ArticlePDF Available

Th17 and Treg cells function in SARS-CoV2 patients compared with healthy controls

Authors:

Abstract

In the course of the coronavirus disease 2019 (COVID-19), raising and reducing the function of Th17 and Treg cells, respectively, elicit hyperinflammation and disease progression. The current study aimed to evaluate the responses of Th17 and Treg cells in COVID-19 patients compared with the control group. Forty COVID-19 intensive care unit (ICU) patients were compared with 40 healthy controls. The frequency of cells, gene expression of related factors, as well as the secretion levels of cytokines, were measured by flow cytometry, real-time poly-merase chain reaction, and enzyme-linked immunosorbent assay techniques, respectively. The findings revealed a significant increase in the number of Th17 cells, the expression levels of related factors (RAR-related orphan receptor gamma [RORγt], IL-17, and IL-23), and the secretion levels of IL-17 and IL-23 cytokines in COVID-19 patients compared with controls. In contrast, patients had a remarkable reduction in the frequency of Treg cells, the expression levels of correlated factors (Forkhead box protein P3 [FoxP3], transforming growth factor-β [TGF-β], and IL-10), and cytokine secretion levels (TGF-β and IL-10). The ratio of Th17/Treg cells, RORγt/FoxP3, and IL-17/IL-10 had a considerable enhancement in patients compared with the controls and also in dead patients compared with the improved cases. The findings showed that enhanced responses of Th17 cells and decreased responses of Treg cells in 2019-n-CoV patients compared with controls had a strong relationship with hyperinflammation, lung damage, and disease pathogenesis. Also, the high ratio of Th17/Treg cells and their associated factors in COVID-19-dead patients compared with improved cases indicates the critical role of inflammation in the mortality of patients. K E Y W O R D S COVID-19, Th17, Th17/Treg cells ratio, Treg
Received: 14 July 2020
|
Revised: 12 August 2020
|
Accepted: 31 August 2020
DOI: 10.1002/jcp.30047
ORIGINAL RESEARCH ARTICLE
Th17 and Treg cells function in SARSCoV2 patients
compared with healthy controls
Armin Sadeghi
1,2
|Safa Tahmasebi
3
|Arshad Mahmood
4
|Maria Kuznetsova
5
|
Hamed Valizadeh
1,2
|Ali Taghizadieh
1,2
|Masoud Nazemiyeh
1,2
|
Leili AghebatiMaleki
6
|Farhad JadidiNiaragh
7
|Sanaz AbbaspourAghdam
8
|
Leila Roshangar
8
|Haleh Mikaeili
1,2
|Majid Ahmadi
8
1
Tuberculosis and Lung Disease Research
Center of Tabriz University of Medical
Sciences, Tabriz, Iran
2
Department of Internal Medicine, School of
Medicine, Tabriz University of Medical
Sciences, Tabriz, Iran
3
Department of Immunology, Healthy Faculty,
Tehran University of Medical Sciences,
Tehran, Iran
4
School of Management, Universiti Sains
Malaysia, Penang, Malaysia
5
Department of Propaedeutics of Dental
Diseases, I.M. Sechenov First Moscow State
Medical University (Sechenov University),
Moscow, Russia
6
Immunology Research Center, Tabriz
University of Medical Sciences, Tabriz, Iran
7
Department of Immunology, Tabriz
University of Medical Sciences, Tabriz, Iran
8
Stem Cell Research Center, Tabriz University
of Medical Sciences, Tabriz, Iran
Correspondence
Haleh Mikaeili, Department of Internal
Medicine, School of Medicine, Tabriz
University of Medical Sciences, Tabriz, Iran.
Email: mikaiili@hotmail.com
Majid Ahmadi, Stem Cell Research Center,
Tabriz University of Medical Sciences,
Tabriz, Iran.
Email: Ahmadi.m@tbzmed.ac.ir
Funding information
Tabriz University of Medical Sciences,
Grant/Award Number: 65235
Abstract
In the course of the coronavirus disease 2019 (COVID19), raising and reducing
the function of Th17 and Treg cells, respectively, elicit hyperinflammation and
disease progression. The current study aimed to evaluate the responses of Th17
and Treg cells in COVID19 patients compared with the control group. Forty
COVID19 intensive care unit (ICU) patients were compared with 40 healthy
controls. The frequency of cells, gene expression of related factors, as well as the
secretion levels of cytokines, were measured by flow cytometry, realtime poly-
merase chain reaction, and enzymelinked immunosorbent assay techniques, re-
spectively. The findings revealed a significant increase in the number of Th17 cells,
the expression levels of related factors (RARrelated orphan receptor gamma
[RORγt], IL17, and IL23), and the secretion levels of IL17 and IL23 cytokines in
COVID19 patients compared with controls. In contrast, patients had a remarkable
reduction in the frequency of Treg cells, the expression levels of correlated factors
(Forkhead box protein P3 [FoxP3], transforming growth factorβ[TGFβ], and
IL10), and cytokine secretion levels (TGFβand IL10). The ratio of Th17/Treg
cells, RORγt/FoxP3, and IL17/IL10 had a considerable enhancement in patients
compared with the controls and also in dead patients compared with the improved
cases. The findings showed that enhanced responses of Th17 cells and decreased
responses of Treg cells in 2019nCoV patients compared with controls
had a strong relationship with hyperinflammation, lung damage, and disease
pathogenesis. Also, the high ratio of Th17/Treg cells and their associated factors in
COVID19dead patients compared with improved cases indicates the critical role
of inflammation in the mortality of patients.
KEYWORDS
COVID19, Th17, Th17/Treg cells ratio, Treg
J Cell Physiol. 2020;111. wileyonlinelibrary.com/journal/jcp © 2020 Wiley Periodicals LLC
|
1
1|INTRODUCTION
In late 2019, the novel severe acute respiratory syndrome cor-
onavirus 2 (SARSCoV2) originated from Wuhan, China, and was
introduced to the world by the World Health Organization (WHO),
namely coronavirus disease 2019 (COVID19; F. Wu, Zhao, et al.,
2020;Y.C. Wu, Chen, & Chan, 2020). The SARSCoV2, as the third
zoonotic coronavirus, belongs to the βcoronavirus cluster,
which includes the Middle East respiratory syndrome coronavirus
(MERSCoV) and SARSCoV (Zhou et al., 2020). Due to the rapid
spread and humantohuman transmission of the novel coronavirus
(2019nCoV), it has become a serious crisis around the world. Hence,
to manage this pandemic, the SARSCoV2 immunopathogenesis
needs to be better understood (Ghaebi, Osali, Valizadeh, Roshangar,
& Ahmadi, 2020)
By recognizing the immunological mechanisms involved in the
disease, appropriate diagnostic and therapeutic methods can be
employed to diagnose and treat COVID19 infected patients, effi-
ciently (Tahmasebi, Khosh, & Esmaeilzadeh, 2020). Transmission of
SARSCoV2 generally occurs through the respiratory system dro-
plets, and close personal contact. There has been a high risk of
contracting the SARSCoV2 in middleaged and older adults, as well
as individuals with underlying diseases (Riou & Althaus, 2020; Zhou
et al., 2020). In brief, from a clinical point of view, 2019nCoV elicits
a series of pathogenesis from coldlike infection to lethal sickness,
severe respiratory infections like acute respiratory disease syndrome
(ARDS), and damage to various organs, including the nervous system,
liver, kidneys, and gastrointestinal system (Lu et al., 2020; Yin &
Wunderink, 2018). The clinical symptoms that characterize the
COVID19, mainly include fever, dry cough, respiratory problems,
and pneumonia, which are caused by inflammation and alveolar da-
mage (Q. Li, Guan, et al., 2020).
Immunologically, a decreased frequency of lymphocytes, espe-
cially T CD4
+
and CD8
+
cells brings lymphopenia, imbalance of the
immune system, and hyperinflammation due to the increased levels
of inflammatory cytokines. They are important paraclinical symp-
toms, identified as the main causes of respiratory damage and death
in novel coronavirus disease (Wan et al., 2020). It should be noted
that among the immune cells, natural killer (NK) cells, macrophages,
and neutrophils in innate immune responses, as well as T cells (CD4
+
and CD8
+
) and B cells in adaptive immune responses, play principal
roles in defending against the SARSCoV2. However, the con-
tamination of immune cells by virus and their alterations in the
course of the disease disturb the balance of the immune system,
which results in severe inflammation and disease progression (Lin,
Lu, Cao, & Li, 2020; Shi et al., 2020).
In intracellular infections, such as viral infections, the immune
system shifts the T cell responses toward the Th1 and Th17 pheno-
types, which in turn counteract the infection by producing the in-
flammatory factors. In infections, Th17 cells are differentiated by
RARrelated orphan receptor γt(RORγt) transcription factor and
interleukin23 (IL23), which play an antiviral role by inducing the
production of inflammatory cytokines, including IL17, TNFα, and
IL6 (Annunziato et al., 2007; Harrington et al., 2005). In contrast to
the mentioned inflammatory cells, regulatory T (Treg) cells are known
as immune systemregulating cells that have antiinflammatory effects
and involve in maintaining the immune system balance by producing
antiinflammatory cytokines, such as IL10, IL35, and TGFβ.The
FoxP3 transcription factor has a role in development and differ-
entiation of Tregs (Ahmadi et al., 2017; Braitch et al., 2009).
Accordingly, in 2019nCoV cases, antiviral activity and in-
flammatory responses are the responsibility of neutrophils, macro-
phages, Th1, and Th17 cells. It is noteworthy that these cells,
especially T cells, provoke a cytokine storm by producing large
amounts of inflammatory cytokines, including IL1β,IL2, 8, and 12,
TNFα, IFNγ,GCSF, GMCSF, (monocyte chemotactic protein1
[MCP1], macrophage inflammatory protein1A [MIP1A], and IP10
(Wan et al., 2020). In turn, cytokine storm leads to hyperinflamma-
tion, disease progression, and respiratory system injury (Jose
& Manuel, 2020). On the other hand, it has been reported that the
lack of Treg cells and Th2 cells, as well as the imbalance of Th17/Treg
cell ratio, alter the immune responses toward inflammatory pheno-
type and disease progression in COVID19 infected patients (Li,
Geng, Peng, Meng, & Lu, 2020c; Prompetchara, Ketloy, & Palaga,
2020). The current study aimed to investigate the inflammatory
condition in COVID19 patients admitted to the intensive care unit
(ICU) in comparison with healthy controls by measuring the number
of Th17 and Treg cells, their relevant transcription factors (RORɣt
and FoxP3, respectively), Th17 cell cytokines (IL17 and IL23), and T
reg cell cytokines (IL10 and TGFβ).
