercial
colorimetric assay kit obtained from Elab science Biotechnology Co., Ltd., Houston, TX,
USA. (Cat No. E-BC-K355-M). H
2
S reacts with an acetate solution to form ZnS which can be
dissolved in an alkaline solution. Methylene blue is formed in the presence of Fe
3+
and can
absorb at 665 nm. 3-nitrotyrosine (3-NT) was determined with a kit provided by LifeSpan
BioSciencies, Seattle, WA, USA) (3-nitrotyrosine ELISA kit No. LS-40120). This assay is
based on the competitive ELISA principle and is measured at a wavelength of 450 nm. The
measurements were made using a visible light micro plate reader (Stat Fax 3200 Awareness
Technology Palm City, FL, USA).
2.7. Oxidative Stress Markers
2.7.1. Nitrites (NO2)
The NO
2
levels in plasma were determined by the Griess reaction. In total, 100
µ
L of
plasma previously deproteinated with 0.5 N, NaOH and 10%, ZnSO
4
were centrifuged at
1789
×
gfor10 min. The supernatant was recovered and 200
µ
L of 1% sulfanilamide and
200
µ
L of 0.1% N-naphthyl-ethyl diamine were added. The total volume was adjusted to
1 mL. The calibration curve was obtained with a solution of KNO
2
5–0.156 nM and the
absorbance was measured at 540 nm [17].
2.7.2. Lipid Peroxidation Levels (LPO)
In total, 50
µ
L CH
3
-OH with 4% BHT plus phosphate buffer pH 7.4 were added to
100
µ
L of plasma. The reaction tube was incubated to 100
C for 1 hour and centrifuged
at 936 g at room temperature for 2 min. Then, the n-butanol phase was extracted, and the
absorbance was measured at 532 nm [
18
]. This test is based on the reaction of malondialde-
hyde, a secondary product of the oxidation of fatty acids with three or more bonds, with
Cells 2022,11, 932 5 of 23
thiobarbituric acid in an acid medium and at high temperature, generating a pink-colored
product, the value is expressed in nM of malondialdehyde (MDA) per 1 mL of plasma.
2.7.3. Evaluation of Total Antioxidant Capacity (TAC)
In total, 100
µ
L of plasma were suspended in 1.5 mL of a reaction mixture prepared
as follows: 300 mM acetate buffer pH 3.6, 20 mM FeCl
3
6H
2
O and 10 mM of 2,4,6-Tris-2-
pyridyl-s-triazine dissolved in 40 mM HCl. These reactants were added in a relation of
10:1:1 v/v, respectively. After mixing, the samples were incubated at 37
C for 15 min in
the dark. The absorbance was measured at 593 nm [19].
2.7.4. Thiol Concentrations
The technique used was previously described by Erel and Neselioglu [
20
], with some
adaptations and modifications carried out in our laboratory. In total, 50
µ
L of plasma were
suspended reduced with 100
µ
L of KBH
4
10 mM dissolved in CH
3
OH-bidistilled H
2
O,
(1:1 vol/vol) for 3 min, then, 700
µ
L of buffer (6.7 mM formaldehyde, 10 mM EDTA and
Tris 100 mM, pH 8.2) was added for 3 min. Then, 100
µ
L of DTNB 10 mM in CH
3
OH was
added for 4 min. The calibration curve was obtained with solution GSSG 1 mg/1 mL and
the absorbance was measured at 415 nm.
2.7.5. Glutathione Levels (GSH)
In total, 100
µ
L of plasma previously deproteinized with 20% trichloroacetic acid
(vol/vol) and centrifugated to 10,000
×
gfor 5 min plasma was added to 800
µ
L of phosphate
buffer 50 mM, pH 7.3, plus 100
µ
L of 5, 5
0
-dithiobis-2-nitrobenzoic acid 1 M. The mixture
was incubated at room temperature for 5 min and absorbance was read at 412 nm [18].
2.7.6. Selenium
The technique used was previously described by Soto et al. [
21
]. All solutions were
made with tridistilled H
2
O and were only used for the assay and discarded. In brief, in
new Corning sterile polypropylene centrifuge tubes, 200
µ
L of plasma and 500
µ
L of acid
mixture (4:1 vol/vol of HNO
3
+ HCl) plus 500
µ
L of 10% H
2
O
2
were added and incubated
at 120
C for 4 h. After incubation, 100
µ
L of tridistilled H
2
O, 150
µ
L of 0.5 N NaOH,
200
µ
L of 30% formaldehyde, 200
µ
L of a mixture containing 0.5N of N
2
S and 0.5 N of
Na
2
SO
3
, plus 250
µ
L of 0.01 M of EDTA (pH 10.2), and 300
µ
L of 4 mM of toluidine blue
were added. Samples were incubated for 15min at 25
C. At the end of the incubation,
they were centrifuged at 448rcf for 2 min and the absorbance was read at 600 nm. The
calibration curve was performed using 100 ng/mL Na
2
SeO
3,
and the samples were treated
under conditions similar to those of the experimental samples.
2.8. Statistical Analysis
The Sigma Plot 14 program (Jendel Corporation, San José, CA, USA, 1986–2017) was
used to generate the analysis and graphs. Statistical significance was determined by the
Mann–Whitney rank sum test followed by the normality test (Shapiro–Wilk). Differences
were considered statistically significant when p
0.05. The calculation of the antibodies
against N and S proteins of the SARS-CoV-2 virus was carried out by the analysis of
frequency performed using Fisher’s exact test.
3. Results
3.1. Demographic Characteristics
A total of 61 COVID-19 patients were examined, out of which 44 (72%) were men
and 17 (28%) were women. Patients had an age range of 56
±
13 years. The average body
mass index was 29
±
4 kg/m
2
. Normal weight was found in 13 (21%) and overweight
in 24 (39%). Comorbid conditions prior to SARS-CoV-2 infection were dyslipidemia in
11 (18%), systemic arterial hypertension (SAH) in 7 (8%), DM in 6 (10%), DM plus dys-
lipidemia in 5 (8%), DM plus SAH in 5 (8%), SAH plus dyslipidemia in 3 (5%), SM in 9
Cells 2022,11, 932 6 of 23
(15%), chronic obstructive lung disease in 1 (1.6%) and chronic kidney disease in 2 (3.3%).
Temperature was 36.6
±
0.46
C. Other variables expressed as median and minimum–
maximum ranges, respectively, are included such as arterial blood oxygen pressure (PaO
2
,
66.9,
34–223 mmHg
), partial pressure of carbon dioxide (PCO
2
, 31.7,
12.2–81.2 mmHg
),
Kirby’s index which is PaCO
2
/inspired fraction of oxygen (FiO
2
128, 26.8–299 mmHg),
oxygen saturation (SpO
2
/FiO
2
138, 50–280 mmHg), urea (16,
5.6–106.7 mg/dL
), ureic
nitrogen (16,
5.6–196.7 mg/dL
), total cholesterol (136,
69–217 mg/dL
), triglycerides (133,
62–726 mg/dL
), high-density lipoprotein (31,
14–60 mg/dL
), low-density lipoprotein (70,
28–40 mg/dL
), total bilirubin (0.60,
0.12–4.10 mg/dL
), direct bilirubin (0.20,
10–1.20 mg/dL
),
leukocytes (8.8, 2–25 10
3
/
µ
L), lymphocytes 0.8, 0.14–9.6 10
3
/
µ
L), platelets median (244,
16–576 103/µL
), ferritin 541, 147–2592 ng/mL, CRP 146, 20–2450 mg/L), IL-6 67,
7.8–638.5 pg/mL
) and D-dimer 700, 136–16,400
µ
g/mL). The demographic characteris-
tics of the COVID-19 patients are shown in the Table 1.
Table 1. Demographic characteristics at admission of patients infected with COVID-19.
Total
n= 61 (100) Number
and Percentage
Moderate
n= 34 (56) Number
and Percentage
Severe
n= 27 (44) Number
and Percentage
p
Women 17 (28) 11 (32) 6 (22) NS
Men 44 (72) 23 (68) 21 (78) NS
Age 56 ±13 54 ±12 59 ±14 NS
BMI (kg/m2)29 ±4 29 ±4 29 ±4 NS
Temperature (C) 36.6 ±0.46 36.5 ±0.43 36.7 ±0.49 NS
Laboratory at the admission Median (Min–Max) range
PAO2(58.5–67.1 mmHg) 66.9 (34–223) 67 (34–223) 66.4 (62–93) NS
PCO2(30.4–40 mmHg) 31.7 (12.2–81.2) 32 (12.2–81.2) 31.6 (22–71) NS
PAO2/FIO2(>164) 128 (26.8–299) 145 (26.8–281) 113 (30–299) NS
SpO2/FIO2(>300) 138 (50–280) 157 (88–240) 128 (50–280) 0.007
Urea (<40 mg/dL) 16 (5.6–106.7) 29.2 (16–60) 30 (13–224) NS
Creatinine (mg/dL) 0.90 (0.5–5.3) 1 (0.5–2.5) 0.8 (0.5–5.3) NS
Ureic Nitrogen (7–25 mg/L) 16 (5.6–196-7) 15.7 (7.5–36) 17 (5.6–106.7) NS
TC (<200 mg/dL) 136 (69–217) 140 (69–217) 132 (86–190) NS
HDL (mg/dL) 31 (14–60) 32 (14–60) 31 (14–45) NS
LDL (mg/dL) 70 (28–140) 64 (35–135) 79 (28–140) NS
DHL (mg/dL) 253 (124–515) 233 (128–412) 255 (124–515) NS
TB (mg/dL) 0.60 (0.12–4.10) 0.50 (0.12–1.3) 0.70 (0.33–4.10) 0.02
DB (mg/dL) 0.20 (0.10–1.20) 0.20 (0.10–1.2) 0.20 (0.10–0.8) NS
TG (<150 mg/dL) 133 (62–726) 137 (62–726) 133 (77–328) NS
Leukocytes (3.5–10.3 ×103/µL)8.8 (2–25) 8.2 (2–14) 11.5 (3–25) 0.003
Lymphocytes (0.99–3.2 ×103/µL)0.8 (0.14–9.6) 0.92 (0.42–9.6) 0.69 (0.14–8.2) 0.01
Platelets (150,000–500,000 ×103/µL)244 (16–576) 226 (16–576) 254 (122–412) 0.057
Ferritin (11–307 ng/mL) 541 (147–2592) 513 (147–2100) 592 (175–2592) NS
IL-6 (pg/mL9) 67 (7.8–638.5) 30.2 (7.8–304) 94 (7.8–639) 0.003
Index N/L 11.5 (1–89) 10 (3–89) 13 (1–83) NS
D-Dimer (0–24 µg/mL) 700 (136–16440) 615 (136–5130) 810 (210–16,640) 0.08
CRP (1–3 mg/L) 146 (20–2450) 280 (20–1380) 146 (32–2450) NS
Comorbidities (%)
DM 6 (10) 4 (7) 2 (3) NS
SAH 5 (8) 3 (5) 2 (3) NS
Dyslipidemia 11 (18) 8 (13) 3 (5) NS
DM + Dyslipidemia 5 (8) 2 (3) 3 (5) NS
DM + SAH 5 (8) 2 (3) 3 (5) NS
Cells 2022,11, 932 7 of 23
Table 1. Cont.
Total
n= 61 (100) Number
and Percentage
Moderate
n= 34 (56) Number
and Percentage
Severe
n= 27 (44) Number
and Percentage
p
SAH + Dyslipidemia 3 (5) 0 2 (5) NS
SM 9 (15) 6 (10) 3 (5) NS
Healthy Subjects without comorbidities 17 (28) 8 (13) 9 (15) NS
Normal weight 13 (21) 6 (18) 7 (26) NS
Overweight 24 (39) 17 (50) 7 (26) 0.06
Obesity 24 (39) 11 (33) 13 (48) NS
COPD 1 (1.6) 0 1 (4) NS
ECKD 2 (3.3) 0 2 (8) NS
Norepinephrine 18 (29) 1 (3) 17 (63)
0.0001
Enteral nutrition 26 (43) 22 (65) 4 (15) 0.001
Deaths 1 (1.6) 0 1 (4) NS
Abbreviations: BMI = Body mass index, HR = Heart rate, MAP = Mean arterial pressure,
HDL = High-density lipoproteins
, LDL = Low-density lipoproteins, TB = Total bilirubin, DB = Direct bilirubin,
IL = Interleukin
,
N/L = neutrophil&/lymphocyte
, DM = Diabetes mellitus, SAH = Systemic arterial hyperten-
sion,
CD = Cardiovascular disease
, COPD = Chronic obstructive pulmonary disease, ECKD = End-stage chronic
kidney disease. FiO
2
= inspired fraction of oxygen, PAO
2
= Blood pressure oxygen, PCO
2
= Partial pressure carbon
dioxide, SpO
2
= Arterial oxygen saturation, ECKD = End-stage chronic kidney disease, TC = Total cholesterol,
TG = Triglycerides, MS = Metabolic syndrome.
