You are on page 1of 7

Hindawi

BioMed Research International


Volume 2021, Article ID 6372128, 7 pages
https://doi.org/10.1155/2021/6372128

Research Article
N-Acetylcysteine Improves Inflammatory Response in COPD
Patients by Regulating Th17/Treg Balance through Hypoxia
Inducible Factor-1α Pathway

Xiaopeng Liu, Zhixiong Hu, and Haiying Zhou


Department of Respiratory Medicine, Jinshan Hospital, Fudan University, Shanghai 201508, China

Correspondence should be addressed to Haiying Zhou; mipanba234265682@163.com

Received 24 April 2021; Accepted 17 June 2021; Published 29 June 2021

Academic Editor: Tao Huang

Copyright © 2021 Xiaopeng Liu et al. This is an open access article distributed under the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Introduction. This study was aimed to investigate the effects of N-acetylcysteine (NAC) on chronic obstructive pulmonary disease
(COPD) and the change of Th17/Treg cytokine imbalance. Material and Methods. A total of 121 patients with stable COPD at the
stage of C or D were consecutively enrolled and randomly divided into 2 groups. Patients in the treatment group received NAC
granules (0:2 g × 10 bags, 0.4 g each time, 3 times/d) for half a year. The control group was treated with the same amount of
placebo therapy. The peripheral blood of the patient was collected and the cytokine, T lymphocyte subsets were detected.
Results. We found the oral administration of NAC could regulate Th17/Treg balance to resist inflammation in COPD patients.
Serum testing showed that the proportion of Treg in CD4+ T cells has increased and the Th17/Treg ratio has decreased during
the NAC treatment. In vitro studies, we found that NAC regulated Th17/Treg balance through Hypoxia Inducible Factor-1α
pathway. Conclusions. Our result could provide new diagnosis and treatment for elderly patients with COPD from the
perspective of immunity ideas.

1. Introduction thione [7]. Oral NAC has been shown to affect the body’s
redox balance through increasing both plasma glutathione
Chronic obstructive pulmonary disease (COPD) belongs to a levels and lung lavage glutathione levels in patients with sta-
class of common chronic respiratory diseases characterized ble COPD. However, the clinical effectiveness of NAC in
by irreversible continuous airflow limitation and high fatal- COPD treatment is still controversial. Although it was found
ity rate, which currently places a huge burden on the world’s in some early studies that NAC had no effect, more recent
disease economy [1]. It is associated with both airway and studies have shown that increasing the dose (1200 mg daily
extrapulmonary inflammation, as indicated by increased or above) of NAC can reduce the exacerbation of COPD
reactive oxygen species (ROS) levels and proinflammatory patients and extend the time to the first exacerbation [8–
markers in peripheral blood. COPD manifests as a series of 11]. And the most principle of related mechanism is for its
overlapping phenotypes, including chronic bronchitis, antioxidative effect, such as reducing ROS production by
emphysema, small airway disease, and frequent exacerba- alveolar macrophages as well as reducing exhaled H2O2.
tions. Despite differences in pathology, they share many There are limited data on the anti-inflammatory activity of
common but complex pathogenetic processes, including NAC in COPD patients.
inflammation, excessive oxidative stress, apoptosis, and In recent years, lymphocytes, especially CD4+ T cells
autoimmunity [2–6]. involved in the pathogenesis of COPD, have become an
N-acetylcysteine (NAC) belongs to a class of antioxi- active topic of research [3, 5, 6, 12]. Studies have revealed that
dants, which has both antioxidant and anti-inflammatory the imbalance between T helper (Th)17 cells and regulatory
properties. NAC exerts its antioxidant effect by acting T (Treg) cells played a vital role in the development and out-
directly as a ROS scavenger and a precursor of reduced gluta- come of COPD [5, 12]. Interestingly, though Th17 and Treg
2 BioMed Research International