2|MATERIALS AND METHODS
2.1 |Study design and patients
In the present study, 40 COVID19 patients admitted into the ICU of
Imam Reza Hospital of Tabriz University of Medical Sciences (TUMS),
who were confirmed to be positive for SARSCoV2 by realtime
polymerase chain reaction (RTPCR), were evaluated in comparison
with 40 healthy controls without underlying diseases. The patient
and control groups were randomly selected, and both were between
20 and 80 years old. Of note, written informed consent was obtained
from all included patients and healthy controls. This study was ap-
proved by the Research Ethics Committee of Tabriz University of
Medical Sciences (IR. TBZMED. REC.1399.011).
According to the inclusion criteria of the current study, 2019
nCoV patients who were confirmed in terms of clinical criteria,
etiological characteristics, positive RTPCR testing and were willing
to cooperate in the study were included. In contrast, based on the
exclusion criteria of the current study, COVID19 patients with a
historyofchronicdiseasessuchasinfectiousdiseases(chronic
hepatitis B or C, chronic brucellosis, and HIV), cancers (leukemia
and lymphoma), and allergic disorders were excluded. The demo-
graphic information and clinical features of the patients are given in
Table 1.
2
|
SADEGHI ET AL.
2.2 |Blood sampling, PBMCs isolation, and cell
culture
From each patient and healthy individual, 8 ml of whole blood was
collected by heparinized syringes for cellular and molecular tests.
The peripheral blood mononuclear cells (PBMCs) were separated by
the densitygradient method using the standard Ficoll (lymphosep)
1.077 g/ml (Biosera) and then centrifuged (25 min, 450 g), which was
followed by twice washing with phosphate buffer saline (PBS; Sigma
Aldrich). Thereafter, separated PBMCs were cultured in a culture
medium consisting of 10% heatinactivated fetal bovine serum,
100 U/ml penicillin, 10 ng/ml of PMA, and 200 mM Lglutamine
(eBioscience). PBMCs in the culture medium were incubated for
48 hr at 37°C and 5% CO
2
. Finally, the cultured cells were subjected
to cell count by flow cytometry and gene expression by RTPCR.
Also, supernatants of isolated cells were employed to measure the
secretion levels of cytokines by enzymelinked immunosorbent assay
(ELISA).
2.3 |Cell separation and flow cytometry
The frequency of Th17 and Treg cells of ICUadmitted COVID19
patients and healthy controls were assessed by the flow cytometry
technique. To detect the Th17 and Treg cells, CD4
+
T cells were
separated from cultured PBMCs by magneticactivated cell sorting
(MACS; Miltenyi Biotec). For this purpose, to robust the intracellular
staining of IL17A, the PBMCs were triggered by 25 ng/ml of PMA
along with 1 μg/ml of ionomycin and then, incubated for 4 hr in the
presence of 1.7 μg/ml monensin (all from eBioscience). To detect
the Th17 cells, incubation of PBMCs with allophycocyanin
(APC)conjugated antiCD4 was conducted at 4°C for 15 min, and
subsequent staining was applied by phycoerythrin (PE)conjugated
antiIL17A (eBioscience). Moreover, the detection of Tregs was
performed by the incubation of cultured PBMCs with the fluorescein
isothiocyanate (FITC)labeled antihuman CD4, PElabeled anti
human CD25, and PerCPCy5.5conjugated antihuman CD127
(eBioscience) for 15 min at 4°C. In the current study, PE and FITC
mouse IgG1, κisotype control were considered as isotype controls.
Afterward, cell count analysis was done by the FACSCalibur flow
cytometer with FlowJo software (Becton Dickinson).
2.4 |RNA extraction and cDNA synthesis
Realtime PCR was conducted to quantify the expression profiles of
factors related to Th17 cells (RORɣt, IL17, and IL23) and Treg cells
(FoxP3, IL10, and TGFβ) in cultured PBMCs of COVID19 and
control groups using the SYBR Green approach, along with the re-
levant forward and reverse primers. Accordingly, total RNA
was extracted from isolated cells exploiting RNXPLUS Solution
(SinaClon), which is followed by complementary DNA (cDNA)
synthesis utilizing the random hexamer primer and Revert Aidre-
verse transcriptase (Thermo Fisher Scientific). Thereafter, the real
time PCR was performed to assay the gene expression as follow
steps: First of all, DNA was denaturated in the denaturation phase at
95°C for 10 s, which was repeated for 40 cycles. Then, the cycles
were continued to the annealing phase at 60°C for 30 s, and exten-
sion phase at 72°C for 20 s. To plot the standard curves, the six
TABLE 1 Subjects' clinical and laboratory information
COVID19
patients
(n= 40)
Healthy
control
group (n= 40) pvalue
Age (years) 2173
(54.2 ± 9.1)
2171
(52.4 ± 8.5)
Sex NS
Men 28 (70%) 28 (80%)
Women 12 (30%) 12 (20%)
Current smoking 10 (25%) 11 (27.5%) NS
Fever 40 (100%) 0 _
<37.3°C 3 (7.5%)
37.338.0°C 21 (52.5%)
38.139.0°C 11 (27.5%)
>39.0°C 4 (10%)
Cough 23 (57.5%) 0 _
Headache 7 (17.5%) 0 _
Dyspnea 12 (30%) 0 _
White blood cell count,
×10
9
/L
.0001
<4 11 (27.5%) 4 (10%)
410 20 (50%) 31 (77.5%)
>10 9 (22.5%) 5 (12.5%)
Lymphocyte count, ×10
9
/L .0001
<1.0 32 (80%) 18 (45%)
1.0 8 (20%) 22 (55%)
Platelet count, ×10
9
/L .0001
<100 25 (62.5%) 9 (22.5%)
100 15 (37.5%) 31 (77.5%)
Creatinine, μmol/L .0001
133 37 (92.5%) 2 (5%)
>133 3 (7.5%) 39 (95%)
Lactate
dehydrogenase,
U/L
.0001
245 23 (57.5%) 4 (10%)
>245 17 (42.5%) 38 (90%)
Bilateral involvement
of chest
radiographs
19 (95%) 0 _
Abbreviations: FoxP3, forkhead box P3; IL, interleukin; RORɣt, retinoic
acidrelated orphan receptor ɣ; TGFβ, transforming growth factor β.
SADEGHI ET AL.
|
3
standards were provided from 10fold serial dilutions of genes con-
centrated samples. Moreover, to confirm the amplification, 2%
agarose gel and Biosystems (Seqlab)were employed for Electro-
phoresis analysis and DNA sequencing. The βactin molecule was
considered as a housekeeping gene to compare the gene expression
of target genes. Eventually, the comparative C
t
method using the
2
CΔΔ
t
formula was utilized to analyze the data and compare the
relative expression of target genes with βactin. The sequences of
primers have been listed in Table 2.
2.5 |Detection of cytokine profile by ELISA
Th17 cellsecreted cytokines (IL17 and IL23), as inflammatory cy-
tokines, and Treg cellsecreted cytokines (IL10 and TGFβ), as anti
inflammatory cytokines, were measured by an ELISA kit (MyBio-
Source) using the supernatant of cultured PBMCs. In short, the ELISA
process was done as follow steps: coating the ELISA plate by adding
100 μl of coating antibody with overnight incubation, washing by PBS
containing 0.05% Tween20, incubating with a blocking buffer on a
shaker for an hour, pouring 100 μl samples and standards, incubating
for an hour, washing, adding 100 μl of a biotinylated antibody, in-
cubating for an hour, pouring 100 μl of the avidinbiotinperoxidase
complex, incubating for 30 min, adding 100 μl of tetra-
methylbenzidine (TMB) substrate, incubating for 30 min, stopping
the reaction and reading. The absorbance values were read by a
Medgenix ELISA reader (BP800; Biohit) at 450 nm.
2.6 |Statistical analysis
SPSS PC Statistics Software (version 19.0; SPSS Inc.) was used out
for statistical analysis. Accordingly, the unpaired ttest and the
MannWhitney Utest compared the normally and abnormally
distributed data between the healthy controls, and 2019nCoV
ICUadmitted patients, respectively. The mean ± SD describes the
descriptive data. Moreover, the ShapiroWilk test controlled the
data appropriateness to normal ranges. Additionally, p< .05 was
described as statistically significant. GraphPad Prism (version 7.00
for Windows; GraphPad Software; www.graphpad.com) was applied
to plot the graphs.
3|RESULTS
3.1 |Frequency of Th17 cells in controls and
COVID19 patients
The flow cytometry technique also analyses the circulating propor-
tion of circulating Th17 (CD4
+
IL17A
+
) cells in PBMCs of 2019nCoV
patients and the controls to compare the alteration of Th17 cells in
both groups. Based upon this, findings illustrated that the frequency
of Th17 cells was considerably built up in COVID19 patients than
controls (6.767 ± 2.831 vs. 3.306 ± 1.628, p< .0001; Figure 1a,b).
3.2 |Frequency of Treg cells in controls and
COVID19 patients
To determine the alteration of the peripheral Treg cell population
(CD4
+
CD25
+
CD127
cells) in both healthy controls and COVID19
patients, the frequency of Tregs was quantified in PBMCs by flow
cytometry analysis. Based on the obtained results, we found that the
number of circulating Treg cells was meaningfully reduced in patients
in comparison with the control group (3.08 ± 1.613 vs. 4.741 ± 2.052,
p< .0001; Figure 2a,b).
3.3 |Measurement of the RORɣt and FoxP3
expression levels
Due to the alterations in the number and the relevant factors of T cell
subsets in COVID19 patients, the expression levels of Th17 and Treg
transcription factors (RORɣt and FoxP3, respectively) were also mea-
sured by Realtime PCR. Considering the results, we found that gene
expression of RORɣt transcription factor had a significant increase in
COVID19 patients than controls (2.018 ± 0.830 vs. 0.992 ± 0.086,
p= .0001) (Figure 1c). In contrast, the gene expression of FoxP3 had a
remarkable decrease in patients compared with healthy individuals
(0.518 ± 0.291 vs. 1.001 ± 0.714, p< .0001; Figure 2c).