3.2. Detection of Antibodies against the N and S Proteins of the SARS-CoV-2 Virus
The presence of anti-N protein and anti-S protein antibodies of the SARS-CoV-2 virus
was determined in the present study at the time of admission. In the baseline measurement,
53 of the 61 patients tested positive for the presence of anti-N protein antibodies (86.9%),
while 8 patients, 4 corresponding to the group who had a moderate respiratory disease,
and 4 corresponding to the group of patients who progressed poorly and required the
use of an artificial respirator, showed an absence of anti-N protein antibodies. A week
later, seven of the eight patients who did not have detectable anti-N protein antibodies
at the beginning, already tested positive for the presence of this type of antibody. The
frequencies of positivity of the antibodies against the N and S proteins were of 31/34 with
91.2% frequency and 7/34 with 20.6% frequency, respectively, in patients with a moderate
illness and of 26/27 with 96.3% frequency and 7/27 with 25.9% frequency, respectively,
in patients with severe COVID-19. There was no statistical difference between the groups
with moderate or severe acute respiratory disease in the analyses of frequency performed
using Fisher’s exact test. Therefore, the results suggest that the serological response to the
SARS-CoV-2 virus does not appear to be the condition responsible for progression to a
more severe stage requiring artificial ventilation.
3.3. Glucose, Insulin and HOMA-IR Concentrations
The moderate and severe COVID-19 patients showed a significant increase (p< 0.001)
in glucose, insulin concentrations and HOMA-IR in plasma in comparison with the HS,
(Figure 1a–c, respectively).
Cells 2022,11, 932 8 of 23
Cells 2022, 11, x FOR PEER REVIEW 8 of 22
Figure 1. (a) The values of glucose significantly increased in the moderate and severe COVID-19
patients in comparison with HS. (b) Insulin had a significant increase in the moderate and severe
COVID-19 patients in comparison to HS. (c) The HOMA index significantly increased in the mod-
erate and severe COVID-19 patients in comparison with HS. Abbreviations: HS = healthy subjects,
HOMA index = marker of insulin resistance which if the values are greater than 2.5 is an indication
of insulin resistance.
HS
0
100
200
300
400
500
600
Glucose (mg / dL)
COVID-19
Moderate COVID-19
Severe
p<0.001
p<0.001
HS
0
1
2
3
4
5
COVID-19
Moderate COVID-19
Severe
Insulin (ng / mL)
p<0.001
p<0.001
HS
0
20
40
60
80
HOMA-index
COVID-19
Moderate COVID-19
Severe
p<0.001
p<0.001
A
B
C
a
b
c
Figure 1.
(
a
) The values of glucose significantly increased in the moderate and severe COVID-19
patients in comparison with HS. (
b
) Insulin had a significant increase in the moderate and severe
COVID-19 patients in comparison to HS. (
c
) The HOMA index significantly increased in the moderate
and severe COVID-19 patients in comparison with HS. Abbreviations: HS = healthy subjects, HOMA
index = marker of insulin resistance which if the values are greater than 2.5 is an indication of
insulin resistance.
Cells 2022,11, 932 9 of 23
3.4. 8-Isoprostane, Vitamin D, H2S, and 3-Nitrotyrosine Concentrations
The 8-isoprostane and 3-NT concentrations showed a significant increase (p< 0.001,
Figure 2a,b). However, Vitamin D (p= 0.03 and p= 0.01, Figure 3a) and H
2
S (p< 0.001 and
p= 0.007, Figure 3b) levels showed a decrease in moderate and severe COVID-19 patients
in comparison to HS.
HS
0
200
400
600
800
1000
8-isoprostane (pg/ml of plasma)
COVID-19
Moderate COVID-19
Severe
p<0.001
p<0.001
HS
-0.01
0.00
0.01
0.02
0.03
0.04
0.05
3-NT
(nM / ml plasma)
COVID-19
Moderate COVID-19
Severe
p<0.001
p<0.001
A
B
a
b
Figure 2.
(
a
) The 8-isoprostane concentrations statistically increased in both the moderate and severe
COVID-19 patients in comparison with HS. (
b
) 3-NT statistically increased in the moderate and severe
COVID-19 patients in comparison to HS. Abbreviations: HS = healthy subjects, 3-NT = 3-nitrotyrosine.
Cells 2022,11, 932 10 of 23
Cells 2022, 11, x FOR PEER REVIEW 10 of 22
Figure 3. (a) Changes in the values of Vit D and (b) changes in the concentration of the H2S, there
was a significant decrease in the moderate and severe COVID-19 patients in comparison with HS.
Abbreviations: Vit D = Vitamin D, H2S = hydrogen sulfide, HS = healthy subjects.
3.5. Oxidative Stress Markers
The LPO index showed a significant increase (p = 0.02 and p = 0.004, Figure 4a). How-
ever, the TAC (p < 0.001, Figure 4b), NO2 (p < 0.001, Figure 4c), the thiols concentrations
(p = 0.02 and p = 0.006, Figure 5a) and GSH levels (p < 0.001, Figure 5b) showed a decrease
in moderate and severe COVID-19 patients in comparison with HS.
HS
-20
-10
0
10
20
30
40
50
60
Vit D (ng/ml of plasma)
COVID-19
Moderate COVID-19
Severe
p=0.03
p=0.01
HS
H2S (mmol/L of plasma)
20
25
30
35
40
45
COVID-19
Moderate
p<0.001
p=0.007
COVID-19
Severe
A
B
a
b
Figure 3.
(
a
) Changes in the values of Vit D and (
b
) changes in the concentration of the H
2
S, there
was a significant decrease in the moderate and severe COVID-19 patients in comparison with HS.
Abbreviations: Vit D = Vitamin D, H2S = hydrogen sulfide, HS = healthy subjects.
3.5. Oxidative Stress Markers
The LPO index showed a significant increase (p= 0.02 and p= 0.004, Figure 4a). How-
ever, the TAC (p< 0.001, Figure 4b), NO
2
(p< 0.001, Figure 4c), the thiols concentrations
(
p= 0.02
and p= 0.006, Figure 5a) and GSH levels (p< 0.001, Figure 5b) showed a decrease
in moderate and severe COVID-19 patients in comparison with HS.
Cells 2022,11, 932 11 of 23
Cells 2022, 11, x FOR PEER REVIEW 11 of 22
Figure 4. (a) The LPO index significantly increased in the moderate and severe COVID-19 patients
in comparison with HS. (b) The TAC significantly decreased in the moderate and severe COVID-19
patients in comparison to HS. (c) The NO2 concentration significantly decreased in the moderate
and severe COVID-19 patients in comparison with HS. Abbreviations: LPO = Lipid peroxidation,
TAC = Total antioxidant capacity, NO2 = Nitrites.
0
2
4
6
8
LPO (nM MDA / ml of plasma)
HS COVID-19
Moderate COVID-19
Severe
p=0.02
p=0.004
0
5000
10000
15000
20000
25000
30000
35000
TAC (nM Trolox/ml of plasma)
p<0.001
p<0.001
HS COVID-19
Moderate COVID-19
Severe
A
B
p<0.001
0
20
40
60
80
HS COVID-19
Moderate COVID-19
Severe
NO2- (nM / ml of plasma)
p<0.001
C
a
b
c
Figure 4.
(
a
) The LPO index significantly increased in the moderate and severe COVID-19 patients
in comparison with HS. (
b
) The TAC significantly decreased in the moderate and severe COVID-19
patients in comparison to HS. (
c
) The NO
2
concentration significantly decreased in the moderate
and severe COVID-19 patients in comparison with HS. Abbreviations: LPO = Lipid peroxidation,
TAC = Total antioxidant capacity, NO2= Nitrites.
Cells 2022,11, 932 12 of 23
Cells 2022, 11, x FOR PEER REVIEW 12 of 22
Figure 5. (a) The thiol concentrations significantly decreased in the moderate and severe COVID-19
patients in comparison to HS. (b) GSH concentration statistically decreased in the moderate and
severe COVID-19 patients in comparison with HS. (c) The Se concentration significantly decreased
in both the moderate and severe COVID-19 patients in comparison with HS. Abbreviations: HS =
healthy subjects, GSH = glutathione, Se = selenium.
HS
GSH (mM/ ml of plasma)
0.00
0.01
0.02
0.03
0.04
0.05
0.06
0.07
COVID-19
Moderate COVID-19
Severe
p<0.001
p<0.001
HS
-0.002
0.000
0.002
0.004
0.006
0.008
0.010
0.012
0.014
0.016
Se (nM / ml of plasma)
COVID-19
Moderate COVID-19
Severe
p<0.001
a
b
c
0
5
10
15
20
25
30
35
Thiols (mM /ml of plasma)
p=0.02
p=0.006
HS COVID-19
Moderate COVID-19
Severe
Figure 5.
(
a
) The thiol concentrations significantly decreased in the moderate and severe COVID-
19 patients in comparison to HS. (
b
) GSH concentration statistically decreased in the moderate
and severe COVID-19 patients in comparison with HS. (
c
) The Se concentration significantly de-
creased in both the moderate and severe COVID-19 patients in comparison with HS. Abbreviations:
HS = healthy subjects, GSH = glutathione, Se = selenium.
Cells 2022,11, 932 13 of 23
3.6. Selenium
The Selenium concentration only decreased in severe COVID-19 patients in compari-
son to HS (p< 0.001, Figure 5c).
4. Discussion
In the present study, the presence of anti-N protein, and anti-S protein antibodies of the
SARS-CoV-2 virus was determined at the time of admission of the patients. In most of the
patients, antibodies against these proteins were present at the time of admission and in the
rest of them, the antibodies appeared a few days later. During the analytical validation of
the ELISA to measure anti-N protein antibodies, it was found that up to 11% of subjects may
not have measurable amounts of anti-N protein antibodies at the time of the manifestation
of clinical symptoms. This was because these subjects are slow to produce antibodies and
require a longer time to respond. However, between 4 and 10 days after the onset of the
manifestations of clinical symptoms, patients already have measurable concentrations of
anti-N protein antibodies. These results confirm the known fact that the measurement of
anti-N protein antibodies is the most effective serological measurement for diagnosing
the SARS-CoV-2 virus infection [
15
,
22
]. This is due to the fact that the N protein of the
SARS-CoV-2 virus is a highly immunogenic protein that is overexpressed and released into
the bloodstream simultaneously with the viral particles. Therefore, its measurement is a
highly useful analytical tool for the epidemiological management of the pandemic [
22
].
The low positivity values for anti-S protein antibodies, both in the frequency of positive
patients, and in the percentage of positivity, suggests that these patients had difficulty in
producing neutralizing antibodies, necessary to counteract the infectious capacity of the
SARS-CoV-2 virus [
15
,
22
]. For this reason, these subjects required hospitalization and were
in need of specialized clinical management and even the use of artificial respirators.
On the other hand, several studies have shown that COVID-19 triggers a transient
hyperglycemia and impairs pancreatic
β
-cell function. This has been associated with
inflammation and the cytokine storm that may lead to IR. These changes contribute to a
positive feedback cycle in the development and progression of hyperglycemia in COVID-
19 patients. Hyperglycemia may also induce OS and glucolipotoxicity. However, in
subjects with MS these changes are more aggressive, and comorbidities may lead to fatal
outcomes [
23
]. Our results show that the hyperglycemia, hyperinsulinemia and IR were
present in our series of 61 moderate and severe COVID-19 patients.
The association of the COVID-19 infection and altered glucose metabolism has previ-
ously been reported [
24
], and explained by the capacity of the virus to hijack mitochondrial
function leading to an anaerobic function [
5
]. In this condition, there is an increase in lactate
levels that provokes an increase in the dependency on glycolysis in hepatocytes, which is
reflected in an increase in glucose and lactate levels in the blood. This state favors viral
replication since the virus requires large amounts of energy for biosynthetical process [
5
].
Moreover, the increase in glucose levels elevates the pool of free fatty acids (FFA) which are
necessary for the formation of viral membranes. It also increases nucleotides needed for
RNA synthesis for viral replication [5].
As a consequence of glucose alterations, DM and MS patients have an increased risk of
severe pneumonia by COVID-19 with fatal outcomes. This is reflected in a higher mortality
in subjects with these comorbidities [
25
]. In addition, prolonged hyperglycemia could
worsen the course of COVID-19 via glycation of pancreatic ACE2 and the transmembrane
serine protease 2 (TMPRSS2), thus facilitating the SARS-CoV-2 binding and entrance to
pancreatic
β
-cells [
2
]. However, SARS-CoV-2 may infect the pancreatic
β
-cells through
other proteinases such as neuroplin-1 and transferrin receptor [
26
,
27
]. Studies in human
islets infected
in vitro
and studies in postmortem autopsied tissue found a phenotypic alter-
ation or trans-differentiation of the
β
-cells with decreased insulin and increased glucagon
secretion that could lead to a fatal outcome [
26
,
27
]. However, these studies had great
limitations such as the lack of a determination of circulating insulin concentrations in
COVID-19 patients.