cells shared a common requirement for TGF-β in their differ- Table 1: Comparison of baseline data between the two groups.
entiation, they express distinct transcriptional regulators
(RORγt versus Foxp3, respectively) and present opposing Control group Treatment group P
Term
functions (inflammatory versus anti-inflammatory). Recent (N = 61) (N = 60) value
studies have shown that NAC could play as a regulatory role Gender
28 30 0.269
on T lymphocyte subsets differentiation and improve the (male)
patient’s immune function [13–16]. However, the effect of Age 72:65 ± 7:96 70:89 ± 8:62 0.116
NAC for T lymphocyte subtype transformation on patients (mean ± SD)
with stable COPD is still unknown. Smoking
569:07 ± 264:32 592:76 ± 228:54 0.305
In this study, we explored the treatment effect of oral index
NAC on elderly patients with COPD in clinic. And we also mMRC score 2:62 ± 0:85 2:69 ± 0:91 0.277
determined the change of Th17/Treg balance in peripheral
blood of patients who accepted NAC, in order to provide
new diagnosis and treatment for elderly patients with COPD or mMRC score of 2 or more, FEV1/pred% <50% and/or
from the perspective of immunity ideas. acute exacerbations ≥2 times per year, or the number of hos-
pitalizations for acute exacerbations is once or more.
2. Materials and Methods
2.4. Inclusion and Exclusion Criteria for the Study. The inclu-
2.1. Ethics Statement. This research was approved by the sion criteria were as follows: (1) patients confirmed as having
research ethics committees of Jinshan Hospital Affiliated to COPD at stage of C and D; (2) patients who received no anti-
Fudan University, China (approval number: Jinshan Medical biotics, glucocorticoids, and theophylline medications within
Ethics Research 2019-13-02). All patients were approached 2 weeks before entering the study; and (3) patients who
in accordance with the approved ethical guidelines. They signed informed consent forms for voluntary participation
had agreed before participating in this study and signed in the study. The exclusion criteria were as follows: (1)
informed consent. All methods used in the study were per- patients with acute exacerbations which may affect the natu-
formed in accordance with the relevant guidelines and regu- ral process of IL-17 and IL-10 in COPD; (2) patients accom-
lations developed by the aforementioned ethics committees. panied by pulmonary interstitial fibrosis, tuberculosis,
bronchial pneumonia, and lung cancer; (3) patients with
2.2. Participants and Trial Design. A total of 121 patients with major diseases of nonrespiratory system, such as diabetes,
stable COPD at the stage of C or D based on the Global Ini- severe cardiovascular and cerebrovascular diseases, and neu-
tiative for Chronic Obstructive Lung Disease randomly rological diseases or liver and kidney dysfunction; and (4)
selected from September 2018 to June 2019 were recruited patients with mental illness and cognitive impairment.
in this research (Jinshan Hospital Affiliated to Fudan Univer-
sity, China). They were randomly divided into two groups. 2.5. Blood Samples and Measurements. The peripheral
Patients in the treatment group were treated with conven- venous blood of the patient was collected in tubes containing
tional inhalation of bronchodilators (long-acting anticholin- ethylene diamine tetraacetate acid (EDTA) in the early
ergic drugs (LAMA, tiotropium, 18 μg daily) and oral NAC morning. The blood samples were immediately centrifuged
granules (0:2 g × 10 bags, 0.4 g each time, 3 times/d, Guang- at 3,000 rpm for 15 minutes after collected, and serum was
dong Baiao Pharmaceutical) for half a year. The control obtained and stored at −80°C until used. The serum levels
group was treated with conventional airway expansion ther- of IL-17, IL-10, IL-9, TGF-β, TNF-a, and IL-1β were deter-
apy and the same amount of placebo therapy. No other mined by using ELISA test kits (R&D Systems, Minneapolis,
phlegm-resolving drugs, glucocorticoids, bronchodilators MN), following the manufacturer’s instructions. Briefly, the
were used during the study. Lung function of each patient antibody was diluted to a content of 5 μg/mL, and then, 0.1
was evaluated at the beginning of the study. No significant mL of that was added into the plate hole and placed at 4°C
differences were observed in terms of age, gender, and smok- overnight. The next day, the solution in the plate was discard-
ing status of patients between the treatment and control indi- ing, and a 0.1 mL aliquot of the sample was added into the
viduals (P > 0:05). Clinical data of all individuals are given in reaction well. Then, place the reaction well into a humid
detail in Table 1. chamber at 37°C for 1 hour. Then, discarding the solution
and 0.1 mL of freshly diluted enzyme-labeled antibody was
2.3. Diagnosis of COPD. Diagnosis of COPD was confirmed added at 37°C for 1 hour. Then, a 0.1 mL aliquot of substrate
in accordance with the diagnostic criteria of the Global initia- solution was added to each well at 37°C for 30 minutes, and
tive for chronic Obstructive Lung Disease. The diagnostic cri- 0.05 mL of stop solution was added to each reaction well.
teria for group C were as follows: CAT (COPD assessment The color intensity was measured at a wavelength of 450
test) score <10 or mMRC (modified British Medical Research nm by using a photometer. The standard curve was plotted
Council) score was between 0 and 1, the FEV1/pred% is less based on the concentration of the standard sample and the
than 50%, and/or acute exacerbations occur more than 2 OD of each well.
times in one year or at least one acute exacerbation occurs
which is required in hospital treatment. The diagnostic cri- 2.6. Inflammation-Pathway Microarray. Expression microar-
teria for group D were as follows: CAT score of 10 or more ray analysis was conducted with HWayen inflammation-
BioMed Research International 3