3.4 |Assessment of cytokine expression profile
To compare the alteration of cytokines in COVID19 patients and
controls, the expression levels of cytokines associated with Th17
TABLE 2 Primer sequence
Gene Primer Sequence
RORγtForward ACTCAAAGCAGGAGCAATGGAA
Reverse AGTGGGAGAAGTCAAAGATGGA
FoxP3 Forward TCATCCGCTGGGCCATCCTG
Reverse GTGGAAACCTCACTTCTTGGTC
TGFβForward CGACTACTACGCCAAGGA
Reverse GAGAGCAACACGGGTTCA
IL10 Forward CAT CGA TTT CTT CCC TGT GAA
Reverse TCTTGGAGCTTATTAAAGGCATTC
IL17 Forward CATAACCGGAATACCAATACCAAT
Reverse GGATATCTCTCAGGGTCCTCATT
IL23 Forward GGACAACAGTCAGTTCTGCTT
Reverse CACAGGGCTATCAGGGAGC
βActin Forward AGAGCTACGAGCTGCCTGAC
Reverse AGCACTGTGTTGGCGTACAG
4
|
SADEGHI ET AL.
cells (IL17 and IL23) and Treg cells (IL10 and TGFβ) were assessed
by realtime PCR. On one hand, the results demonstrated that the
mean cytokine expression of IL17 and IL23 cytokines was notably
escalated in patients when compared with controls (3.867 ± 2.383,
p< .0001, and 3.172 ± 1.240, p< .0001). On the other hand, notice-
able downregulation of IL10 and TGFβcytokines was detected in
the patient group compared with controls (0.376 ± 0.274, p< .0001
and 0.417 ± 0.297, p< .0001, respectively; Figure 3).
3.5 |The cytokine secretion levels in serum and
supernatant of cultured PBMCs
To examine the changes in secretion levels of cytokines between
patients and healthy subjects, the levels of IL17, IL23, IL10, and
TGFβcytokines were measured in serum samples and the super-
natant of PBMCs by ELISA. According to obtained results, we found
that due to the decrease in the level of Tregs in SARSCoV2 patients,
the mean secretion levels of the relevant cytokines, including IL10
and TGFβwere significantly reduced in patients rather than controls
in both the serum sample (17.35 ± 11.13 vs. 33.81 ± 21.87, p= .0005
and 37.64 ± 27.6 vs. 100.7 ± 57, p< .0001, respectively) and super-
natant of PBMCs (392.5 ± 235.6 vs. 812.6 ± 519.7, p= .0001 and
130.2 ± 82.83 vs. 229.5 ± 82.28, p< .0001, respectively) (Figure 4a,b).
By contrast, considering the elevated number of Th17 cells in
COVID19 patients, the mean secretion levels of IL17 and IL23
cytokines were strongly grown in both serum (27.35 ± 11.78 vs.
18.38 ± 7.775, p= .0012 and 41.69 ± 21.26 vs. 25.58 ± 13.37,
p= .0031, respectively) and the supernatant of cultured PBMCs
(179.0 ± 74.45 vs. 101.8 ± 53.46, p= .0001 and 0.417 ± 0.297 vs.
0.376 ± 0.274, p= .0001, respectively) of patients than that in heal-
thy individuals (Figure 4c,d).
3.6 |Th17/Treg ratios in COVID19 patient and
control groups
The ratios of Th17/Treg cells, their main cytokines, and transcription
factors were compared in both patient and control groups. As a re-
sult, the ratio of Th17/Treg cells was found greater in COVID19
infected patients when compared with the control group
(1.338 ± 0.544 vs. 0.706 ± 0.364, p< .0001). Additionally, the ex-
pression ratios of RORɣt/FoxP3 transcription factors and IL17/IL10
cytokines were markedly increased in COVID19 patients than in
healthy subjects (2.269 ± 1.241 vs. 0.512 ± 0.351, p< .0001 and
1.771 ± 1.23 vs. 0.51 ± 0.39, p< .0001, respectively; Figure 5).
3.7 |Th17/Treg cell ratio in COVID19 improved
and dead patients
To investigate the effects of Th17 and Treg cells, the imbalance of
the immune system, and the resulting inflammation in COVID19
mortality, we assessed the expression ratios of Th17/Treg cells, and
their associated cytokines and transcription factors in both 2019
nCoV improved and dead patients. Hence, we found that the ex-
pression ratios of all Th17/Treg cells (1.638 ± 0.592 vs.
1.189 ± 0.462, p= .023), IL17/IL10 cytokines (2.26 ± 0.564 vs.
1.76 ± 1.13, p= .017), and RORɣt/Foxp3 transcription factors
(2.501 ± 0.744 vs. 2.263 ± 1.241, p= .028) were notably higher in
dead patients when compared with improved cases (Figure 6).
4|DISCUSSION
With regard to the rapid outbreak of novel SARSCoV2 in the world
that originated from Wuhan, China, for rapid diagnosis and treat-
ment of infected patients, more knowledge of the viral
FIGURE 1 The number of Th17 cells and the expression level of
RORɣt in 2019nCoV patients and healthy subjects. (a) The counting
of the Th17 cells was done according to the representative dot plots
shown, which indicates the percentage of Th17 cells (CD4
+
IL17
+
).
(b) According to the flow cytometric results, the frequency of Th17
cells was considerably upregulated in 2019nCoV infected patients in
comparison with healthy subjects (p< .0001). (c) In comparison to
healthy controls, significant elevation was found in the expression
level of RORɣt in 2019nCoV patients based on Realtime PCR
findings (p= .0001). Patients group, n= 40; control group, n= 40.
Results were presented as mean ± SD;p< .05 was described as
statistically significant. Th, T helper; ROR, RARrelated orphan
receptors; 2019nCoV; 2019novel coronavirus
SADEGHI ET AL.
|
5
immunopathogenesis is required (Tahmasebi et al., 2020;Y.C. Wu,
Chen, et al., 2020). Inflammation caused by COVID19 elicits a range
of disorders from a mild coldlike infection to severe ARDS and da-
mage to other organs, including the liver, kidneys (Taghizadieh,
Mikaeili, Ahmadi, & Valizadeh, 2020), nervous system, and gastro-
intestinal tract. Humantohuman transmission of the virus from in-
fected patients or asymptomatic carriers causes the disease and is
characterized by serious symptoms such as fever, cough, shortness of
breath, and severe pneumonia, which mostly lead to death in patients
(Abdulamir & Hafidh, 2020; Riou & Althaus, 2020). It is important to
note that the risk of disease development and severe infection is
greater in older individuals and people with underlying diseases such
as hypertension, cardiovascular diseases, diabetes, autoimmunities,
immunodeficiencies, cancers, and viral infections (Q. Li, Guan, et al.,
2020; Stebbing et al., 2020).
ARDS caused by inflammatory responses led to the respiratory
dysfunction in 2019nCov disease. This is justified by the fact
that viral immunopathogenesis is initiated by the penetration of
SARSCoV2 into the alveolar epithelium of the lungs, which express
the virus receptor, termed as Angiotensinconverting enzyme 2
(ACE2). Thereafter, it can result in viral infection in the lungs and
other organs by entering the bloodstream. Following the entry of the
virus and its binding to the receptor, innate, and adaptive immune
responses are induced to defend against the infection. By the im-
mune system activation, antiviral responses are generated through
the immune cells and their produced inflammatory factors (G. Li, Fan,
et al., 2020; X. Li, Geng, Peng, Meng, & Lu, 2020).
As cytotoxic T cell (CTLs) and T helper (Th) cells, particularly Th1
and Th17 cells are the main defense against intracellular infections
like viral infections, so in COVID19, CTLs and Th1/17 cells strongly
fight the virus by secreting the perforin/granzyme and inflammatory
cytokines, respectively. Th cells also induce the activation of B cells,
macrophages, and neutrophils against the virus by secretingcytokines
(Liu, Du, et al., 2020;Tay,Poh,Rénia,MacAry,&Ng,2020). In the
course of the disease, inflammatory cytokines (IL1β,IL6, IL8, IL17,
GCSF, GMCSF, TNFα,IFNγ,IP10, and MCP1) are produced due to
the imbalance of the immune system, uncontrolled immune responses,
reduction of T regs and upregulation of inflammatory Th1 and Th17
cells. These inflammatory cytokines elicit the cytokine storm or cy-
tokine release syndrome (CRS), which results in severe inflammation
and respiratory system injury in infected patients (Huang et al., 2020;
Mehta et al., 2020). In most autoimmune and inflammatory diseases,
the disturbance balance of the Th17/Treg cells, the predominance of
the inflammatory responses of Th1 and Th17 cells, and the dysfunc-
tion of the regulatory T cells are the reasons for disease progression
(Dolati et al., 2018; Severson & Hafler, 2010).
FIGURE 2 Frequency of Treg cells and FoxP3 expression in COVID19 patients and healthy subjects. (a) The enumeration of the Treg cell
population was performed based on the representative dot plots shown, which demonstrates the percentage of Treg cells
(CD4
+
CD25
+
CD127
). (b) Flow cytometric findings indicated the meaningful reduction of Treg frequency in patients compared with controls
(p< .0001). (c) According to realtime PCR analysis, a remarkable smaller expression level of the FoxP3 transcription factor was detected in
COVID19 patients compared with controls (p< .0001). Patients group, n= 40; control group, n= 40. Results were presented as mean ± SD;
p< .05 was described as statistically significant. COVID19, coronaviruses disease2019; FoxP3, forkhead box P3; Treg, regulatory T
6
|
SADEGHI ET AL.