Cells 2022,11, 932 14 of 23
Our results showed that hyperglycemia and hyperinsulinemia were present in plasma
of patients with and without comorbidities upon admission to the ICU. These results agree
with those reported by Montefusco et al. who showed that in 551 patients hospitalized for
COVID-19, the SARS-CoV-2 induces IR and disrupts
β
-cells function which can result in
clinically evident hyperglycemia detectable even in the post-acute phase [
28
]. This suggests
that both the hyperglycemia and hyperinsulinemia depend on the SARS-CoV-2 infection,
the viral replication cycle and the viral load. As the disease progresses, there is an increase
in glucose levels elicited by the demand of the virus for its replication, which favors a
continuous overstimulation of the
β
-cell [
24
]. This results in an increase in insulin levels
that eventually leads to depletion of insulin stores. This worsens hyperglycemia, and
finally deteriorates
β
-cell function.
In vitro
studies have demonstrated that the hepatitis
virus (HCV) infection of human
β
-cells also leads to hypersecretion of insulin in the initial
stages of the infection and that it later causes a reduction in insulin in the
β
-cells [
29
].
Another study in pancreatic cells infected with HCV demonstrated that hyperinsulinemia
and pancreatic
β
-cell hyperfunctionality is aimed to maintain glucose homeostasis [
30
].
Furthermore, hyperinsulinemia may disturb fibrinolysis by elevating the plasminogen
activator inhibitor type 1 and increase thrombosis in COVID-19 patients [31].
An inadequate insulin secretion by decompensated
β
-cells may cause increasingly
higher blood glucose levels that bathe the islet. This leads to a spectrum of consequences
for the
β
-cell, including glucose desensitization,
β
-cell exhaustion, and eventually glu-
cose toxicity [
32
]. Furthermore, this sustained response of the pancreas aimed towards
maintaining glucose homeostasis, leads to the activation of pathways of apoptosis and trans-
differentiation if it does not decrease the infection. These result, with time, in a decreased
secretion of insulin by the
β
-cells and lower glucose-stimulated insulin secretion [
33
]. In
addition, subjects without comorbidities may develop hyperglycemia after three days of
the COVID-19 infection. This elevation in glucose levels can be reversed within 2 weeks.
However, 10% of patients could develop DM later in time. This is important because these
findings are not observed in other viral forms of pneumonia, suggesting the involvement of
the pancreatic axis in the coronavirus infection [
34
]. Furthermore, subjects with DM show
pre- and post-prandial hyperglycemia as well as diabetic ketoacidosis when infected by
SARS-CoV-2 [
34
]. In our experimental series, out of 61 patients infected with SARS-CoV-2,
45 had comorbidities while 17 did not report comorbidities and, nevertheless, all presented
alterations in glucose metabolism [
34
]. Hyperglycemia inactivates the glucose transporters
(GLUT) which are normally triggered by insulin. In fibroblasts infected with the human
cytomegalovirus, the IE72 trans activator of viral promoters decreases the mRNA of GLUT1
and increases the mRNA of GLUT-3, -4 and -8 from the early stage of infection that are
3 times more effective in transporting glucose. This increase is independent from the
Akt-mediated metabolic pathway that depends on insulin concentrations. This suggests
that the SARS-CoV-2 could probably have a similar metabolic pathway in glucose transport
independent of hyperglycemia and hyperinsulinemia present in COVID-19 patients [35].
In addition, our results showed that the IR state in COVID-19 patients is present. In
this sense, it has been described that binding of insulin to the insulin receptor or insulin-like
growth factor 1 receptor (IGF1R) results in the autophosphorylation of insulin receptor
substrate 1/2 (IRS1/2) at its tyrosine residues and in the subsequent activation of two main
pathways, the phosphoinositide-3-kinase (PI3K-Akt) pathway and the mitogen activated
protein kinase (MAPK) pathway. Conversely, serine phosphorylation of the IRS1/2 attenu-
ates insulin signaling by decreasing insulin-stimulated tyrosine phosphorylation and this
can produce IR [
36
]. A study using liver biopsy specimens obtained from non-diabetic
HCV-infected patients showed that HCV impairs the insulin-stimulated tyrosine phospho-
rylation of hepatic IRS1, resulting in reduced PI3K-Akt activation without any alteration in
the MAPK pathway [
37
]. Another study in a transgenic mouse model expressing the HCV
genotype 1 core protein indicated that the core protein was responsible for inducing IR
in the liver via suppression of tyrosine phosphorylation in IRS1, which is associated with
DM [38].
Cells 2022,11, 932 15 of 23
During IR, the action of insulin at the cellular level is reduced in several tissues, which
increases the secretion of this hormone by the pancreas. The HOMA-IR index is used
as an indicator of IR [
39
]. IR is defined by an augmented production of glucose by the
liver, reduced glucose uptake by muscle and elevated lipolysis. In this condition, muscles,
adipose tissue and the liver do not respond to insulin. Moreover, IR is related to a range
of risk factors for cardiovascular disease, including MS, hypertension, OS, dyslipidemia,
inflammation and glucose intolerance [
40
,
41
]. Different studies have demonstrated that
SARS-CoV-2 induces IR [
42
]. Our results show that COVID-19 patients presented an
increase in the HOMA index. This result implies that during the progression of the
infection, IR forces
β
-cells to elevate the synthesis of insulin trying to restore the normal
blood glucose level [
43
]. Furthermore, IR leads to an increased pancreatic expression of
the ACE2 receptors, increasing the binding affinity for the S protein of the SARS-CoV-2.
Therefore, there is an increased vulnerability for the COVID-19 infection in subjects with
IR [44].
The alterations in the glucose–insulin axis in COVID-19 patients with or without
comorbidities, suggest that it is very important to control the glycemic state using dif-
ferent drugs such as the metformin, which improves IR and peripheral glucose uptake
through the activation of AMP-dependent protein kinase [
45
]. Metformin also exerts
pleiotropic effects through the AMP-independent pathway including anti-inflammatory
and immunomodulatory effects and it also inhibits the synthesis and release of CRP, IL-1
β
-
induced IL-6, and ferritin from macrophages, endothelial cells, smooth muscle vascular
cells and hepatocytes [
46
]. Metformin also reduces the binding of SARS-CoV-2 to ACE2 by
inducing functional changes in the transmembrane enzyme by AMP-dependent phospho-
rylation [47].
Hyperglycemia, hyperinsulinemia and IR contribute to increase ROS that attack free
fatty acid (FFA) or esterified arachidonic acid in the phospholipids of the membrane and
constitute conditions that are frequently found in COVID-19 and in patients with MS.
Hyperglycemia, hyperinsulinemia and IR increase the rate of lipolysis which is accompa-
nied with liberation of FFA to the circulation. An increased fatty acid (FA) metabolism is
essential for the formation of the viral membranes including those of SARS-CoV-2 [
48
].
Furthermore, the increase in FFA and the alteration in FA metabolism increase gluconeo-
genesis, reduce
β
-cells mass and provide substrates for the synthesis of triglycerides that
are elevated in COVID-19 patients [
48
]. The attack of the membrane phospholipids and
arachidonic acid by ROS result in the formation of isoprostanes that are molecules that are
considered as markers of LPO and oxidative injury. In this sense, 8-isoprostane has been
considered an ideal marker for pathophysiology such as bronchoalveolar lavage fluid of
patients with interstitial lung disease [
49
]. Our results show that 8-isoprostanes and IL-6
were elevated in both moderate and severe COVID-19 patients. A study in rats with EPOC
also showed an increase in 8-isoprostanes and IL-6 in lung lysates associated with ACE2
overexpression and another study demonstrated an increase in urinary 8-isoprostanes
levels in patients with chronic obstructive pulmonary disease [
50
,
51
]. This suggests that
the 8-isoprostanes are associated in pneumonia by SASR-CoV-2 and comorbidities present
in MS and that their synthesis is a consequence of the alteration of the FA metabolism
and the oxidative background. These alterations contribute to ACE2 over-expression that
facilitates the entrance of the virus into the host cells. However, more investigations are
necessary to verify this hypothesis. In addition, the alterations in the glucose–insulin axis
and FA metabolism [
48
] may lead to an increase in OS, increased inflammation, interleukin
storm, and decreased mobilization of leukocytes, phagocytic activity, TAC and impairment
of endothelial function [
52
]. In this sense, it has been reported that the TAC levels were
considerably lower in serum of mild and severe COVID-19 patients in comparison with
control subjects [
53
]. Our results showed alterations in some markers of OS such as LPO,
TAC, thiols, 8-isoprotanes, GSH, 3-NT and NO
2
concentrations. In ARDS and COPD
associated with COVID-19, the hypoxic conditions result in inflammation that may lead to
OS. Increased OS leads, as a consequence, to a decrease in nitric oxide (NO) release mainly
Cells 2022,11, 932 16 of 23
because ROS interfere with the activity of the NO synthases and cause an inadequate
production of tetrahydrobiopterin [
54
]. Optimal concentrations of O
2
are required for the
synthesis of NO by the nitric oxide synthases.
Changes in the activity of these synthases are reflected in a decrease in their metabo-
lites including NO
2
. This metabolite was decreased in moderate and severe patients in
our study. Furthermore, the newly synthetized NO might be oxidized by ROS to peroxini-
trite (ONOO
) which is the most aggressive RNS and may contribute to the inflammatory
process associated with the interleukin storm in the COVID-19 patients. Low levels of NO
also induce proliferation of vascular smooth muscle cells, platelet aggregation, elevated
pro-inflammatory cytokines such as IL-6, chemokine expression, low-density lipoprotein
oxidation, and expression of vascular cell adhesion molecule-1, stimulation of thrombolysis
and monocyte chemotactic protein-1 through the inhibition of the NF-
κ
B signaling pathway.
Moreover, decreased NO following an oxidative burst can stimulate the production of
metalloproteinases-2 and -9 that contribute to pulmonary damage [
55
]. Hypoxia also in-
creases the presence of protons, in a similar way as in a neutral or acidic pH, compromising
the stability of ONOO
and decomposing peroxynitrous acid to form nitrates (NO
3
) and
NO
2
[
54
]. ONOO
causes the irreversible process of nitration of tyrosines in proteins,
it oxidizes the thiol groups in them [
56
] and it also lowers the H
2
S concentration. In this
sense, H
2
S is a potent gasotransmitter that is decreased in COVID-19 patients [
57
]. Recent
studies provide evidence of the beneficial role of H
2
S in COVID-19 patients. In 10 patients
with severe COVID-19 intravenous N-acetylcysteine a potential H
2
S releaser decreased
the inflammation [
58
]. H
2
S is able to positively modulate concentrations of cytokines by
reducing pro-inflammatory IL-6 and TNF-
α
. This suggests that the decrease in the H
2
S
levels contributes in part to a pro-inflammatory state in SARS-CoV-2 infection. In addition,
moderate or high IL-6 levels have been associated with the decrease in the cysteine and
taurine concentration which are essential amino acids for H
2
S synthesis [
59
]. Our results
show that 3-NT in plasma proteins was increased both in moderate and severe patients.
This result suggests that COVID-19 courses with nitrosative stress (NSS) [
5
,
60
], which is
associated with ferroptosis [5].
Ferroptosis results from mitochondrial sequestration by the viruses and contributes to
an oxidative environment by increasing the Fenton–Haber–Weiss reaction which results
in OS and NSS production [
5
], and in the ferroptosis present in COVID-19 patients [
61
].
The NSS and OS damage disulphide bonds (thiols), which are necessary to stabilize the
architecture of proteins. Viruses and pathogens rely on the proper redox state for their
–S–S– bonds or sulfhydryl groups that are needed for their entrance to the host cells. The S1
subunit is responsible for receptor binding and it contains the receptor-binding domain
which contains disulphide bridges that are indispensable for the union with the ACE2.
Moreover, the ACE2 receptor and the antioxidants enzymes also contain disulphide bridges,
and therefore, dynamic disulphide bridge homeostasis is very important for both viral
replication and for the antioxidant defense in the patient [
3
]. Our results show that the level
of thiols in plasma from patients with COVID-19 was decreased in moderate and severe
cases. This result is similar to the one found in another study where it was demonstrated
that COVID-19 patients’ course with low levels of thiols and that their level can even
be a marker that reflects the gradual increase in the severity of the infection [
62
]. This
result is very important because different investigations have reported that the treatment
with agents capable of elevating the reducing thiol groups such as GSH, allicin, garlic and
N-acetylcysteine, can restore the homeostasis of thiols and decrease the degree of viral
infection increasing the TAC and decreasing the LPO in COVID-19 patients [63].
On another hand, the low levels of H
2
S favor a decrease in the thiol concentration
because this gasotransmitter reduces the –S–S– in the proteins and enzymes and increases
the expression of TMPRSS2 which facilities the SARS-CoV-2 entrance into cells [
64
]. Low
levels of H
2
S have been reported in ARDS and COPD associated with the SARS-CoV-2
infection [
65
]. In this sense, exogenously applied H
2
S donors such as NAC protect from
lung damage, including that produced by ARDS, COPD, ALI, asthma pulmonary fibrosis
Cells 2022,11, 932 17 of 23
and hypoxia-induced pulmonary hypertension in animal models and in humans. This
therapy has therefore been proposed as an alternative strategy in this pandemic [66].
Moreover, levels of Vitamin D are low in diseases that course with hyperinsulinemia
such as COVID-19 and in the pathologies that comprise the MS [
67
,
68
]. Vitamin D exerts its
effects by binding to a nuclear Vitamin D receptor, which is expressed in various immune
cells, with particularly high levels in dendritic cells, macrophages, T and B lymphocytes that
are increased when the innate immune response is elicited [
69
]. During hyperinsulinaemic
states, Vitamin D3 is sequestered into adipocytes, and inactivated in the kidney [
42
]. Our
results show that Vitamin D levels were decreased in moderate and severe COVID-19
patients. This suggests that hyperinsulinemia may decrease the concentration of this
vitamin having an unfavorable impact upon the innate immune response. Moreover,
this deficiency may increase the interaction of the S protein with ACE2, because its low
levels favor an increase in the TMPRSS2 protease which is essential for the entrance of the
virus [
70
]. Vitamin D deficiency also decreases innate cellular immunity by lowering the
expression of defensins which maintain the gap junctions in the endothelial cells of the lung.
Rupture of these junctions is caused by the SARS-CoV-2 leading to ARDS and pulmonary
edema [
71
]. High concentrations of Vitamin D may also have benefic effects such as the
induction of the vasorelaxant ACE2/Ang-(1-7)/Mas receptor axis, which protects against
acute lung injury and ARDS [
72
]. In addition, low levels of Vitamin D are associated with Se
deficiency and are related to COVID-19 severity, old age, obesity, diabetes and dyslipidemia.
However, in COVID-19 patients without comorbidities, there is also a decrease in Vitamin D
and Se in comparison with the healthy subjects [
68
]. The participation of Se is also important
in COVID-19 because it has synergic effects with Vitamin D and E acting as antioxidants.
Furthermore, Se is indispensable for the presence of 25 seleno-enzymes such glutathione
peroxidase, thioredoxin reductases (TrxR), deiodinases and methionine sulfoxide reductases
which play an important role in maintaining the redox homeostasis [
73
]. Our results show
that the concentrations of Se and GSH were decreased in severe COVID-19 patients. This
suggests that the redox homeostasis which is dependent on the 25 seleno-enzymes is altered
and this condition is associated with severe COVID-19. Moreover, this impacts on the
presence of low thiols GSH and H
2
S concentrations in plasma, which contribute to an
increase in OS and NSS present in these patients. This is reflected in the rise of the LPO
index accompanied with depletion of the TAC. In this sense, Se deficiency in mice was
associated with enhanced virulence of enteroviruses and the development of myocardial
lesions [
73
]. The homeostasis of thiols and GSH depend on TrxR and glutathione reductase
which are seleno-enzymes. Low levels of the GSH in COVID-19 patients are associated
with ferroptosis and with a down-regulation of GPX4 and TrxR [
5
,
61
]. A deficiency in
GSH is linked to HIV progression and poor survival of HIV-infected individuals [
74
], and
high concentrations of GSH in blood decrease the virulence of the infection by dengue
and chikungunya [
75
]. Our results suggest that a reduction in the GSH concentration may
contribute to an increase in OS and NSS and favor the decrease and increase, respectively,
in the TAC and LPO in COVID-19 patients. Figure 6summarizes the alterations in the
glucose–insulin axis by SARS-CoV-2.
Cells 2022,11, 932 18 of 23
Cells 2022, 11, x FOR PEER REVIEW 18 of 22
Figure 6. Alteration of the glucoseinsulin axis by SARS-CoV-2, and the impact on some antioxidant
markers in patients. (1) Entry of COVID-19 through various receptors in the pancreas. (2) COVID-
19 increases lactate levels, favoring viral replication. (3) Increased free fatty acids are used for viral
membrane formation. (4) Increased glucose overstimulates pancreatic β-cells and subsequently im-
pairs their function. (5) Chronically increased insulin concentrations will lead to depletion of insulin
reserves. (6) Hyperinsulinemia may favor the development of thrombosis. (7) Hyperglycemia inac-
tivates GLUT 1 but SARS-CoV-2 could increase -3, -4 and -8, transporters. (8) Increased lipolysis
blocks the response to insulin by the liver, adipose tissue and muscles. (9) Metformin prevents the
interaction of the ACE2 receptor and COVID-19. (10) Increased free fatty acid synthesis favors the
generation of 8-isoprostanes, which favor ACE2 receptor overexpression. (11) ONOO favors inter-
leukin storm. (12) ONOO decreases H2S concentration. (13) Nitrosative stress and depletion of GSH
concentrations is associated with ferroptosis and mitochondrial sequestration by the virus. (14) Un-
der conditions of hyperinsulinemia, Vitamin D is sequestered in adipose tissue and inactivated in
the kidney, which favors the interaction of virus S protein with the ACE2 receptor. (15) Vitamin D
deficiency generates ruptures in the gap junctions of lung endothelial cells. (16) Vitamin D defi-
ciency is associated with Se deficiency which affects selenoenzymes. Abbreviations: ARDS = acute
respiratory distress syndrome, H2O2 = hydrogen peroxide, H2S = sulfhydryl acid, G-6-P = glucose 6
phosphate, GR = glutathione reductase, GSH = glutathione, GSSG = oxidized glutathione, NO = ni-
tric oxide, NSS = nitrosative stress, O2 = superoxide, ONOO = peroxynitrite, ROS = reactive oxygen
species, SOD = Superoxide dismutase.
5. Conclusions
The results suggest that infection with SARS-CoV-2 in patients with and without
comorbidities results in alterations in the glucoseinsulin axis which leads to hyperglyce-
mia, hyperinsulinemia and IR. These alterations increase OS and NSS that is reflected in
increases or decreases in some oxidative markers in plasma with major impacts or fatal
consequences in patients that course with MS. Moreover, subjects without comorbidities
could have long-term alterations in the redox homeostasis after infection.
Figure 6.
Alteration of the glucose–insulin axis by SARS-CoV-2, and the impact on some an-
tioxidant markers in patients. (
1
) Entry of COVID-19 through various receptors in the pancreas.
(
2
) COVID-19 increases lactate levels, favoring viral replication. (
3
) Increased free fatty acids are
used for viral membrane formation. (
4
) Increased glucose overstimulates pancreatic
β
-cells and
subsequently impairs their function. (
5
) Chronically increased insulin concentrations will lead
to depletion of insulin reserves. (
6
) Hyperinsulinemia may favor the development of thrombo-
sis. (
7
) Hyperglycemia inactivates GLUT 1 but SARS-CoV-2 could increase -3, -4 and -8, trans-
porters. (
8
) Increased lipolysis blocks the response to insulin by the liver, adipose tissue and muscles.
(
9
) Metformin prevents the interaction of the ACE2 receptor and COVID-19. (
10
) Increased free
fatty acid synthesis favors the generation of 8-isoprostanes, which favor ACE2 receptor over-
expression. (
11
) ONOO
favors interleukin storm.
(12) ONOO
decreases H
2
S concentration.
(
13
) Nitrosative stress and depletion of GSH concentrations is associated with ferroptosis and mi-
tochondrial sequestration by the virus. (
14
) Under conditions of hyperinsulinemia, Vitamin D is
sequestered in adipose tissue and inactivated in the kidney, which favors the interaction of virus S pro-
tein with the ACE2 receptor. (
15
) Vitamin D deficiency generates ruptures in the gap junctions of lung
endothelial cells.
(16) Vitamin D
deficiency is associated with Se deficiency which affects selenoen-
zymes. Abbreviations:
ARDS = acute respiratory distress syndrome
,
H2O2= hydrogen peroxide
,
H
2
S = sulfhydryl acid, G-6-P = glucose 6 phosphate, GR = glutathione reductase,
GSH = glutathione
,
GSSG = oxidized glutathione
, NO = nitric oxide,
NSS = nitrosative stress
,
O2= superoxide
,
ONOO= peroxynitrite, ROS = reactive oxygen species, SOD = Superoxide dismutase.
5. Conclusions
The results suggest that infection with SARS-CoV-2 in patients with and without
comorbidities results in alterations in the glucose–insulin axis which leads to hyperglycemia,
hyperinsulinemia and IR. These alterations increase OS and NSS that is reflected in increases
or decreases in some oxidative markers in plasma with major impacts or fatal consequences
Cells 2022,11, 932 19 of 23
in patients that course with MS. Moreover, subjects without comorbidities could have
long-term alterations in the redox homeostasis after infection.
5.1. Study Limitations
One of the limitations of this study is that we could not study SARS-CoV-2-infected
subjects with mild symptoms or asymptomatic COVID-19. The center in which this work
was carried out was a reference center and received patients with moderate to severe
symptoms sent from other hospitals. Another limitation is that in the evaluation of certain
parameters such as D-Dimer, Ferritin, N/L index, there is a tendency to an elevation in
moderate and critical patients; however, this cannot be defined in a concrete way because a
larger sample size would be required for this specific evaluation. However, it is known that
these parameters increase, and our results show this same tendency. It is important to state
that the sample for this study was adjusted for the specific question of the investigation.
One of the limitations of this study is also that we were not able to measure markers with
thromboelastography to define the co-participation of a hyperglycemic state with changes
in PAI-1 and fibrinolysis. It is currently known that a subset of patients with COVID-19
have a higher risk of bleeding, and high levels of tissue-type plasminogen activator (tPA)
and plasminogen activator inhibitor 1 (PAI- 1) have been associated with a worse lung
function. An elevated tPA is independently correlated with mortality. The levels of any of
these molecules can increase independently of the other; however, a change in one can have
consequences on another. The interaction of plasminogen activators, both tissue-type (tPA)
and urokinase-type, and their main inhibitor, PAI-1, play a key role in the regulation of
fibrinolytic activity. Impaired fibrinolysis has been suggested in COVID-19 patients, which
could further increase thrombotic risk. This has been evidenced by markedly reduced clot
lysis at 30 min by thromboelastography in critically ill COVID-19 patients [
76
]. Another
limitation of the study is that the oxidative markers can be altered by the age, gender
and pathological comorbidities which was not contemplated in this work but would be
interesting to address in a future investigation.
5.2. Perspectives
According to the results of our study, the use of a combined therapy with antioxidants
such as NAC and metformin could attenuate SARS-CoV-2 infection in patients with COVID-
19, as has already been shown in various studies [11].
Author Contributions:
I.P.-T., M.E.S. and V.G.-L. designed the study and wrote the manuscript.
V.G.-L
. revised and structured the manuscript. I.P.-T. and M.E.S. designed the laboratory determina-
tions, the tables, performed, and planned the statistical analysis, contributed to the conceptualization
of the project, methodology and statistical analysis. E.D.-D. designed and made glucose, insulin,
HOMA index and detection of N and S proteins antibodies. L.M.-P. designed and made the graphical
abstract and Selenium determination. A.P.-C., R.R.V.-V., A.A.-Á. and H.S.-O. treated and recruited
the patients in the intensive care unit and collected the biochemical results. All authors have read
and agreed to the published version of the manuscript.
Funding:
This work was supported by Consejo Nacional de Ciencia y Tecnologia (CONACYT)
México, Project Number 312167.
Institutional Review Board Statement:
The studies involving human participants were reviewed
and approved by ethical approval from the local ethics committee on 19 August 2020 (Control-
9867/2020, register REG. CONBIOETICA-09CEI-011-20160627). A written informed consent for
enrollment or consent to use patient data was obtained from each patient or their legal surrogate. The
protocol was registered (TRIAL REGISTRATION: ClinicalTrials.gov Identifier: NCT 04570254).
Informed Consent Statement:
The patients/participants provided their written informed consent
to participate in this study. Written informed consent was obtained from the individual(s) for the
publication of any potentially identifiable images or data included in this article.