Accordingly, the current study was aimed to investigate the al-
terations of the Th17 and Treg cells and their relevant factors, in-
cluding transcription factors (RORγt and Foxp3) and the main
cytokines (IL17, IL23, IL10, and TGFβ) in 40 COVID19 patients
and 40 healthy controls. Also, to investigate the effect of in-
flammatory responses in pathogenesis and disease progression, we
compared the expression ratios of Th17/Treg cells, IL17/IL10
cytokines, and RORγt/FoxP3 transcription factors between
SARSCoV2 infected patients and healthy subjects and also between
two groups of dead and improved COVID19 patients. As mentioned,
Th17 and Treg cells are among the most important cells known in
COVID19 patients. The role of Th17 cells is well known in in-
flammatory and autoimmune diseases (Harrington et al., 2005). In
this regard, to examine the inflammatory responses of Th17 cells in
SARSCoV2 patients, the frequency of these cells was assessed by
flow cytometry. Based on the results, we observed that the number
of Th17 cells was noticeably higher in novel coronavirus infected
patients than in the healthy control group. Due to the importance of
the RORγt transcription factor and Th17 cellrelated cytokines (IL17
and IL23) that differentiate and mediate the inflammatory re-
sponses, respectively (EghbalFard et al., 2019), the gene expression
and protein secretion levels of the mentioned factors were evaluated
by Realtime PCR and ELISA techniques, respectively. The findings
illustrated that the expression levels of RORγt, IL17, and IL23
factors were strongly enhanced in patients compared with healthy
individuals. Additionally, the secretion levels of IL17 and IL23 cy-
tokines were notably higher in patients. IL17 is introduced as an
inflammatory and multifocal cytokine that leads to inflammatory
responses, whereas, IL23 is a modulatory cytokine that has a role in
the differentiation and expansion of TH17 cells. Th17 cells are dif-
ferentiated from Th0 and expanded by RORγt, IL6, and IL23 fac-
tors. It can be suggested that Th17 cells can cause inflammatory
responses in SARSCoV2 patients by producing the proinflammatory
cytokines (IL17 [A to F)] IL21, IL22, and IL26; Annunziato et al.,
2007), along with the inflammatory cytokines secreted by Th1 cells.
This inflammation leads to respiratory system injury and other organ
failures in the course of the disease.
Also, as mentioned previously, the T reg cells are reduced in
autoimmune and inflammatory diseases, which cause disease pro-
gression (Riou & Althaus, 2020). Therefore, the frequency of Treg
cells was also measured, and the results showed a momentous de-
crease in the number of Tregs in 2019nCoV patients compared with
controls. Accordingly, we designed the study to compare the al-
terations in Tregs and related factors (FoxP3, IL10, and TGFβ)
between the patient and control groups, as well. The results showed
that the number of Tregs in patients was remarkably decreased
compared with controls. Besides this, a considerable reduction in the
expression levels of FoxP3 transcription factor and IL10 and TGFβ
cytokines and also the secretion levels of the mentioned cytokines
were detected in COVID19 patients. Treg cells
(CD4+CD25+FoxP3+) are the immune system regulators that char-
acterized by expressing the CTLA4, CD95 (Fas), and TNFR family
proteins. Tregs inhibit the overactivation of T cells and maintain the
immune system balance, tolerance, and hemostasis during the au-
toimmune and inflammatory diseases (Pette et al., 1990). In this
context, FoxP3 as a transcription factor of Tregs is induced by TGFβ,
which mediates the differentiation and development of Treg cells
from naive CD4+ T cells. IL10, TGFβ, and IL35 are the main anti
inflammatory cytokines of Tregs that play principal roles in sup-
pressing the immune responses and inflammation (Kulkarni et al.,
1993; Lee, Severin, & LovettRacke, 2017). Based on these data, in-
flammation can be considered as the main cause of disease patho-
genesis due to the high frequency of Th17 cells and their relevant
factors (RORγt, IL17, and IL23) as well as the low number of Tregs
and their factors (FoxP3, IL10, and TGFβ), in 2019nCoV patients.
In this regard, some other studies support these findings. Accord-
ingly, in a study by Qin et al. (2020), different subsets of T cells were
examined in a cohort of 452 patients with laboratoryconfirmed
2019nCoV in Wuhan, China. The findings showed the increased
levels of naive Th cells and decreased levels of Tregs (mainly induced
Tregs), in severe COVID19 patients, which suggested the important
role of Th and Treg cells in disease progression and immune system
regulation, respectively. Furthermore, recent studies have high-
lighted that the enhanced migration of neutrophils and inflammatory
responses of Th17 cells play substantial roles in inflammation,
pneumonia, and edema in COVID19 patients; whereas, Tregs are
downregulated in the course of the disease (Hoe et al., 2017;Wu&
Yang, 2020). On the other hand, given that the IL17 cytokine can
FIGURE 3 The cytokine expression levels of Treg and Th17 cells
in COVID19 patients and controls. The gene expression analysis
revealed the considerable enhancement in the expression levels of
IL10 and TGFβ, the antiinflammatory cytokines of Tregs, in
COVID19 patients compared with healthy subjects (p< .0001 and
p< .0001, respectively). In contrast, results showed that IL17 and
IL23, the inflammatory cytokines of Th17 cells, were significantly
downregulated in the patient groups than controls (p< .0001 and
p< .0001, respectively). Patients group, n= 40; control group, n= 40.
Results were presented as mean ± SD.p< .05 was described as
statistically significant. COVID19, coronaviruses disease
2019; FoxP3, forkhead box P3; IL, interleukin; ROR, RARrelated
orphan receptors; TH, T helper; Treg, regulatory T
SADEGHI ET AL.
|
7
induce pulmonary eosinophilic and allergic responses, it can cause
inflammation in SARSCoV2 disease by recruiting the eosinophils
into the lungs (AlRamli et al., 2009; Cheung, Wong, & Lam, 2008). In
another investigation conducted by Liu, Zhang, et al. (2020), it was
found that inflammatory cytokines, including IL2, 4, 7, 10, 12,
and 17, IFNγ, IFNα2, MCSF, GCSF, and IP10 were built up in
COVID19 ICU patients compared with nonICU cases, which were
associated with lung injury. Some of these cytokines, such as IL1β
and TNFα, induce the responses of Th17 and Th1 cells. Thereby, it
has been suggested that Th17 cells play a prominent role in the
FIGURE 4 The secretion levels of cytokines in serum samples and supernatant of cultured PBMCs in COVID19 patients and control
individuals. (a) The secretion levels of IL10 and TGFβin the serum sample of patients were noticeably lower than controls (p= .0005 and
p<.0001, respectively). (b) A significant downregulation was found in the secretion levels of IL10 and TGFβcytokines in PBMCs' supernatant
of patients than the control group (p= .0001 and p< .0001, respectively). (c) Both IL17 and IL23 inflammatory cytokines were greatly
increased in the serum samples of COVID19 patients compared with healthy subjects (p= .0012 and p= .0031, respectively). (d) In the patient
group, there was a remarkable elevation in the secretion levels of IL17 and IL23 cytokines in the supernatant of PBMCs compared with the
control group (p= .0001 and p= .0008, respectively). Patients group, n= 40; control group, n= 40. Results were presented as mean ± SD;p< .05
was described as statistically significant. COVID19, coronaviruses disease2019; IL, interleukin; PBMC, peripheral blood mononuclear cell;
TGFβ, transforming growth factorβ
FIGURE 5 The expression ratio of Th17/Treg cells and their related factors in COVID19 cases and healthy controls. (a) In comparison
between patients and controls, the expression ratio of IL17/IL10 cytokines and RORɣt /FoxP3 transcription factors were notably higher in
COVID19 patients (p< .0001 and p< .0001, respectively). (b) According to obtained results, the expression ratio of Th17/Treg cells was
increased in patients when compared with controls (p< .0001). Patients group, n= 40; control group, n= 40. Results were presented as
mean ± SD.p< .05 was described as statistically significant. COVID19; coronaviruses disease2019; FoxP3, forkhead box P3; IL, interleukin;
ROR, RARrelated orphan receptors; Th, T helper; Treg, regulatory T
8
|
SADEGHI ET AL.
disease inflammation by producing the IL17, IL21, IL22, and GM
CSF cytokines.
Moreover, Xu et al. (2020) documented the elevated levels of
CCR6+ Th17 cells in SARSCoV2 patients with severe conditions,
which were involved in cytokine storm and inflammation, further
supporting our findings. In another investigation, Huang et al. found
that the intensivecare COVID19 patients had boosted in-
flammatory cytokine levels like IL17 compared with nonintensive
care patients. Therefore, the hypothesis was suggested that cytokine
inhibition could reduce inflammation, improve the patient outcomes,
and reduce the mortality rate (Antalis et al., 2019; Huang et al., 2020;
Ma, Yao, Peng, & Chen, 2019). Furthermore, the results of studies on
SARS and MERS coronaviruses evidenced the raised levels of Th17
cells and IL17related pathways in the mentioned diseases that had
inflammatory roles in disease progression (Faure et al., 2014; Josset
et al., 2013). Also, higher levels of IL17 cytokine and decreased
levels of IFNγand IFNαcytokines have been reported, as in-
flammatory responses in MERSCoV patients (Faure et al., 2014).
Some related studies have shown a close correlation between the
severity of the MERSCoV, SARSCoV, and SARSCoV2 diseases and
the grown levels of Th17 cells and their relevant cytokines, including
IL1, IL6, IL15, IL17, IFNγ, and TNFα(Liu, 2019; Mahallawi,
Khabour, Zhang, Makhdoum, & Suliman, 2018).
In conclusion, based on the results of our study, as well as
supporting studies on MERSCoV, SARSCoV, and SARSCoV2, it can
be suggested that the elevated levels of Th17 cells and their asso-
ciated factors (RORγt, IL17, and IL23), the decreased frequency of
Treg cells and their relevant factors (FoxP3, TGFβ, and IL10), and
imbalanced ratios of Th17/Treg cells may play a critical role in in-
creasing the inflammatory responses and the disease pathogenesis in
COVID19 patients. Accordingly, among these responses, an im-
paired respiratory system can be strongly related to the hyperin-
flammation, eosinophilic responses, allergic disorders, and CRS
derived from the function of Th17 cells.
ACKNOWLEDGMENTS
This study was supported by the Tuberculosis and Lung Disease
Research Center of Tabriz University of Medical Sciences, Tabriz,
Iran (Grant Number: 65235).
CONFLICT OF INTERESTS
The authors declare that there are no conflict of interests.
AUTHOR CONTRIBUTIONS
Conceptualization, patient selection, writingoriginal draft: Armin
Sadeghi. Conceptualization, writingoriginal draft: Safa Tahmasebi.