Data Availability Statement:
The datasets generated and analyzed during the current study are
available from the corresponding author on reasonable request.
Cells 2022,11, 932 20 of 23
Acknowledgments:
Thank to Instituto Nacional de Cardiología “Ignacio Chávez” for the payment
for the open access to this paper.
Conflicts of Interest:
The authors declare that they have no known competing financial interest or
personal relationships that could have appeared to influence the work reported in this paper.
References
1.
Soto, M.E.; Guarner-Lans, V.; Soria-Castro, E.; Manzano-Pech, L.; Pérez-Torres, I. Is antioxidant therapy a useful complementary
measure for COVID-19 treatment? An algorithm for its application. Medicina 2020,56, 386. [CrossRef] [PubMed]
2.
Landstra, C.P.; de Koning, E.J.P. COVID-19 and diabetes: Understanding the interrelationship and risks for a severe course. Front.
Endocrinol. 2021,12, 649525. [CrossRef] [PubMed]
3.
Pérez-Torres, I.; Manzano-Pech, L.; Rubio-Ruíz, M.E.; Soto, M.E.; Guarner-Lans, V. Nitrosative stress and its association with
cardiometabolic disorders. Molecules 2020,25, 2555. [CrossRef] [PubMed]
4.
Cuschieri, S.; Stephan, G. COVID-19 and diabetes: The why, the what and the how. J. Diabetes Complicat.
2020
,34, 107637.
[CrossRef] [PubMed]
5.
Soria-Castro, E.; Soto, M.E.; Guarner-Lans, V.; Rojas, G.; Perezpeña-Diazconti, M.; Críales-Vera, S.A.; Manzano-Pech, L.; Pérez-
Torres, I. The kidnapping of mitochondrial function associated with the SARS-CoV-2 infection. Histol. Histopathol.
2021
,36,
947–965.
6.
Mahrooz, A.; Muscogiuri, G.; Buzzetti, R.; Maddaloni, E. The complex combination of COVID-19 and diabetes: Pleiotropic
changes in glucose metabolism. Endocrine 2021,72, 317–325. [CrossRef]
7.
Barberis, E.; Timo, S.; Amede, E.; Vanella, V.V.; Puricelli, C.; Cappellano, G.; Raineri, D.; Cittone, M.G.; Rizzi, E.;
Pedrinelli, A.R.; et al
. Large-scale plasma analysis revealed new mechanisms and molecules associated with the host
response to SARS-CoV-2. Int. J. Mol. Sci. 2020,21, 8623. [CrossRef]
8.
Ceriello, A.; De Nigris, V.; Prattichizzo, F. Why is hyperglycaemia worsening COVID-19 and its prognosis? Diabetes Obes. Metab.
2020,22, 1951–1952. [CrossRef]
9.
Paul, A.K.; Hossain, M.K.; Mahboob, T.; Nissapatorn, V.; Wilairatana, P.; Jahan, R.; Jannat, K.; Bondhon, T.A.; Hasan, A.; de
Lourdes, P.M.; et al. Does oxidative stress management help alleviation of COVID-19 symptoms in patients experiencing diabetes?
Nutrients 2022,14, 321. [CrossRef]
10.
Delgado-Roche, L.; Mesta, F. Oxidative stress as key player in severe acute respiratory syndrome coronavirus (SARS-CoV)
infection. Arch. Med. Res. 2020,51, 384–387. [CrossRef]
11.
Chavarría, A.P.; Vázquez, R.R.V.; Cherit, J.G.D.; Bello, H.H.; Suastegui, H.C.; Moreno-Castañeda, L.; Alanís-Estrada, G.;
Hernández, F.; González-Marcos, O.; Saucedo-Orozco, H.; et al. Antioxidants and pentoxifylline as coadjuvant measures to
standard therapy to improve prognosis of patients with pneumonia by COVID-19. Comput. Struct. Biotechnol. J.
2021
,19,
1379–1390. [CrossRef] [PubMed]
12.
Lambden, S.; Laterre, P.F.; Levy, M.M.; Francois, B. The SOFA score-development, utility, and challenges of accurate assessment in
clinical trials. Crit. Care 2019,23, 374. [CrossRef] [PubMed]
13. Horovitz, J.H.; Carrico, C.J.; Shires, G.T. Pulmonary response to major injury. Arch. Surg. 1974,108, 349–355. [CrossRef]
14. Ranieri, V.M.; Rubenfeld, G.D.; Thompson, B.T.; Ferguson, N.D.; Caldwell, E.; Fan, E. Acute respiratory distress syndrome: The
Berlin Definition. JAMA 2012,307, 2526–2533.
15.
Jiménez, I.U.; Díaz-Díaz, E.; Castro, J.S.; Ramos, J.P.; León, M.C.; Alvarado-Ríos, J.A.; Auriostigue, B.J.C.; Correa-Rotter, R.;
Aguilar, S.C.A.; Larrea, F. Circulating concentrations of advanced glycation end products, its association with the development of
diabetes mellitus. Arch. Med. Res. 2017,48, 360–369. [CrossRef]
16.
Matthews, D.R.; Hosker, J.P.; Rudenski, A.S.; Naylor, B.A.; Treacher, D.F.; Turner, R.C. Homeostasis model assessment: Insulin
resistance and beta-cell function from fasting plasma glucose and insulin concentrations in man. Diabetologia
1985
,28, 412–419.
[CrossRef]
17.
Griess, P. Bemerkungen zu der Abhandlung der HH. Weselsky und Benedikt Ueber einige Azoverbindungen. Ber. Deutsch. Chem.
Ges. 1879,12, 426–428. [CrossRef]
18.
Manzano-Pech, L.G.; Caballero-Chacón, S.C.; Guarner-Lans, V.; Díaz-Díaz, E.; Moreno, G.A.; Pérez-Torres, I. Effect of
oophorosalpingo-hysterectomy on serum antioxidant enzymes in female dogs. Sci. Rep. 2019,9, 9674. [CrossRef]
19.
Benzie, I.F.; Strain, J.J. The ferric reducing ability of plasma (FRAP) as a measure of “antioxidant power”: The FRAP assay. Anal.
Biochem. 1996,239, 70–76. [CrossRef]
20.
Erel, O.; Neselioglu, S. A novel and automated assay for thiol/disulphide homeostasis. Clin. Biochem.
2014
,47, 326–332.
[CrossRef]
21.
Soto, M.E.; Manzano-Pech, L.G.; Guarner-Lans, V.; Díaz-Galindo, J.A.; Vásquez, X.; Castrejón-Tellez, V.; Gamboa, R.; Huesca, C.;
Fuentevilla-Alvárez, G.; Pérez-Torres, I. Oxidant/antioxidant profile in the thoracic aneurysm of patients with the Loeys-Dietz
syndrome. Oxid. Med. Cell. Longev. 2020,2020, 5392454. [CrossRef] [PubMed]
22.
González-Fiallo, S.; Mena-Rodríguez, I.; Doeste-Hernández, V.M.; Castro-Batista, P.; Espinosa-Reyes, S. COVID-19 rapid test
validation. Isla de la Juventud, Cuba. Vaccimonitor 2021,30, 105–114.
Cells 2022,11, 932 21 of 23
23.
Pérez-Torres, I.; Castrejón-Téllez, V.; Soto, M.E.; Rubio-Ruiz, M.E.; Manzano-Pech, L.; Guarner-Lans, V. Oxidative stress, plant
natural antioxidants, and obesity. Int. J. Mol. Sci. 2021,22, 1786. [CrossRef] [PubMed]
24.
Müller, J.A.; Groß, R.; Conzelmann, C.; Krüger, J.; Merle, U.; Steinhart, J.; Weil, T.; Koepke, L.; Bozzo, C.P.; Read, C.; et al.
SARS-CoV-2 infects and replicates in cells of the human endocrine and exocrine pancreas. Nat. Metab.
2021
,3, 149–165. [CrossRef]
25.
Zhang, Y.; Wang, S.; Xia, H.; Guo, J.; He, K.; Huang, C.; Luo, R.; Chen, Y.; Xu, K.; Gao, H.; et al. Identification of monocytes
associated with severe COVID-19 in the PBMCs of severely infected patients through single-cell transcriptome sequencing.
Engineering 2021, in press. [CrossRef]
26.
Tang, X.; Uhl, S.; Zhang, T.; Xue, D.; Bo, L.; Vandana, J.J.; Acklin, J.A.; Bonnycastle, L.L.; Narisu, N.; Erdos, M.R.; et al. SARS-CoV-2
infection induces beta cell transdifferentiation. Cell. Metab. 2021,33, 1577–1591.e7. [CrossRef]
27.
Wu, C.T.; Lidsky, P.V.; Xiao, Y.; Lee, I.T.; Cheng, R.; Nakayama, T.; Jiang, S.; Demeter, J.; Bevacqua, R.J.; Chang, C.A.; et al.
SARS-CoV 2 infects human pancreatic beta cells and elicits beta cell impairment. Cell. Metab.
2021
,33, 1565–1576.e5. [CrossRef]
28.
Montefusco, L.; Ben-Nasr, M.; D’Addio, F.; Loretelli, C.; Rossi, A.; Pastore, I.; Daniele, G.; Abdelsalam, A.; Maestroni, A.;
Dell’Acqua, M.; et al. Acute and long-term disruption of glycometabolic control after SARS-CoV-2 infection. Nat. Metab.
2021
,3,
774–785. [CrossRef]
29.
Huang, J.F.; Yu, M.L.; Dai, C.Y.; Chuang, W.L. Glucose abnormalities in hepatitis C virus infection. Kaohsiung J. Med. Sci.
2013
,29,
61–68. [CrossRef]
30.
Kawaguchi, Y.; Mizuta, T. Interaction between hepatitis C virus and metabolic factors. World. J. Gastroenterol.
2014
,20, 2888–2901.
[CrossRef]
31.
Douglas, M.W.; George, J. Molecular mechanisms of insulin resistance in chronic hepatitis C. World. J. Gastroenterol.
2009
,15,
4356–4364. [CrossRef] [PubMed]
32. Robertson, R.P. Beta-cell deterioration during diabetes: What’s in the gun? Trends Endocrinol. Metab. 2009,20, 388–393.
33.
Clark, A.L.; Mirmira, R.G. SARS-CoV-2 infection of islet
β
cells: Evidence and implications. Cell. Rep. Med.
2021
,2, 100380.
[CrossRef]
34.
Zhu, L.; She, Z.G.; Cheng, X.; Qin, J.J.; Zhang, X.J.; Cai, J.; Lei, F.; Wang, H.; Xie, J.; Wang, W.; et al. Association of blood glucose
control and outcomes in patients with COVID-19 and pre-existing type 2 diabetes. Cell Metab.
2020
,31, 1068–1077.e3. [CrossRef]
35.
Yu, Y.; Maguire, T.G.; Alwine, J.C. Human cytomegalovirus activates glucose transporter 4 expression to increase glucose uptake
during infection. J. Virol. 2011,85, 1573–1580. [CrossRef] [PubMed]
36.
Tanti, J.F.; Jager, J. Cellular mechanisms of insulin resistance: Role of stress-regulated serine kinases and insulin receptor substrates
(IRS) serine phosphorylation. Curr. Opin. Pharmacol. 2009,9, 753–762. [CrossRef] [PubMed]
37.
Aytug, S.; Reich, D.; Sapiro, L.E.; Bernstein, D.; Begum, N. Impaired IRS-1/PI3-kinase signaling in patients with HCV: A
mechanism for increased prevalence of type 2 diabetes. Hepatology 2003,38, 1384–1392. [CrossRef] [PubMed]
38.
Shintani, Y.; Fujie, H.; Miyoshi, H.; Tsutsumi, T.; Tsukamoto, K.; Kimura, S.; Moriya, K.; Koike, K. Hepatitis C virus infection and
diabetes: Direct involvement of the virus in the development of insulin resistance. Gastroenterology
2004
,126, 840–848. [CrossRef]
39.
Pérez-Torres, I.; Gutiérrez-Alvarez, Y.; Guarner-Lans, V.; Díaz-Díaz, E.; Manzano-Pech, L.; Caballero-Chacón, S.D.C. Intra-
abdominal fat adipocyte hypertrophy through a progressive alteration of lipolysis and lipogenesis in metabolic syndrome rats.
Nutrients 2019,11, 1529. [CrossRef]
40.
Mirzaei, F.; Khodadadi, I.; Vafaei, S.A.; Abbasi-Oshaghi, E.; Tayebinia, H.; Farahani, F. Importance of hyperglycemia in COVID-19
intensive-care patients: Mechanism and treatment strategy. Prim. Care. Diabetes 2021,15, 409–416. [CrossRef]
41.
Singh, A.K.; Singh, R. Hyperglycemia without diabetes and new-onset diabetes are both associated with poorer outcomes in
COVID-19. Diabetes Res. Clin. Pract. 2020,167, 108382. [CrossRef] [PubMed]
42.