Revised article and final edition: Arshad Mahmood and Maria
Kuznetsova. Conceptualization, introducing, and selecting patients: Leili
AghebatiMaleki and Hamed Valizadeh. Conceptualization, data cura-
tion: Ali Taghizadieh.Conceptualization, data curation: Masoud
Nazemiye. Formal analysis: Farhad JadidiNiaragh. Investigation: Sanaz
AbbaspourAghdam. Investigation: Leila Roshangar. Supervision,
Validation, review & editing: Haleh Mikaeili. Supervision, validation, re-
view and editing: Majid Ahmadi.
DATA AVAILABILITY STATEMENT
Data supporting the findings in this study are immediately available
upon reasonable request.
ORCID
Leila Roshangar http://orcid.org/0000-0001-5329-0951
Majid Ahmadi http://orcid.org/0000-0001-9845-9140
REFERENCES
Abdulamir, A. S., & Hafidh, R. R. (2020). The possible immunological
pathways for the variable immunopathogenesis of COVID19
infections among healthy adults, elderly and children. Electronic
Journal of General Medicine,17(4), 14.
Ahmadi, M., AbdolmohammadiVahid, S., Ghaebi, M., AghebatiMaleki, L.,
Dolati, S., Farzadi, L., Yousefi, M. (2017). Regulatory T cells
FIGURE 6 The expression ratio of Th17/Treg cells and their related factors in COVID19 improved and dead patients. (a) A significant
increase in the expression ratios of both IL17/IL10 cytokines and RORɣt/FoxP3 transcription factors were detected in COVID19 dead
patients compared with the improved cases (p= .017 and p= .028, respectively). (b) The COVID19 dead patients have a considerably higher
ratio of Th17/Treg cells than improved patients (p= .023). Patients group, n= 40; control group, n= 40. Results were presented as mean ± SD.
p< .05 was described as statistically significant. COVID19, coronaviruses disease2019; FoxP3, forkhead box P3; IL, interleukin;
ROR, RARrelated orphan receptors; TH, T helper; Treg, regulatory T
SADEGHI ET AL.
|
9
improve pregnancy rate in RIF patients after additional IVIG
treatment. Systems Biology in Reproductive Medicine,63(6), 350359.
AlRamli, W., Préfontaine, D., Chouiali, F., Martin, J. G., Olivenstein, R.,
Lemière, C., & Hamid, Q. (2009). TH17associated cytokines (IL17A
and IL17F) in severe asthma. Journal of Allergy and Clinical
Immunology,123(5), 11851187.
Annunziato, F., Cosmi, L., Santarlasci, V., Maggi, L., Liotta, F., Mazzinghi, B.,
Frosali, F. (2007). Phenotypic and functional features of human
Th17 cells. Journal of Experimental Medicine,204(8), 18491861.
Antalis, E., Spathis, A., Kottaridi, C., Kossyvakis, A., Pastellas, K.,
Tsakalos, K., Tsiodras, S. (2019). Th17 serum cytokines in relation
to laboratoryconfirmed respiratory viral infection: A pilot study.
Journal of Medical Virology,91(6), 963971.
Braitch, M., Harikrishnan, S., Robins, R., Nichols, C., Fahey, A., Showe, L., &
Constantinescu, C. (2009). Glucocorticoids increase CD4+
CD25high cell percentage and Foxp3 expression in patients with
multiple sclerosis. Acta Neurologica Scandinavica,119(4), 239245.
Cheung, P. F., Wong, C. K., & Lam, C. W. (2008). Molecular mechanisms
of cytokine and chemokine release from eosinophils activated by
IL17A, IL17F, and IL23: Implication for Th17 lymphocytes
mediated allergic inflammation. The Journal of Immunology,180(8),
56255635.
Dolati, S., Ahmadi, M., Rikhtegar, R., Babaloo, Z., Ayromlou, H.,
AghebatiMaleki, L., Yousefi, M. (2018). Changes in Th17 cells
function after nanocurcumin use to treat multiple sclerosis.
International Immunopharmacology,61,7481.
EghbalFard, S., Yousefi, M., Heydarlou, H., Ahmadi, M., Taghavi, S.,
Movasaghpour, A., AghebatiMaleki, L. (2019). The imbalance of
Th17/Treg axis involved in the pathogenesis of preeclampsia.
Journal of Cellular Physiology,234(4), 51065116.
Faure, E., Poissy, J., Goffard, A., Fournier, C., Kipnis, E., Titecat, M.,
Dessein, R. (2014). Distinct immune response in two MERSCoV
infected patients: Can we go from bench to bedside? PLOS One,9(2),
e88716.
Ghaebi, M., Osali, A., Valizadeh, H., Roshangar, L., & Ahmadi, M. (2020).
Vaccine development and therapeutic design for 2019nCoV/SARS
CoV2: Challenges and chances. Journal of Cellular Physiology.
https://doi.org/10.1002/jcp.29771
Harrington, L. E., Hatton, R. D., Mangan, P. R., Turner, H., Murphy, T. L.,
Murphy, K. M., & Weaver, C. T. (2005). Interleukin 17producing
CD4+ effector T cells develop via a lineage distinct from the T
helper type 1 and 2 lineages. Nature Immunology,6(11), 11231132.
Hoe, E., Anderson, J., Nathanielsz, J., Toh, Z. Q., Marimla, R., Balloch, A., &
Licciardi, P. V. (2017). The contrasting roles of Th17 immunity in
human health and disease. Microbiology and Immunology,61(2),
4956.
Huang, C., Wang, Y., Li, X., Ren, L., Zhao, J., Hu, Y., Gu, X. (2020). Clinical
features of patients infected with 2019 novel coronavirus in Wuhan,
China. The Lancet,395(10223), 497506.
Jose, R. J., & Manuel, A. (2020). COVID19 cytokine storm: The interplay
between inflammation and coagulation. The Lancet Respiratory
Medicine,8, e46e47.
Josset, L., Menachery, V. D., Gralinski, L. E., Agnihothram, S., Sova, P.,
Carter, V. S., Katze, M. G. (2013). Cell host response to infection
with novel human coronavirus EMC predicts potential antivirals and
important differences with SARS coronavirus. mBio,4(3),
e0016500113.
Kulkarni, A. B., Huh, C.G., Becker, D., Geiser, A., Lyght, M., Flanders, K. C.,
Karlsson, S. (1993). Transforming growth factor beta 1 null
mutation in mice causes excessive inflammatory response and early
death. Proceedings of the National Academy of Sciences,90(2),
770774.
Lee, P. W., Severin, M. E., & LovettRacke, A. E. (2017). TGFβregulation
of encephalitogenic and regulatory T cells in multiple sclerosis.
European Journal of Immunology,47(3), 446453.
Li, G., Fan, Y., Lai, Y., Han, T., Li, Z., Zhou, P., Liu, X. (2020). Coronavirus
infections and immune responses. Journal of Medical Virology,92(4),
424432.
Li, Q., Guan, X., Wu, P., Wang, X., Zhou, L., Tong, Y., Wong, J. Y. (2020).
Early transmission dynamics in Wuhan, China, of novel
coronavirusinfected pneumonia. New England Journal of Medicine,
382, 11991207.
Li, X., Geng, M., Peng, Y., Meng, L., & Lu, S. (2020). Molecular immune
pathogenesis and diagnosis of COVID19. Journal of Pharmaceutical
Analysis,10, 102108.
Lin, L., Lu, L., Cao, W., & Li, T. (2020). Hypothesis for potential
pathogenesis of SARSCoV2 infectionA review of immune
changes in patients with viral pneumonia. Emerging Microbes &
Infections,9(1), 727732.
Liu, Y. (2019). Novel coronavirus (2019nCoV) infections trigger an ex-
aggerated cytokine response aggravating lung injury.
Liu, Y., Du, X., Chen, J., Jin, Y., Peng, L., Wang, H. H., Zhao, Y. (2020).
Neutrophiltolymphocyte ratio as an independent risk factor for
mortality in hospitalized patients with COVID19. Journal of
Infection,81,e6e12.
Liu, Y., Zhang, C., Huang, F., Yang, Y., Wang, F., Yuan, J., Zhao, D. (2020).
Elevated plasma level of selective cytokines in COVID19 patients
reflect viral load and lung injury. National Science Review,7, 10031011.
Lu, R., Zhao, X., Li, J., Niu, P., Yang, B., Wu, H., Zhu, N. (2020). Genomic
characterisation and epidemiology of 2019 novel coronavirus:
Implications for virus origins and receptor binding. The Lancet,
395(10224), 565574.
Ma, W. T., Yao, X. T., Peng, Q., & Chen, D. K. (2019). The protective and
pathogenic roles of IL17 in viral infections: friend or foe? Open
Biology,9(7), 190109.
Mahallawi, W. H., Khabour, O. F., Zhang, Q., Makhdoum, H. M., &
Suliman, B. A. (2018). MERSCoV infection in humans is associated
with a proinflammatory Th1 and Th17 cytokine profile. Cytokine,
104,813.
Mehta, P., McAuley, D. F., Brown, M., Sanchez, E., Tattersall, R. S., &
Manson, J. J. (2020). COVID19: Consider cytokine storm syndromes
and immunosuppression. The Lancet,395(10229), 10331034.
Pette, M., Fujita, K., Kitze, B., Whitaker, J., Albert, E., Kappos, L., &
Wekerle, H. (1990). Myelin basic proteinspecific T lymphocyte lines
from MS patients and healthy individuals. Neurology,40(11), 1770.
Prompetchara, E., Ketloy, C., & Palaga, T. (2020). Immune responses in
COVID19 and potential vaccines: Lessons learned from SARS and
MERS epidemic. Asian Pacific Journal of Allergy and Immunology,
38(1), 19.
Qin, C., Zhou, L., Hu, Z., Zhang, S., Yang, S., Tao, Y., Wang, W. (2020).
Dysregulation of immune response in patients with COVID19 in
Wuhan, China. Clinical Infectious Diseases.
Riou, J., & Althaus, C. L. (2020). Pattern of early humantohuman
transmission of Wuhan 2019 novel coronavirus (2019nCoV),
December 2019 to January 2020. Eurosurveillance,25(4), 2000058.
Severson, C., & Hafler, D. A. (2010). Tcells in multiple sclerosis. Results
and Problems in Cell Differentiation,51,7598.
Shi, Y., Tan, M., Chen, X., Liu, Y., Huang, J., Ou, J., & Deng, X. (2020).
Immunopathological characteristics of coronavirus disease 2019
cases in Guangzhou. China. medRxiv.
Stebbing, J., Phelan, A., Griffin, I., Tucker, C., Oechsle, O., Smith, D., &
Richardson, P. (2020). COVID19: Combining antiviral and anti
inflammatory treatments. The Lancet Infectious Diseases,20(4),
400402.
Taghizadieh, A., Mikaeili, H., Ahmadi, M., & Valizadeh, H. (2020). Acute
kidney injury in pregnant women following SARSCoV2 infection: A
case report from Iran. Respiratory Medicine Case Reports,30, 101090.
Tahmasebi, S., Khosh, E., & Esmaeilzadeh, A. (2020). The outlook for
diagnostic purposes of the 2019novel coronavirus disease. Journal
of Cellular Physiology.119. Advance online publication.
10
|
SADEGHI ET AL.
Tay, M. Z., Poh, C. M., Rénia, L., MacAry, P. A., & Ng, L. F. (2020). The
trinity of COVID19: Immunity, inflammation and intervention.
Nature Reviews Immunology,20(6), 363374.
Wan, S., Yi, Q., Fan, S., Lv, J., Zhang, X., Guo, L., Yi, Z. (2020).
Characteristics of lymphocyte subsets and cytokines in peripheral
blood of 123 hospitalized patients with 2019 novel coronavirus
pneumonia (NCP). MedRxiv.
Wu, D., & Yang, X. O. (2020). TH17 responses in cytokine storm of
COVID19: An emerging The outlook for diagnostic purposes of
thetarget of JAK2 inhibitor Fedratinib. Journal of Microbiology,
Immunology and Infection,53, 368370.
Wu, F., Zhao, S., Yu, B., Chen, Y.M., Wang, W., Song, Z.G., Pei, Y.Y.
(2020). A new coronavirus associated with human respiratory
disease in China. Nature,579(7798), 265269.
Wu, Y.C., Chen, C.S., & Chan, Y.J. (2020). The outbreak of COVID19:
An overview. Journal of the Chinese Medical Association,83(3),
217220.
Xu, Z., Shi, L., Wang, Y., Zhang, J., Huang, L., Zhang, C., Wang, F. S.
(2020). Pathological findings of COVID19 associated with acute
respiratory distress syndrome. The Lancet,8(4), 420422.
Yin, Y., & Wunderink, R. G. (2018). MERS, SARS and other coronaviruses
as causes of pneumonia. Respirology,23(2), 130137.
Zhou, P., Yang, X.L., Wang, X.G., Hu, B., Zhang, L., Zhang, W., Huang,
C.L. (2020). A pneumonia outbreak associated with a new
coronavirus of probable bat origin. Nature,579(7798), 270273.
How to cite this article: Sadeghi A, Tahmasebi S, Mahmood A,
et al. Th17 and Treg cells function in SARSCoV2 patients
compared with healthy controls. J Cell Physiol. 2020;111.
https://doi.org/10.1002/jcp.30047
SADEGHI ET AL.
|
11
... As far as we know, an elevated number of Th17 lymphocytes and their related factors, a reduced number of regulatory T (Treg) cells, the disruption of the Th17/Treg cells balance, as well as hyperinflammation all play an important role in the pathogenesis of several immunological diseases, including SARS-CoV2 [28], MS [29], ankylosing spondylitis (AS) [30,31], Behcet's disease [32], Recurrent implantation failure [33,34], recurrent pregnancy loss [35,36], and recurrent miscarriage [37]. In the course of COPD, the lack of balance between pro-inflammatory and anti-inflammatory immune cell responses regulated by Th17 and Treg has been demonstrated to play a critical function in the pathogenesis of COPD [38]. ...
Article
Full-text available
Chronic obstructive pulmonary disease (COPD) is a chronic inflammatory process in the airways that results in airflow obstruction. It is mainly linked to cigarette smoke exposure. Th17 cells have a role in the pathogenesis of COPD by secreting pro-inflammatory cytokines, which cause hyperinflammation and progression of the disease. This study aimed to assess the potential therapeutic effects of nanocurcumin on the Th17 cell frequency and its responses in moderate and severe COPD patients. This study included 20 patients with severe COPD hospitalized in an intensive care unit (ICU) and 20 patients with moderate COPD. Th17 cell frequency, Th17-related factors gene expression (RAR-related orphan receptor t (RORγt), IL-17, IL-21, IL-23, and granulocyte-macrophage colony-stimulating factor), and serum levels of Th17-related cytokines were assessed before and after treatment in both placebo and nanocurcumin-treated groups using flow cytometry, real-time PCR, and ELISA, respectively. According to our findings, in moderate and severe nanocurcumin-treated COPD patients, there was a substantial reduction in the frequency of Th17 cells, mRNA expression, and cytokines secretion level of Th17-related factors compared to the placebo group. Furthermore, after treatment, the metrics mentioned above were considerably lower in the nanocurcumin-treated group compared to before treatment. Nanocurcumin has been shown to decrease the number of Th17 cells and their related inflammatory cytokines in moderate and severe COPD patients. As a result, it might be used as an immune-modulatory agent to alleviate the patient's inflammatory state.
... Firstly, Tregs, gamma delta T cells, CD8 + T cells, resting memory CD4 + T cells, resting NK cells, Neutrophils and Monocytes were associated with the immune patterns and ICI scores in this work. Lower level of Tregs could aggravate the damage in severe COVID-19 patients and Tregs played an immune-related role in the disease progression of COVID-19 [26,27]. It was demonstrated that gamma delta T cells served as a determining function in severity of COVID-19, and thus were often considered as the main indicator for COVID-19 symptoms [28,29]. ...
Article
Full-text available
Purpose Although the role of SARS-CoV-2-specfic immune cells has been revealed, a comprehensive understanding of immune patterns remains unknown. Methods In this work, unsupervised consensus clustering analysis was used to classify 240 coronavirus disease 2019 (COVID-19) patients into different immune subtypes. Next, we performed differentially expressed analysis between different immune subtypes. Functional enrichment and pathway analyses were employed to reveal the biological significance of these differentially expressed genes (DEGs). Besides, we compared feature score of some DEGs between whole blood and lung tissues. Then, we utilized the “GSVA” algorithm to construct an immune cell infiltrating (ICI) tool based on the categories of these DEGs. Finally, we developed a nomogram associated with severity of COVID-19. Results As a result, we identified two immune subtypes, and 238 DEGs which mainly participated in some immune-related functions and the COVID-19 pathway. Most importantly, the 238 DEGs could reflect the characterization of immune patterns in lung tissues. ICI scores were markedly negative associated with immune scores. It was worth noting that ICI score was a strong indicator for severity of COVID-19 and could accurately predict the severity of COVID-19. Conclusion Our findings could provide more valuable strategies for the management of COVID-19.
... This finding is remarkable because besides COVID19 IL-17 is affiliated with other clinical pathophysiologies in which a dysregulation between innate and adaptive immune responses such as myocarditis and lupus (S. Y. Lee et al., 2019 ;Rangachari et al., 2006 ;Sadeghi et al., 2021 ). ...
Preprint
Patients present a wide range of clinical severities in response SARS-CoV-2 infection, but the underlying molecular and cellular reasons why clinical outcomes vary so greatly within the population remains unknown. Here, we report that negative clinical outcomes in severely ill patients were associated with divergent RNA transcriptome profiles in peripheral immune cells compared with mild cases during the first weeks after disease onset. Protein-protein interaction analysis indicated that early-responding cytotoxic NK cells were associated with an effective clearance of the virus and a less severe outcome. This innate immune response was associated with the activation of select cytokine-cytokine receptor pathways and robust Th1/Th2 cell differentiation profiles. In contrast, severely ill patients exhibited a dysregulation between innate and adaptive responses affiliated with divergent Th1/Th2 profiles and negative outcomes. This knowledge forms the basis of clinical triage that may be used to preemptively detect high-risk patients before life-threatening outcomes ensue. – Mild COVID-19 patients presented an early compromise with NK cell function, whereas severe patients do so with neutrophil function. – The identified co-expressed genes give insights into a coordinated transcriptional program of NK cell cytotoxic activity being associated with mild patients. – Key checkpoints of NK cell cytotoxicity that were enriched in mild patients include: KLRD1 , CD247 , and IFNG . – The early innate immune response related to NK cells connects with the Th1/Th2 adaptive immune responses, supporting their relevance in COVID-19 progression.
... In the acute phase of COVID-19, there is T cell lymphopenia with CD8 T cells displaying a hyperactivated phenotype, followed by the appearance of T cells with features of senescence and exhaustion [18]. Furthermore, a shift in T cell responses towards a pro-inflammatory Th17 phenotype [19] and altered composition of regulatory T cells [20] results in severe inflammation and respiratory system injury in COVID-19, with this Th17/T reg imbalance associated with poor prognosis [21]. The number of Natural Killer (NK) cells, which also play a vital role in the clearance of viral infections, are reduced in the acute phase of COVID-19, and these cells also show features of senescence and functional impairment in severe disease [22]. ...
Article
Full-text available
Background The striking increase in COVID-19 severity in older adults provides a clear example of immunesenescence, the age-related remodelling of the immune system. To better characterise the association between convalescent immunesenescence and acute disease severity, we determined the immune phenotype of COVID-19 survivors and non-infected controls. Results We performed detailed immune phenotyping of peripheral blood mononuclear cells isolated from 103 COVID-19 survivors 3–5 months post recovery who were classified as having had severe ( n = 56; age 53.12 ± 11.30 years), moderate ( n = 32; age 52.28 ± 11.43 years) or mild ( n = 15; age 49.67 ± 7.30 years) disease and compared with age and sex-matched healthy adults ( n = 59; age 50.49 ± 10.68 years). We assessed a broad range of immune cell phenotypes to generate a composite score, IMM-AGE, to determine the degree of immune senescence. We found increased immunesenescence features in severe COVID-19 survivors compared to controls including: a reduced frequency and number of naïve CD4 and CD8 T cells ( p < 0.0001); increased frequency of EMRA CD4 ( p < 0.003) and CD8 T cells ( p < 0.001); a higher frequency ( p < 0.0001) and absolute numbers ( p < 0.001) of CD28 −ve CD57 +ve senescent CD4 and CD8 T cells; higher frequency ( p < 0.003) and absolute numbers ( p < 0.02) of PD-1 expressing exhausted CD8 T cells; a two-fold increase in Th17 polarisation ( p < 0.0001); higher frequency of memory B cells ( p < 0.001) and increased frequency ( p < 0.0001) and numbers ( p < 0.001) of CD57 +ve senescent NK cells. As a result, the IMM-AGE score was significantly higher in severe COVID-19 survivors than in controls ( p < 0.001). Few differences were seen for those with moderate disease and none for mild disease. Regression analysis revealed the only pre-existing variable influencing the IMM-AGE score was South Asian ethnicity ( $$\beta$$ β = 0.174, p = 0.043), with a major influence being disease severity ( $$\beta$$ β = 0.188, p = 0.01). Conclusions Our analyses reveal a state of enhanced immune ageing in survivors of severe COVID-19 and suggest this could be related to SARS-Cov-2 infection. Our data support the rationale for trials of anti-immune ageing interventions for improving clinical outcomes in these patients with severe disease.