Govender, N.; Khaliq, O.P.; Moodley, J.; Naicker, T. Insulin resistance in COVID-19 and diabetes. Prim. Care. Diabetes
2021
,15,
629–634. [CrossRef] [PubMed]
43.
Daryabor, G.; Atashzar, M.R.; Kabelitz, D.; Meri, S.; Kalantar, K. The Effects of Type 2 diabetes mellitus on organ metabolism and
the immune system. Front. Immunol. 2020,11, 1582. [CrossRef] [PubMed]
44.
Santos, A.; Magro, D.O.; Evangelista-Poderoso, R.; Saad, M.J.A. Diabetes, obesity, and insulin resistance in COVID-19: Molecular
interrelationship and therapeutic implications. Diabetol. Metab. Syndr. 2021,13, 23. [CrossRef] [PubMed]
45.
Cory, T.J.; Emmons, R.S.; Yarbro, J.R.; Davis, K.L.; Pence, B.D. Metformin suppresses monocyte immunometabolic activation by
SARS-CoV-2 spike protein subunit 1. Front. Immunol. 2021,12, 733921. [CrossRef]
46.
Zangiabadian, M.; Nejadghaderi, S.A.; Zahmatkesh, M.M.; Hajikhani, B.; Mirsaeidi, M.; Nasiri, M.J. The Efficacy and potential
mechanisms of metformin in the treatment of COVID-19 in the diabetics: A systematic review. Front. Endocrinol.
2021
,12, 645194.
[CrossRef] [PubMed]
47.
Shin, J.; Toyoda, S.; Nishitani, S.; Fukuhara, A.; Kita, S.; Otsuki, M.; Shimomura, I. Possible involvement of adipose tissue in
patients with older age, obesity, and diabetes with SARS-CoV-2 infection (COVID-19) via GRP78 (BIP/HSPA5): Significance of
hyperinsulinemia management in COVID-19. Diabetes 2021,70, 2745–2755. [CrossRef]
48.
Pérez-Torres, I.; Guarner-Lans, V.; Soria-Castro, E.; Manzano-Pech, L.; Palacios-Chavarría, A.; Valdez-Vázquez, R.R.; Domínguez-
Cherit, J.G.; Herrera-Bello, H.; Castillejos-Suastegui, H.; Moreno-Castañeda, L.; et al. Alteration in the lipid profile and the
desaturases activity in patients with severe pneumonia by SARS-CoV-2. Front. Physiol. 2021,12, 667024. [CrossRef]
49.
Montuschi, P.; Corradi, M.; Ciabattoni, G.; Nightingale, J.; Kharitonov, S.A.; Barnes, P.J. Increased 8-isoprostane, a marker of
oxidative stress, in exhaled condensate of asthma patients. Am. J. Respir. Crit. Care. Med. 1999,160, 216–220. [CrossRef]
Cells 2022,11, 932 22 of 23
50. Praticò, D.; Basili, S.; Vieri, M.; Cordova, C.; Violi, F.; Fitzgerald, G.A. Chronic obstructive pulmonary disease is associated with
an increase in urinary levels of isoprostane F
2α
-III, an index of oxidant stress. Am. J. Respir. Crit. Care. Med.
1998
,158, 1709–1714.
[CrossRef]
51.
Chuang, H.C.; Chen, Y.Y.; Hsiao, T.C.; Chou, H.C.; Kuo, H.P.; Feng, P.H.; Ho, S.C.; Chen, J.K.; Chuang, K.J.; Lee, K.Y. Alteration in
angiotensin-converting enzyme 2 by PM
1
during the development of emphysema in rats. ERJ. Open. Res.
2020
,6, 00174–02020.
[CrossRef] [PubMed]
52.
Al-Kuraishy, H.M.; Al-Gareeb, A.I.; Alblihed, M.; Guerreiro, S.G.; Cruz-Martins, N.; Batiha, G.E. COVID-19 in relation to
hyperglycemia and diabetes mellitus. Front. Cardiovasc. Med. 2021,8, 644095. [CrossRef] [PubMed]
53.
Yaghoubi, N.; Youssefi, M.; Azad, F.J.; Farzad, F.; Yavari, Z.; Avval, F.Z. Total antioxidant capacity as a marker of severity of
COVID-19 infection: Possible prognostic and therapeutic clinical application. J. Med.Virol. 2022,94, 1558–1565. [CrossRef]
54.
Kouhpayeh, S.; Shariati, L.; Boshtam, M.; Rahimmanesh, I.; Mirian, M.; Esmaeili, Y.; Najaflu, M.; Khanahmad, N.; Zeinalian, M.;
Trovato, M.; et al. The Molecular basis of COVID-19 pathogenesis, conventional and nanomedicine therapy. Int. J. Mol. Sci.
2021
,
22, 5438. [CrossRef] [PubMed]
55.
Prakash, A.; Kaur, S.; Kaur, C.; Prabha, P.K.; Bhatacharya, A.; Sarma, P.; Medhi, B. Efficacy and safety of inhaled nitric oxide in the
treatment of severe/critical COVID-19 patients: A systematic review. Indian. J. Pharmacol. 2021,53, 236–243. [PubMed]
56.
Wang, J.; Mei, F.; Bai, L.; Zhou, S.; Liu, D.; Yao, L.; Ahluwalia, A.; Ghiladi, R.A.; Su, L.; Shu, T.; et al. Serum nitrite and nitrate: A
potential biomarker for post-COVID-19 complications? Free. Radic. Biol. Med. 2021,175, 216–225. [CrossRef]
57.
Kalem, A.K.; Kayaaslan, B.; Neselioglu, S.; Eser, F.; Hasanoglu, ˙
I.; Aypak, A.; Akinci, E.; Akca, H.N.; Erel, O.; Guner, R.A. Useful
and sensitive marker in the prediction of COVID-19 and disease severity: Thiol. Free. Radic. Biol. Med.
2021
,166, 11–17. [CrossRef]
58.
Ibrahim, H.; Perl, A.; Smith, D.; Lewis, T.; Kon, Z.; Goldenberg, R.; Yarta, K.; Staniloae, C.; Williams, M. Therapeutic blockade of
inflammation in severe COVID-19 infection with intravenous N-acetylcysteine. Clin. Immunol. 2020,219, 108544. [CrossRef]
59.
Bourgonje, A.R.; Offringa, A.K.; van Eijk, L.E.; Abdulle, A.E.; Hillebrands, J.L.; van der Voort, P.H.J.; van Goor, H.; van Hezik, E.J.
N-Acetylcysteine and hydrogen sulfide in coronavirus disease 2019. Antioxid. Redox. Signal. 2021,35, 1207–1225. [CrossRef]
60.
Parimala, N.; Pandey, V.; Phanithi, P.B. Role of nitric oxide and hydrogen sulfide in ischemic stroke and the emergent epigenetic
underpinnings. Mol. Neurobiol. 2019,56, 1749–1769.
61.
Ming, Y.; Lai, C.L. SARS-CoV-2 infection: Can ferroptosis be a potential treatment target for multiple organ involvement? Cell
Death Discov. 2020,6, 130.
62.
Grishin, A.M.; Dolgova, N.V.; Landreth, S.; Fisette, O.; Pickering, I.J.; George, G.N.; Falzarano, D.; Cygler, M. Disulfide bonds play
a critical role in the structure and function of the receptor-binding domain of the SARS-CoV-2 Spike antigen. J. Mol. Biol.
2022
,
434, 167357. [CrossRef] [PubMed]
63.
Erel, O.; Ne¸selio˘glu, S.; Tunçay, M.E.; O˘guz, E.F.; Eren, F.; Akku¸s, M.S.; Güner, H.R.; Ate¸s, ˙
I. A sensitive indicator for the severity
of COVID-19: Thiol. Turk. J. Med. Sci. 2021,51, 921–928. [CrossRef] [PubMed]
64.
Mösbauer, K.; Fritsch, V.N.; Adrian, L.; Bernhardt, J.; Gruhlke, M.C.H.; Slusarenko, A.J.; Niemeyer, D.; Antelmann, H. The effect
of allicin on the proteome of SARS-CoV-2 infected Calu-3 Cells. Front. Microbiol. 2021,12, 746795. [CrossRef] [PubMed]
65.
Yang, G. H
2
S as a potential defense against COVID-19? Am. J. Physiol. Cell. Physiol.
2020
,319, C244–C249. [CrossRef] [PubMed]
66.
Renieris, G.; Katrini, K.; Damoulari, C.; Akinosoglou, K.; Psarrakis, C.; Kyriakopoulou, M.; Dimopoulos, G.; Lada, M.;
Koufargyris, P
.; Giamarellos-Bourboulis, E.J. Serum hydrogen sulfide and outcome association in pneumonia by the SARS-CoV-2
coronavirus. Shock 2020,54, 633–637. [CrossRef] [PubMed]
67.
Ao, T.; Kikuta, J.; Ishii, M. The effects of vitamin D on immune system and inflammatory diseases. Biomolecules
2021
,11, 1624.
[CrossRef]
68.
Ghelani, D.; Alesi, S.; Mousa, A. Vitamin D and COVID-19: An overview of recent evidence. Int. J. Mol. Sci.
2021
,22, 10559.
[CrossRef]
69.
Cooper, I.D.; Crofts, C.A.P.; DiNicolantonio, J.J.; Malhotra, A.; Elliott, B.; Kyriakidou, Y.; Brookler, K.H. Relationships between
hyperinsulinaemia, magnesium, vitamin D, thrombosis and COVID-19: Rationale for clinical management. Open Heart
2020
,
7, e001356. [CrossRef]
70.
Song, Y.; Qayyum, S.; Greer, R.A.; Slominski, R.M.; Raman, C.; Slominski, A.T.; Song, Y. Vitamin D3 and its hydroxyderivatives as
promising drugs against COVID-19: A computational study. J. Biomol. Struct. Dyn. 2021, 1–17. [CrossRef]
71.
Shakoor, H.; Feehan, J.; Al-Dhaheri, A.S.; Ali, H.I.; Platat, C.; Ismail, L.C.; Apostolopoulos, V.; Stojanovska, L. Immune-boosting
role of vitamins D, C, E, zinc, selenium and omega-3 fatty acids: Could they help against COVID-19? Maturitas
2021
,143, 1–9.
[CrossRef] [PubMed]
72.
Kotur, N.; Skakic, A.; Klaassen, K.; Gasic, V.; Zukic, B.; Skodric-Trifunovic, V.; Stjepanovic, M.; Zivkovic, Z.; Ostojic, O.;
Stevanovic, G.; et al
. Association of vitamin D, zinc and selenium related genetic variants with COVID-19 disease severity. Front.
Nutr. 2021,8, 689419. [CrossRef] [PubMed]
73.
Kieliszek, M. Selenium fascinating microelement, properties and sources in food. Molecules
2019
,24, 1298. [CrossRef] [PubMed]
74.
Fratta-Pasini, A.M.; Stranieri, C.; Girelli, D.; Busti, F.; Cominacini, L. Is Ferroptosis a key component of the process leading to
multiorgan damage in COVID-19? Antioxidants 2021,10, 1677. [CrossRef]
Cells 2022,11, 932 23 of 23
75.
Herzenberg, L.A.; De Rosa, S.C.; Dubs, J.G.; Roederer, M.; Anderson, M.T.; Ela, S.W.; Deresinski, S.C.; Herzenberg, L.A.
Glutathione deficiency is associated with impaired survival in HIV disease. Proc. Natl. Acad. Sci. USA
1997
,94, 1967–1972.
[CrossRef]
76.
Zuo, Y.; Warnock, M.; Harbaugh, A.; Yalavarthi, S.; Gockman, K.; Zuo, M.; Madison, J.A.; Knight, J.S.; Kanthi, Y.; Lawrence, D.A.
Plasma tissue plasminogen activator and plasminogen activator inhibitor-1 in hospitalized COVID-19 patients. Sci. Rep.
2021
,
11, 1580. [CrossRef]
... 11 It appears that exposure to SARS-CoV-2 precipitates the onset of type 1 diabetes. 12 In patients with COVID-19 and those recovering from COVID-19, fasting insulin, C-peptide levels, homeostasis model assessment of β cell dysfunction (HOMA-β) and homeostasis model assessment of insulin resistance (HOMA-IR) level were higher, 4,13 indicating multiple effects of SARS-CoV-2 infection on insulin-secreting organs as well as insulin-targeting organs. These phenomena suggested that new-onset diabetes may be different from type 1 diabetes. ...