... Its relevance has been recently explored in other viral diseases, including COVID-19 72 . Furthermore, a few studies have shown that it is important in cryptosporidiosis, with a rapid induction in the intestine of mice and bovines 73,74 . At 24 hpi, the genes involved in this pathway and that were upregulated in BcoV-infected cells included FOS (FC = 2), CXCL2 (FC = 8), CXCL3 (FC = 2.56), IL-8 (FC = 13.95) and PTGS2 (FC = 1.72). ...
Article
Full-text available
Among the causative agents of neonatal diarrhoea in calves, two of the most prevalent are bovine coronavirus (BCoV) and the intracellular parasite Cryptosporidium parvum. Although several studies indicate that co-infections are associated with greater symptom severity, the host–pathogen interplay remains unresolved. Here, our main objective was to investigate the modulation of the transcriptome of HCT-8 cells during single and co-infections with BCoV and C. parvum. For this, HCT-8 cells were inoculated with (1) BCoV alone, (2) C. parvum alone, (3) BCoV and C. parvum simultaneously. After 24 and 72 h, cells were harvested and analyzed using high-throughput RNA sequencing. Following differential expression analysis, over 6000 differentially expressed genes (DEGs) were identified in virus-infected and co-exposed cells at 72 hpi, whereas only 52 DEGs were found in C. parvum-infected cells at the same time point. Pathway (KEGG) and gene ontology (GO) analysis showed that DEGs in the virus-infected and co-exposed cells were mostly associated with immune pathways (such as NF-κB, TNF-α or, IL-17), apoptosis and regulation of transcription, with a more limited effect exerted by C. parvum. Although the modulation observed in the co-infection was apparently dominated by the virus, over 800 DEGs were uniquely expressed in co-exposed cells at 72 hpi. Our findings provide insights on possible biomarkers associated with co-infection, which could be further explored using in vivo models.
Article
Regulatory T (TREG) cells are involved in the antiviral immune response in patients with COVID-19; however, whether TREG cells are involved in the neutralizing antibody (nAb) response remains unclear. Here, we found that individuals who recovered from mild but not severe COVID-19 had significantly greater frequencies of TREG cells and lower frequencies of CXCR3+ circulating TFH (cTFH) cells than healthy controls. Furthermore, TREG and CXCR3+ cTFH cells were negatively and positively correlated with the nAb responses, respectively, and TREG cells was inversely associated with CXCR3+ cTFH cells in individuals who recovered from mild COVID-19 but not in those with severe disease. Mechanistically, TREG cells inhibited memory B-cell differentiation and antibody production by limiting the activation and proliferation of cTFH cells, especially CXCR3+ cTFH cells, and functional molecule expression. This study provides novel insight showing that mild COVID-19 elicits a concerted nAb responses which are shaped by both TREG and TFH cells.
Article
Rationale During the past 3 years of the corona virus disease 2019 (COVID-19) pandemic, COVID-19 has been recognized to cause various neurological complications, including rare posterior reversible encephalopathy syndrome (PRES). In previously reported cases of PRES associated with COVID-19, the majority of patients had severe COVID-19 infection and known predisposing factors for PRES, such as uncontrolled hypertension, renal dysfunction, and use of immunosuppressants. It remains unclear whether these risk factors or infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) contributes to the development of PRES in these patients. Here we report a special case of PRES associated with COVID-19 without any known risk factors for PRES, indicating the SARS-CoV-2’s direct role in the pathogenesis of PRES associated with COVID-19. Patient concerns An 18-year-old female patient presented to the emergency department with abdominal pain. Preliminary investigations showed no abnormalities, except for positive results in novel coronavirus nucleic acid tests using oropharyngeal swabs. However, the patient subsequently developed tonic–clonic seizures, headaches, and vomiting on the second day. Extensive investigations have been performed, including brain MRI and lumbar puncture. Brain MRI showed hypointense T1-weighted and hyperintense T2-weighted lesions in the bilateral occipital, frontal, and parietal cortices without enhancement effect. Blood and cerebrospinal fluid analyses yielded negative results. The patient had no hypertension, renal insufficiency, autoimmune disease, or the use of immunosuppressants or cytotoxic drugs. Diagnoses PRES was diagnosed based on the clinical features and typical MRI findings of PRES. Interventions Symptomatic treatments such as anticonvulsants were administered to the patients. Outcomes The patient fully recovered within 1 week. The initial MRI abnormalities also disappeared completely on a second MR examination performed 11 days later, supporting the diagnosis of PRES. The patient was followed up for 6 months and remained in a normal state. Lessons The current case had no classical risk factors for PRES, indicating that although the cause of PRES in COVID-19 patients may be multifactorial, the infection of SARS-CoV-2 may play a direct role in the pathogenesis of PRES associated with COVID-19.
Preprint
Full-text available
Recent studies have revealed gene-expression signatures in response to vaccination; however, the epigenetic regulatory mechanisms that are underlying vaccine-induced immune responses remains to be elucidated. Here, we analyzed a multiomics single-nucleus data of 267,485 nuclei at 10 timepoints after SARS-CoV-2 inactivated vaccination, which showed critical time points on gene changes in each celltype. We identified some epigenetically distinct monocyte subtypes, which were characterized by persistent chromatin remodeling at AP-1-targeted binding sites after the second dose, which were only transiently activated after the first dose. This remolded chromatin correspond to changes in cytokine and interferon pathways overtime. We found a coordinated regulation of IL1B, CXCL8, CCL3 and CSF2RA by c-Fos, c-Jun, IRF family and RUNX in myeloid cells. Pseudotime analysis revealed that CD14⁺ monocytes tend to differentiate towards a highly inflammatory state, while high-inflammation state is characterized by prolonged open chromatin after transcriptional termination. These findings demonstrate that two-dose vaccination stimulates persistent epigenetic remodeling of the innate immune cells and highlight the potential of temporal and spatial specific regulatory elements to optimizing vaccines.
Preprint
Full-text available
Patients present a wide range of clinical severities in response SARS-CoV-2 infection, but the underlying molecular and cellular reasons why clinical outcomes vary so greatly within the population remains unknown. Here, we report that negative clinical outcomes in severely ill patients were associated with divergent RNA transcriptome profiles in peripheral immune cells compared with mild cases during the first weeks after disease onset. Protein-protein interaction analysis indicated that early-responding cytotoxic NK cells were associated with an effective clearance of the virus and a less severe outcome. This innate immune response was associated with the activation of select cytokine-cytokine receptor pathways and robust Th1/Th2 cell differentiation profiles. In contrast, severely ill patients exhibited a dysregulation between innate and adaptive responses affiliated with divergent Th1/Th2 profiles and negative outcomes. This knowledge forms the basis of clinical triage that may be used to preemptively detect high-risk patients before life-threatening outcomes ensue. Highlights – Mild COVID-19 patients presented an early compromise with NK cell function, whereas severe patients do so with neutrophil function. – The identified co-expressed genes give insights into a coordinated transcriptional program of NK cell cytotoxic activity being associated with mild patients. – Key checkpoints of NK cell cytotoxicity that were enriched in mild patients include: KLRD1 , CD247 , and IFNG . – The early innate immune response related to NK cells connects with the Th1/Th2 adaptive immune responses, supporting their relevance in COVID-19 progression.
Article
Full-text available
Viral infections trigger inflammation by controlling ATP release. CD39 ectoenzymes hydrolyze ATP/ADP to AMP, which is converted by CD73 into anti-inflammatory adenosine (ADO). ADO is an anti-inflammatory and immunosuppressant molecule which can enhance viral persistence and severity. The CD39-CD73-adenosine axis contributes to the immunosuppressive T-reg microenvironment and may affect COVID-19 disease progression. Here, we investigated the link between CD39 expression, mostly on T-regs, and levels of CD73, adenosine, and adenosine receptors with COVID-19 severity and progression. Our study included 73 hospitalized COVID-19 patients, of which 33 were moderately affected and 40 suffered from severe infection. A flow cytometric analysis was used to analyze the frequency of T-regulatory cells (T-regs), CD39+ T-regs, and CD39+CD4+ T-cells. Plasma concentrations of adenosine, IL-10, and TGF-β were quantified via an ELISA. An RT-qPCR was used to analyze the gene expression of CD73 and adenosine receptors (A1, A2A, A2B, and A3). T-reg cells were higher in COVID-19 patients compared to healthy controls (7.4 ± 0.79 vs. 2.4 ± 0.28; p < 0.0001). Patients also had a higher frequency of the CD39+ T-reg subset. In addition, patients who suffered from a severe form of the disease had higher CD39+ T-regs compared with moderately infected patients. CD39+CD4+ T cells were increased in patients compared to the control group. An analysis of serum adenosine levels showed a marked decrease in their levels in patients, particularly those suffering from severe illness. However, this was paralleled with a marked decline in the expression levels of CD73. IL-10 and TGF-β levels were higher in COVID-19; in addition, their values were also higher in the severe group. In conclusion, there are distinct immunological alterations in CD39+ lymphocyte subsets and a dysregulation in the adenosine signaling pathway in COVID-19 patients which may contribute to immune dysfunction and disease progression. Understanding these immunological alterations in the different immune cell subsets and adenosine signaling provides valuable insights into the pathogenesis of the disease and may contribute to the development of novel therapeutic approaches targeting specific immune mechanisms.