... 19 Viral replication requires an enormous supply of ATP, which would result in cellular depletion of ATP. 13 Moreover, the untargeted metabolomic and lipidomic analysis demonstrated a disruption of the TCA cycle in non-severe COVID-19 patients, 33 suggesting the impaired ATP generation. Glucose-dependent insulin secretion of β cells requires ATP, and consequently, ATP deficiency can impair the GSIS function. ...
Article
Full-text available
Coronavirus disease 2019 (COVID-19)-induced new-onset diabetes has raised widespread concerns. Increased glucose concentration and insulin resistance levels were observed in the COVID-19 patients. COVID-19 patients with newly diagnosed diabetes may have worse clinical outcomes and can have serious consequences. The types and exact mechanisms of COVID-19-caused diabetes are not well understood. Understanding the direct effects of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) on pancreatic beta cells and insulin target metabolism organs, such as the liver, muscle, and adipose tissues, will provide new ideas for preventing and treating the new-onset diabetes induced by COVID-19.
... On the other hand, OS is characterized by an increase in ROS and the depletion of the enzymatic and non-enzymatic antioxidant systems. This leads to the disturbance of redox homeostasis [20]. Therefore, the objective of this study was to evaluate and demonstrate whether treatment with MT contributes to decreasing the OS present in COVID-19 patients and whether it may correct the alteration in redox homeostasis. ...
Article
Full-text available
Type II pneumocytes are the target of the SARS-CoV-2 virus, which alters their redox homeostasis to increase reactive oxygen species (ROS). Melatonin (MT) has antioxidant proprieties and protects mitochondrial function. In this study, we evaluated whether treatment with MT compensated for the redox homeostasis alteration in serum from COVID-19 patients. We determined oxidative stress (OS) markers such as carbonyls, glutathione (GSH), total antioxidant capacity (TAC), thiols, nitrites (NO 2 −), lipid peroxidation (LPO), and thiol groups in serum. We also studied the enzymatic activities of glutathione peroxidase (GPx), glutathione-S-transferase (GST), reductase (GR), thioredoxin reductase (TrxR), extracellular superoxide dismutase (ecSOD) and peroxidases. There were significant increases in LPO and carbonyl quantities (p ≤ 0.03) and decreases in TAC and the quantities of NO 2 − , thiols, and GSH (p < 0.001) in COVID-19 patients. The activities of the antioxidant enzymes such as ecSOD, TrxR, GPx, GST, GR, and peroxidases were decreased (p ≤ 0.04) after the MT treatment. The treatment with MT favored the activity of the antioxidant enzymes that contributed to an increase in TAC and restored the lost redox homeostasis. MT also modulated glucose homeostasis, functioning as a glycolytic agent, and inhibited the Warburg effect. Thus, MT restores the redox homeostasis that is altered in COVID-19 patients and can be used as adjuvant therapy in SARS-CoV-2 infection.
... It was found that in the case group (COVID-19-infected group), HOMA score was significantly higher compared to the control group (patients without . In a comparable study performed by Soto et al., which examined glycemic factors and IR in both COVID-19 patients and a control group, they noted that the HOMA score was elevated in the COVID group in contrast to the group of control [112]. ...
Article
Emerging publications indicate that diabetes predisposes patients with COVID-19 to more severe complications, which is partly attributed to inflammatory condition. In the current review, we reviewed recent published literature to provide evidence on the role of insulin resistance (IR) in diabetes, the association between diabetes and COVID-19 severity and mortality, the impact of COVID-19 infection on incident new-onset diabetes, mechanisms responsible for IR in COVID-19 patients, and the predictive value of different surrogates of IR in COVID-19. The literature search performs to find out studies that have assessed the association between IR surrogates and morbidity and mortality in patients with COVID-19. We showed that there is a bulk of evidence in support of the fact that diabetes is a potent risk factor for enhanced morbidity and mortality in COVID-19 patients. COVID-19 patients with diabetes are more prone to remarkable dysglycemia compared to those without diabetes, which is associated with an unfavourable prognosis. Furthermore, SARS-COV2 can make patients predispose to IR and diabetes via activating ISR, affecting RAAS signaling pathway, provoking inflammation, and changing the expression of PPARɣ and SREBP-1. Additionally, higher IR is associated with increased morbidity and mortality in COVID-19 patients and different surrogates of IR can be utilized as a prognostic biomarker for COVID-19 patients. Different surrogates of IR can be utilized as predictors of COVID-19 complications and death.
... Increased lipolysis and elevated circulating nonesterified fatty acids (NEFA) were also described [12]. Damaged mitochondria in peripheral tissues cannot meet the increased energetic demands caused by SARS-CoV-2-triggered inflammation, with a subsequent increase in reactive oxygen species (ROS) production [13]. Beta cells, on the other hand, can be affected by many suggested mechanisms: systemic and islet reninangiotensin-aldosterone system (RAAS) activation, islet redox stress, systemic and islet inflammation, islet amyloid deposition, islet fibrosis, and/or failure due to apoptosis and capillary rarefaction [14]. ...
Article
Full-text available
Abstract Background COVID-19, an infectious disease caused by SARS-CoV-2, was shown to be associated with an increased risk of new-onset diabetes. Mechanisms contributing to the development of hyperglycemia are still unclear. We aimed to study whether hyperglycemia is related to insulin resistance and/or beta cell dysfunction. Materials and methods Survivors of severe COVID-19 but without a known history of diabetes were examined at baseline (T0) and after 3 (T3) and 6 (T6) months: corticosteroids use, indirect calorimetry, and OGTT. Insulin response and sensitivity (IS) were expressed as insulinogenic (IGI), disposition (DI), and Matsuda insulin sensitivity index (ISI). Resting energy expenditure (REE) and respiratory quotient (RQ) was calculated from the gas exchange and nitrogen losses. Results 26 patients (out of 37) with complete outcome data were included in the analysis (age ~59.0 years; BMI ~ 30.4, 35% women). Patients were hypermetabolic at T0 (30.3 ± 4.0 kcal/kg lean mass/day, ~120% predicted) but REE declined over 6 months (ΔT6-T0 mean dif. T6-T0 (95% CI): −5.4 (−6.8, −4.1) kcal/kg FFM/day, p
... Previous studies, which include a common pathway affecting all of these danger elements, have tested a measure of low GSH degrees [51][52][53] . COVID-19 sufferers display changes inside the glucose-insulin axis, main to hyperglycemia and extensive decreases in vitamin D and thiols [54] . GSH depletion can result in immune machine failure and positioned the body's organs at hazard from oxidative pressure resulting from the buildup of ROS that could be a commonplace hallmark in all situations assocated with COVID -19 [34] . ...
Article
Full-text available
Background: The world witnessed a major crisis when the new Corona virus (Covid-19) appeared, and its effects are still continuing, as no gender, race, age, or color was excluded from infection, and given the disease represented by a pandemic that spread in most parts of the world in recent years and still poses a threat to Therefore, it is a fertile field for studies and to delve into it extensively. Methods: the study was conducted in Nineveh governorate on (113), samples of (34) for patients, Covid-19 with diabetes patients (29), and a healthy group (50), whose ages ranged between (35-70) years. They measured (15) parameters of some enzymes, levels of oxidants and its antioxidants, and indicators of kidney function and minerals. Results: The results showed that there was a high significant increase in all the measured enzymes lactoperoxidase (LP), lactate dehydrogenase (LDH), aspartate and alanine aminotansferases (AST, ALT), as well as an increase in the level of interleukin-6(IL-6) when compared between groups of patients COVID-19 with diabetes, and that there was a very high oxidative stress in patients with COVID-19 and diabetes when compared with COVID-19 patients and control group. It was also observed that there was a high significant increase in the levels of total cholesterol, urea, creatinine, and iron, and a high significant decrease in the level of calcium in patients with COVID-19 and diabetes. Conclusion: The study concluded that there are clear effects of the levels of measured enzymes (LP, LDH, ALT and AST) and the level of IL-6 in patients with COVID-19, especially those with diabetes, as well as a high oxidative stress state, and there is a clear effect on the kidneys and their functions in patients with (Covid-19), especially those with diabetes.
... In a study involving 61 adult patients admitted to the ICU of the CITIBANAMEX Center (Mexico City, Mexico) between 20 August and 20 September 2020, the selenium concentration was found decreased in severe COVID-19 patients (n = 27), compared to the levels found in 25 age-and gender-matched healthy subjects [62]. However, no significant decrease was found in moderate COVID-19 patients (n = 34). ...
Article
Full-text available
The purpose of this review is to systematically examine the scientific evidence investigating selenium’s relationship with COVID-19, aiming to support, or refute, the growing hypothesis that supplementation could prevent COVID-19 etiopathogenesis. In fact, immediately after the beginning of the COVID-19 pandemic, several speculative reviews suggested that selenium supplementation in the general population could act as a silver bullet to limit or even prevent the disease. Instead, a deep reading of the scientific reports on selenium and COVID-19 that are available to date supports neither the specific role of selenium in COVID-19 severity, nor the role of its supplementation in the prevention disease onset, nor its etiology.
Article
Full-text available
Mitochondria are critical for providing energy to maintain cell viability. Oxidative phos-phorylation involves the transfer of electrons from energy substrates to oxygen to produce adenosine triphosphate. Mitochondria also regulate cell proliferation, metastasis, and deterioration. The flow of electrons in the mitochondrial respiratory chain generates reactive oxygen species (ROS), which are harmful to cells at high levels. Oxidative stress caused by ROS accumulation has been associated with an increased risk of cancer, and cardiovascular and liver diseases. Glutathione (GSH) is an abundant cellular antioxidant that is primarily synthesized in the cytoplasm and delivered to the mitochondria. Mitochondrial glutathione (mGSH) metabolizes hydrogen peroxide within the mitochondria. A long-term imbalance in the ratio of mitochondrial ROS to mGSH can cause cell dysfunction, apoptosis, necroptosis, and ferroptosis, which may lead to disease. This study aimed to review the physiological functions, anabolism, variations in organ tissue accumulation, and delivery of GSH to the mitochondria and the relationships between mGSH levels, the GSH/GSH disulfide (GSSG) ratio, programmed cell death, and ferroptosis. We also discuss diseases caused by mGSH deficiency and related therapeutics.
Article
Full-text available
Cellular homeostasis is lost or becomes dysfunctional during septic shock due to the activation of the inflammatory response and the deregulation of oxidative stress. Antioxidant therapy administered alongside standard treatment could restore this lost homeostasis. We included 131 patients with septic shock who were treated with standard treatment and vitamin C (Vit C), vitamin E (Vit E), N-acetylcysteine (NAC), or melatonin (MT), in a randomized trial. Organ damage quantified by Sequential Organ Failure Assessment (SOFA) score, and we determined levels of Interleukins (IL) IL1β, Tumor necrosis factor alpha (TNFα), IL-6, monocyte chemoattractant protein-1 (MCP-1), Transforming growth factor B (TGFβ), IL-4, IL-10, IL-12, and Interferon-γ (IFNγ). The SOFA score decreased in patients treated with Vit C, NAC, and MT. Patients treated with MT had statistically significantly reduced of IL-6, IL-8, MCP-1, and IL-10 levels. Lipid peroxidation, Nitrates and nitrites (NO3− and NO2−), glutathione reductase, and superoxide dismutase decreased after treatment with Vit C, Vit E, NAC, and MT. The levels of thiols recovered with the use of Vit E, and all patients treated with antioxidants maintained their selenium levels, in contrast with controls (p = 0.04). The findings regarding oxidative stress markers and cytokines after treatment with antioxidants allow us to consider to future the combined use of antioxidants in a randomized clinical trial with a larger sample to demonstrate the reproducibility of these beneficial effects.
Article
Full-text available
Obesity, diabetes mellitus (mostly type 2), and COVID-19 show mutual interactions because they are not only risk factors for both acute and chronic COVID-19 manifestations, but also because COVID-19 alters energy metabolism. Such metabolic alterations can lead to dysglycemia and long-lasting effects. Thus, the COVID-19 pandemic has the potential for a further rise of the diabetes pandemic. This review outlines how preexisting metabolic alterations spanning from excess visceral adipose tissue to hyperglycemia and overt diabetes may exacerbate COVID-19 severity. We also summarize the different effects of SARS-CoV-2 infection on the key organs and tissues orchestrating energy metabolism, including adipose tissue, liver, skeletal muscle, and pancreas. Last, we provide an integrative view of the metabolic derangements that occur during COVID-19. Altogether, this review allows for better understanding of the metabolic derangements occurring when a fire starts from a small flame, and thereby help reducing the impact of the COVID-19 pandemic.
Article
Full-text available
Severe acute respiratory syndrome (SARS)-CoV-2 virus causes novel coronavirus disease 2019 (COVID-19) with other comorbidities such as diabetes. Diabetes is the most common cause of diabetic nephropathy, which is attributed to hyperglycemia. COVID-19 produces severe complications in people with diabetes mellitus. This article explains how SARS-CoV-2 causes more significant kidney damage in diabetic patients. Importantly, COVID-19 and diabetes share inflammatory pathways of disease progression. SARS-CoV-2 binding with ACE-2 causes depletion of ACE-2 (angiotensin-converting enzyme 2) from blood vessels, and subsequently, angiotensin-II interacts with angiotensin receptor-1 from vascular membranes that produce NADPH (nicotinamide adenine dinucleotide hydrogen phosphate) oxidase, oxidative stress, and constriction of blood vessels. Since diabetes and COVID-19 can create oxidative stress, we hypothesize that COVID-19 with comorbidities such as diabetes can synergistically increase oxidative stress leading to end-stage renal failure and death. Antioxidants may therefore prevent renal damage-induced death by inhibiting oxidative damage and thus can help protect people from COVID-19 related comorbidities. A few clinical trials indicated how effective the antioxidant therapy is against improving COVID-19 symptoms, based on a limited number of patients who experienced COVID-19. In this review, we tried to understand how effective antioxidants (such as vitamin D and flavonoids) can act as food supplements or therapeutics against COVID-19 with diabetes as comorbidity based on recently available clinical, preclinical, or in silico studies.
Article
Full-text available
Infection by the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) leads to multi-organ failure associated with a cytokine storm and septic shock. The virus evades the mitochondrial production of interferons through its N protein and, from that moment on, it hijacks the functions of these organelles. The aim of this study was to show how the virus kidnaps the mitochondrial machinery for its benefit and survival, leading to alterations of serum parameters and to nitrosative stress (NSS). In a prospective cohort of 15 postmortem patients who died from COVID-19, six markers of mitochondrial function (COX II, COX IV, MnSOD, nitrotyrosine, Bcl-2 and caspase-9) were analyzed by the immune colloidal gold technique in samples from the lung, heart, and liver. Biometric laboratory results from these patients showed alterations in hemoglobin, platelets, creatinine, urea nitrogen, glucose, C-reactive protein, albumin, D-dimer, ferritin, fibrinogen, Ca²⁺, K⁺, lactate and troponin. These changes were associated with alterations in the mitochondrial structure and function. The multi-organ dysfunction present in COVID-19 patients may be caused, in part, by damage to the mitochondria that results in an inflammatory state that contributes to NSS, which activates the sepsis cascade and results in increased mortality in COVID-19 patients.
Article
Full-text available
Having diagnostic methods that meet certain attributes is vital to guide health decisions, the current context warrants it. In order to validate the capacity of two rapid tests to detect antibodies due to SARS-CoV-2 infection in Isla de la Juventud, Cuba, from April to May 2020, a descriptive cross-sectional evaluation study was carried out. Rapid tests: Wondfo (SARS-CoV-2 antibody test) and Lungene covid-19 IgG/IgM, were compared to the real-time polymerase chain reaction test. Two homogeneous samples of 250 each were constructed, determining validation indicators. Sensitivity values of 6.6% and 8.3% respectively were obtained for each test, while the specificity was higher for Wondfo (95%). The positive predictive values were very low, the negative ones were adequate, higher in Lungene with 94.8%. Likelihood ratio values were classified as useless. In different scenarios in terms of symptomatic cases, sensitivity of 50% was reached in an interval of 1 to 7 days for Wondfo. The area under the ROC curve for Wondfo was 0.50 (95% CI = 0.46-0.55) and 0.46 for Lungene (95%CI = 0.38-0.55). The kappa index for Wondfo was 0.025 and 0.010 for Lungene. The rapid tests explored showed very low sensitivity, positive predictive value, and inadequate likelihood ratio. The global validity of the tests did not demonstrate a good diagnostic performance, marked by the value of the area under the ROC curve. The degree of agreement was poor.
Article
Full-text available
A hallmark of COVID-19 is a hyperinflammatory state associated with severity. Monocytes undergo metabolic reprogramming and produce inflammatory cytokines when stimulated with SARS-CoV-2. We hypothesized that binding by the viral spike protein mediates this effect, and that drugs which regulate immunometabolism could inhibit the inflammatory response. Monocytes stimulated with recombinant SARS-CoV-2 spike protein subunit 1 showed a dose-dependent increase in glycolytic metabolism associated with production of pro-inflammatory cytokines. This response was dependent on hypoxia-inducible factor-1α, as chetomin inhibited glycolysis and cytokine production. Inhibition of glycolytic metabolism by 2-deoxyglucose (2-DG) or glucose deprivation also inhibited the glycolytic response, and 2-DG strongly suppressed cytokine production. Glucose-deprived monocytes rescued cytokine production by upregulating oxidative phosphorylation, an effect which was not present in 2-DG-treated monocytes due to the known effect of 2-DG on suppressing mitochondrial metabolism. Finally, pre-treatment of monocytes with metformin strongly suppressed spike protein-mediated cytokine production and metabolic reprogramming. Likewise, metformin pre-treatment blocked cytokine induction by SARS-CoV-2 strain WA1/2020 in direct infection experiments. In summary, the SARS-CoV-2 spike protein induces a pro-inflammatory immunometabolic response in monocytes that can be suppressed by metformin, and metformin likewise suppresses inflammatory responses to live SARS-CoV-2. This has potential implications for the treatment of hyperinflammation during COVID-19.
Article
Full-text available
Immune cells, including dendritic cells, macrophages, and T and B cells, express the vitamin D receptor and 1α-hydroxylase. In vitro studies have shown that 1,25-dihydroxyvitamin D, the active form of vitamin D, has an anti-inflammatory effect. Recent epidemiological evidence has indicated a significant association between vitamin D deficiency and an increased incidence, or aggravation, of infectious diseases and inflammatory autoimmune diseases, such as rheumatoid arthritis, systemic lupus erythematosus, and multiple sclerosis. However, the impact of vitamin D on treatment and prevention, particularly in infectious diseases such as the 2019 coronavirus disease (COVID-19), remains controversial. Here, we review recent evidence associated with the relationship between vitamin D and inflammatory diseases and describe the underlying immunomodulatory effect of vitamin D.
Article
Full-text available
Allicin (diallyl thiosulfinate) is the major thiol-reactive organosulfur compound produced by garlic plants ( Allium sativum ) upon tissue damage. Allicin exerts its strong antimicrobial activity against bacteria and fungi via S -thioallylation of protein thiols and low molecular weight thiols. Here, we investigated the effect of allicin on SARS-CoV-2 infected Vero E6 and Calu-3 cells. Toxicity tests revealed that Calu-3 cells showed greater allicin tolerance, probably due to >4-fold higher GSH levels compared to the very sensitive Vero E6 cells. Exposure of infected Vero E6 and Calu-3 cells to biocompatible allicin doses led to a ∼60–70% decrease of viral RNA and infectious viral particles. Label-free quantitative proteomics was used to investigate the changes in the Calu-3 proteome after SARS-CoV-2 infection and the effect of allicin on the host-virus proteome. SARS-CoV-2 infection of Calu-3 cells caused a strong induction of the antiviral interferon-stimulated gene (ISG) signature, including several antiviral effectors, such as cGAS, Mx1, IFIT, IFIH, IFI16, IFI44, OAS, and ISG15, pathways of vesicular transport, tight junctions (KIF5A/B/C, OSBPL2, CLTCL1, and ARHGAP17) and ubiquitin modification (UBE2L3/5), as well as reprogramming of host metabolism, transcription and translation. Allicin treatment of infected Calu-3 cells reduced the expression of IFN signaling pathways and ISG effectors and reverted several host pathways to levels of uninfected cells. Allicin further reduced the abundance of the structural viral proteins N, M, S and ORF3 in the host-virus proteome. In conclusion, our data demonstrate the antiviral and immunomodulatory activity of biocompatible doses of allicin in SARS-CoV-2-infected cell cultures. Future drug research should be directed to exploit the thiol-reactivity of allicin derivatives with increased stability and lower human cell toxicity as antiviral lead compounds.
Article
Full-text available
Even though COVID-19 is mostly well-known for affecting respiratory pathology, it can also result in several extrapulmonary manifestations, leading to multiorgan damage. A recent reported case of SARS-CoV-2 myocarditis with cardiogenic shock showed a signature of myocardial and kidney ferroptosis, a novel, iron-dependent programmed cell death. The term ferroptosis was coined in the last decade to describe the form of cell death induced by the small molecule erastin. As a specific inducer of ferroptosis, erastin inhibits cystine-glutamate antiporter system Xc-, blocking transportation into the cytoplasm of cystine, a precursor of glutathione (GSH) in exchange with glutamate and the consequent malfunction of GPX4. Ferroptosis is also promoted by intracellular iron overload and by the iron-dependent accumulation of polyunsaturated fatty acids (PUFA)-derived lipid peroxides. Since depletion of GSH, inactivation of GPX4, altered iron metabolism, and upregulation of PUFA peroxidation by reactive oxygen species are peculiar signs of COVID-19, there is the possibility that SARS-CoV-2 may trigger ferroptosis in the cells of multiple organs, thus contributing to multiorgan damage. Here, we review the molecular mechanisms of ferroptosis and its possible relationship with SARS-CoV-2 infection and multiorgan damage. Finally, we analyze the potential interventions that may combat ferroptosis and, therefore, reduce multiorgan damage.
Article
Background The pathogenesis of SARS-CoV-2 infection, causative pathogen of the known COVID-19 pandemic is not well clarified. In this regard oxidative stress is one of the topics that need to be investigated. Therefore, the present research was performed to explore the relationship between the oxidant/antioxidant system and COVID-19 exacerbation. Methods Sera were collected from 120 patients with COVID-19 infection and 60 healthy volunteers as the control group. The patient group consisted of 60 cases with mild disease and 60 severely ill patients. Serum levels of total antioxidant capacity (TAC) and nitric oxide (NO) as well as serum activities of the two main anti-oxidant defense enzymes, superoxide dismutase (SOD) and catalase (CAT), were measured. Results TAC levels were considerably lower in patients compared with healthy individuals (P<0.05) and also between patients with mild and severe diseases (P<0·05). A rather decreasing trend was also found in NO concentration as well as SOD and CAT activity, though, the observed differences were not statistically significant (P>0·05). Conclusion These findings suggest that COVID-19 patients may be susceptible to depleted total anti-oxidant capacity. Moreover, showing such variations in blood samples of infected individuals could be considered as a predictive marker of COVID-19 severity. This article is protected by copyright. All rights reserved.
Article
The current coronavirus pandemic is exerting a tremendously detrimental impact on global health. The Spike proteins of coronaviruses, responsible for cell receptor binding and viral internalization, possess multiple and frequently conserved disulfide bonds raising the question about their role in these proteins. Here, we present a detailed structural and functional investigation of the disulfide bonds of the SARS-CoV-2 Spike receptor-binding domain (RBD). Molecular dynamics simulations of the RBD predict increased flexibility of the surface loops when the four disulfide bonds of the domain are reduced. This flexibility is particularly prominent for the disulfide bond-containing surface loop (residues 456-490) that participates in the formation of the interaction surface with the Spike cell receptor ACE2. In vitro, disulfide bond reducing agents affect the RBD secondary structure, lower its melting temperature from 52°C to 36-39°C and decrease its binding affinity to ACE2 by two orders of magnitude at 37°C. Consistent with these in vitro findings, the reducing agents tris(2-carboxyethyl)phosphine (TCEP) and dithiothreitol (DTT) were able to inhibit viral replication at low millimolar levels in cell-based assays. Our research demonstrates the mechanism by which the disulfide bonds contribute to the molecular structure of the RBD of the Spike protein, allowing the RBD to execute its viral function.
Article
Aging, obesity and diabetes are major risk factors for the severe progression and outcome of SARS-CoV-2 infection (COVID-19), but the underlying mechanism is not yet fully understood. In this study, we found that the SARS-CoV-2 spike protein physically interacts with cell surface GRP78, which promotes the binding to and accumulation in ACE2-expressing cells. GRP78 was highly expressed in adipose tissue and increased in older and obese diabetic human and mouse subjects. The overexpression of GRP78 was attributed to hyperinsulinemia in adipocytes, which was in part mediated by the stress-responsive transcription factor XBP-1s. Management of hyperinsulinemia by pharmacological approaches, including metformin, SGLT2 inhibitor or β3-adrenergic receptor agonist, decreased GRP78 gene expression in adipose tissue. Environmental interventions, including exercise, calorie restriction, fasting or cold exposure, reduced the gene expression of GRP78 in adipose tissue. This study provides scientific evidence for the role of GRP78 as a binding partner of the SARS-CoV-2 spike protein and ACE2, which might be related to the severe progression and outcome of COVID-19 in older and obese diabetic patients. The management of hyperinsulinemia and the related GRP78 expression could be a potential therapeutic or preventative target.