Article
Full-text available
A recent outbreak of pneumonia in Wuhan, China was found to be caused by a 2019 novel coronavirus (2019-nCoV or SARS-CoV-2 or HCoV-19). We previously reported the clinical features of 12 patients with 2019-nCoV infections in Shenzhen, China. To further understand the pathogenesis of COVID-19 and find better ways to monitor and treat the disease caused by 2019-nCoV, we measured the levels of 48 cytokines in the blood plasma of those 12 COVID-19 patients. Thirty-eight out of the 48 measured cytokines in the plasma of 2019-nCoV-infected patients were significantly elevated compared to healthy individuals. Seventeen cytokines were linked to 2019-nCoV loads. Fifteen cytokines, namely M-CSF, IL-10, IFN-α2, IL-17, IL-4, IP-10, IL-7, IL-1ra, G-CSF, IL-12, IFN-γ, IL-1α, IL-2, HGF and PDGF-BB, were strongly associated with the lung-injury Murray score and could be used to predict the disease severity of 2019-nCoV infections by calculating the area under the curve of the receiver-operating characteristics. Our results suggest that 2019-nCoV infections trigger extensive changes in a wide array of cytokines, some of which could be potential biomarkers of disease severity of 2019-nCoV infections. These findings will likely improve our understanding of the immunopathologic mechanisms of this emerging disease. Our results also suggest that modulators of cytokine responses may play a therapeutic role in combating the disease once the functions of these elevated cytokines have been characterized.
Article
Full-text available
The ongoing outbreak of the recently emerged 2019 novel coronavirus (nCoV), which has seriously threatened global health security, is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) with high morbidity and mortality. Despite the burden of the disease worldwide, still, no licensed vaccine or any specific drug against 2019-nCoV is available. Data from several countries show that few repurposed drugs using existing antiviral drugs have not (so far) been satisfactory and more recently were proven to be even highly toxic. These findings underline an urgent need for preventative and therapeutic interventions designed to target specific aspects of 2019-nCoV. Again the major factor in this urgency is that the process of data acquisition by physical experiment is time-consuming and expensive to obtain. Scientific simulations and more in-depth data analysis permit to validate or refute drug repurposing opportunities predicted via target similarity profiling to speed up the development of a new more effective anti-2019-nCoV therapy especially where in vitro and/or in vivo data are not yet available. In addition, several research programs are being developed, aiming at the exploration of vaccines to prevent and treat the 2019-nCoV. Computational-based technology has given us the tools to explore and identify potentially effective drug and/or vaccine candidates which can effectively shorten the time and reduce the operating cost. The aim of the present review is to address the available information on molecular determinants in disease pathobiology modules and define the computational approaches employed in systematic drug re-positioning and vaccine development settings for SARS-CoV-2.
Article
Full-text available
At the end of December 2019, a novel acute respiratory syndrome coronavirus 2 (SARS‐CoV2) appeared as the third unheard of outbreak of human coronavirus infection in the 21st century. First, in Wuhan, China , the novel SARS‐CoV2 was named by the World Health Organization (WHO), as 2019‐nCOV (COVID‐19), and spread extremely all over the world. SARS‐CoV2 is transmitted to individuals by human‐to‐human transmission leading to severe viral pneumonia and respiratory system injury. SARS‐CoV2 elicits infections from the common cold to severe conditions accompanied by lung injury, acute respiratory distress syndrome, and other organ destruction. There is a possibility of virus transmission from asymptomatic cases as active carriers, in addition to symptomatic ones, which is a crucial crisis of COVID‐19 that should be considered. Hence, paying more attention to the accurate and immediate diagnosis of suspected and infected cases can be a great help in preventing the rapid spread of the virus, improving the disease prognosis, and controlling the pandemic. In this review, we provide a comprehensive and up‐to‐date overview of the different types of Clinical and Para‐clinical diagnostic methods and their practical features, which can help understand better the applications and capacities of various diagnostic approaches for COVID‐19 infected cases.
Article
Full-text available
We reported a 33-year-old female case with novel coronavirus disease 2019 (COVID-19) accompanied by Acute tubular necrosis (ATN). She had a gestational age of 34 weeks. The patient referred to treatment clinic for COVID-19 in Imam Reza hospital of Tabriz (Iran) after having flu-like symptoms. In radiologic assessment, ground glass opacity (GGO) with consolidation was found in upper right lobe. Lopinavir/ritonavir (200mg/50mg) two tablet tow times, Ribavirin 200mg every six hours, and Oseltamivir 75mg tow times were given for the treatment of COVID-19. The medications used for treatment of pneumonia were Meropenem, Ciprofloxacin, Vancomycin. All doses of medications were administrated by adjusted dose assuming the patient is anephric. Also, a few supplements were also given after ATN development including daily Rocaltrol and Nephrovit (as a multivitamin appropriate for patients with renal failure), Folic acid and Calcium carbonate. The patient is still under ventilator with a Fraction of inspired oxygen (FiO2) of 60% and Positive end-expiratory pressure (PEEP) of eight. SpO2 is 94% but the patient's ATN problem has been resolved. We started weaning from mechanical ventilator. The patient is conscious with full awareness to time, person and place. The maternal well-being is achieved and her neonate was discharged.
Article
Full-text available
Background Several studies have described the clinical characteristics of patients with novel coronavirus (SARS-CoV-2)-infected pneumonia (COVID-19), indicating severe patients tended to have higher neutrophil to lymphocyte ratio (NLR). Whether baseline NLR could be an independent predictor of in-hospital death in Chinese COVID-19 patients remains to be investigated. Methods A cohort of patients with COVID-19 admitted to the Zhongnan Hospital of Wuhan University from January 1 to February 29 was retrospectively analyzed. The baseline data of laboratory examinations, including NLR were collected. Univariate and multivariate logistic regression models were developed to assess the independent relationship between the baseline NLR and in-hospital all-cause death. A sensitivity analysis was performed by converting NLR from a continuous variable to a categorical variable according to tertile. Interaction and stratified analyses were conducted as well. Results 245 COVID-19 patients were included in the final analyses, and the in-hospital mortality was 13.47%. Multivariate analysis demonstrated that there was 8% higher risk of in-hospital mortality for each unit increase in NLR (Odds ratio [OR] = 1.08; 95% confidence interval [95% CI], 1.01 to 1.14; P = 0.0147). Compared with patients in the lowest tertile, the NLR of patients in the highest tertile had a 15.04-fold higher risk of death (OR = 16.04; 95% CI, 1.14 to 224.95; P = 0.0395) after adjustment for potential confounders. Notably, the fully adjusted OR for mortality was 1.10 in males for each unit increase of NLR (OR = 1.10; 95% CI, 1.02 to 1.19; P = 0.016). Conclusions NLR is an independent risk factor of the in-hospital mortality for COVID-19 patients especially for male. Assessment of NLR may help identify high risk individuals with COVID-19.
Article
Full-text available
Novel Coronavirus, COVID-19 discovered in December, 2019 in Wuhan, China started a world-wide epidemic. It is not clear till now what is the pathogenesis of this virus infection in human or the exact strategies of host immune response in combating this novel threat to human beings. However, morbidity and mortality of COVID-19 infections vary widely from asymptomatic, mild to deadly critical. Strangely, children were found to be protected from severe or deadly critical infections, while elderly and immunocompromised adults are most affected badly by this virus. It is necessary to disclose the possible viral and host interactions that lead to such variable morbid effects among patients of COVID-19 infections.
Article
Full-text available
Coronavirus disease 2019 (COVID-19) is an infectious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) with droplets and contact as the main means of transmission. Since the first case appeared in Wuhan, China, in December 2019, the outbreak has gradually spread nationwide. Up to now, according to official data released by the Chinese health commission, the number of newly diagnosed patients has been declining, and the epidemic is gradually being controlled. Although most patients have mild symptoms and good prognosis after infection, some patients developed severe and die from multiple organ complications. The pathogenesis of SARS-CoV-2 infection in humans remains unclear. Immune function is a strong defense against invasive pathogens and there is currently no specific antiviral drug against the virus. This article reviews the immunological changes of coronaviruses like SARS, MERS and other viral pneumonia similar to SARS-CoV-2. Combined with the published literature, the potential pathogenesis of COVID-19 is inferred, and the treatment recommendations for giving high-doses intravenous immunoglobulin and low-molecular-weight heparin anticoagulant therapy to severe type patients are proposed.
Article
Coronavirus disease 2019 (COVID‐19) is a respiratory disorder caused by the highly contagious SARS‐CoV‐2. The immunopathological characteristics of COVID‐19 patients, either systemic or local, have not been thoroughly studied. In the present study, we analyzed both the changes in the number of various immune cell types as well as cytokines important for immune reactions and inflammation. Our data indicate that patients with severe COVID‐19 exhibited an overall decline of lymphocytes including CD4+ and CD8+ T cells, B cells, and NK cells. The number of immunosuppressive regulatory T cells was moderately increased in patients with mild COVID‐19. IL‐6, IL‐10, and C‐reactive protein were remarkably up‐regulated in patients with severe COVID‐19. In conclusion, our study shows that the comprehensive decrease of lymphocytes, the elevation of IL‐6, IL‐10, and C‐reactive protein are reliable indicators of severe COVID‐19.
Article
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of the ongoing coronavirus disease 2019 (COVID-19) pandemic. Alongside investigations into the virology of SARS-CoV-2, understanding the fundamental physiological and immunological processes underlying the clinical manifestations of COVID-19 is vital for the identification and rational design of effective therapies. Here, we provide an overview of the pathophysiology of SARS-CoV-2 infection. We describe the interaction of SARS-CoV-2 with the immune system and the subsequent contribution of dysfunctional immune responses to disease progression. From nascent reports describing SARS-CoV-2, we make inferences on the basis of the parallel pathophysiological and immunological features of the other human coronaviruses targeting the lower respiratory tract — severe acute respiratory syndrome coronavirus (SARS-CoV) and Middle East respiratory syndrome coronavirus (MERS-CoV). Finally, we highlight the implications of these approaches for potential therapeutic interventions that target viral infection and/or immunoregulation. In the short time since SARS-CoV-2 infections emerged in humans, much has been learned about the immunological processes that underlie the clinical manifestation of COVID-19. Here, the authors provide an overview of the pathophysiology of SARS-CoV-2 infection and discuss potential therapeutic approaches.