pathway microarray CSP100. All the procedures and data with oral NAC, compared to the control counterparts. Con-
analysis were performed by HWayan, China. versely, the serum level of IL-10 was upregulated by oral
NAC treatment in the COPD patients, compared to the con-
2.7. Flow Cytometry Analysis. Peripheral blood mononuclear trol counterparts (Figure 1). Meanwhile, other proinflamma-
cells (PBMCs) were freshly isolated by Ficoll density gradient tion cytokines, including IL-9, TGF-β, TNF-a, and IL-1β,
centrifugation (Pharmacia, Uppsala, Sweden). The isolated were all downregulated. These data demonstrated that oral
PBMCs were washed twice with PBS and resuspended at NAC treatment at a dose of 1200 mg/day was helpful for
106 cells/mL in complete culture medium (RPMI 1640 sup- improving the inflammatory status of COPD patients.
plemented with 1% penicillin/streptomycin, 2 mM L-gluta-
mine, and 10% heat-inactivated fetal bovine serum; Gibco 3.2. Oral NAC suppressed circulating Th17 cells while
BRL, Gaithersburg, MD, USA). For Th17 cells analysis, cells promoted Treg cells in the serum of COPD patients. To further
were detected as described previously. For the analysis of explore the effect of oral NAC, we used IL-17 and Foxp3 as
Treg cells, a human regulatory T-cell staining kit (BD Phar- the biomarker of Th17 and Treg cells in the flow cytometry
mingen™, USA) was used to measure CD4+ CD25+ Foxp3 analysis, respectively. As shown in Figures 2(a) and 2(b),
+ cells as Treg cells according to the manufacturer’s protocol. the frequencies of Th17 cells were significantly decreased in
Briefly, PBMCs were incubated with a cocktail of fluorescein the peripheral blood of COPD patients in the NAC treatment
isothiocyanate-conjugated anti-CD4 and APC-conjugated group than those in the control group (P < 0:05). On the con-
anti-CD25 for 30 minutes at 4°C. After fixation and perme- trary, the frequencies of Treg cells were increased accordingly
abilization, the cells were blocked by normal rat serum and (Figures 2(c) and 2(d)).
stained using PE-conjugated anti-Foxp3 for 45 minutes at
4°C. The flow cytometry analyses were performed on a FACS 3.3. Oral NAC rebalances the Th17/Treg ratio. Constantly, we
Calibur flow cytometer (BD biosciences, San Jose, CA, USA) found that the Th17/Treg ratio was evidently decreased after
equipped with the CellQuest software (BD biosciences). Iso- taking oral NAC in the treatment group from 5.7 to 0.28.
type controls were conducted to ensure antibody specificity. This result further proved that NAC could protect patients
with COPD by suppressing Th17 immune responses and
2.8. Western Blot Analysis. The cells were lysed in lysis buffer modulating the Th17/Treg balance in favor of Treg cells.
containing 4% sodium dodecyl sulfate (SDS), 20% glycerol, Interestingly, we found that Th17 cells had a negative corre-
100 mM dithiothreitol (DTT), and Tris-HCl, pH 6.8. A total lation with Treg cells whether COPD patients took NAC or
of 30 μg of the supernatant proteins was loaded and sepa- not (Figure 3).
rated by 8-15% sodium dodecyl sulfate-polyacrylamide gel
electrophoresis. The samples were incubated with primary 3.4. NAC decreased HIF1-α expression of T cells isolated from
antibodies overnight at 4°C after the proteins were trans- peripheral blood of COPD patients. Studies have shown that
ferred to a polyvinylidene fluoride (PVDF) membrane (Milli- NAC can downregulate the protein of HIF-α by posttran-
pore, 0.2 μm or 0.45 μm, as appropriate), following the scription mechanism [22, 23]. And specifically, HIF1-α is
electrophoresis [17]. The primary antibodies were used at considered as a key regulatory molecule that could directly
dilutions of 1 : 1000 for GAPDH (Cell Signaling), 1 : 1000 promote Th17 differentiation by inducing RORγt transcrip-
for HIF1-α (Cell Signaling). Then, the membranes were incu- tion and inhibit Treg differentiation through facilitating
bated with HRP-conjugated secondary antibodies and visual- Foxp3 protein degradation [24]. So, we conducted an
ized with an ECL Chemiluminescent kit (NovexTM). inflammation-pathway array to further analyze the pathway
related to the change (n = 10), and we found that HIF1-a
2.9. Statistical Analysis. All analyses were done by using the and MAPKp38 exhibited the greatest fold changes in Th17
SPSS software, version 20.0 (IBM Corp., Armonk, NY, and Treg cells. Next, we detected the protein and RNA
USA). Continuous variables were reported as mean ± SD change of HIF1-α and p38 expression in Th17 and Treg cells
[18]. For two-group comparisons, the independent sample t after NAC treatment in COPD patients (n = 121). As shown
-test was used for normally distributed data [19]. All tests in Figure 4, in the treatment group, western blot revealed that
were two-tailed, and a value of P < 0:05 was considered sta- HIF1-α significantly decreased in both cells, while p38 had
tistically significant. no significant change. The result might reveal that NAC
improves inflammatory response in COPD patients by regu-
3. Results lating Th17/Treg balance through the HIF1-α pathway.

3.1. Oral NAC downregulated the level of IL-17 while 4. Discussion


upregulated the level of IL-10 in the serum of patients with
COPD. Previous studies demonstrated that increased IL-17 COPD is a type of high-incidence chronic disease in the
level and decreased IL-10 level in serum were positively cor- respiratory system. The occurrence and development of the
related with the progression of COPD [20, 21]. To determine disease have not been completely studied [1]. In recent years,
whether oral NAC regulated the level of IL-17, IL-10, and immune system disorders have been closely related to the
other related inflammation cytokine in the serum, we cause of the disease. Many researchers have conducted in-
obtained peripheral blood from the patient with COPD for depth research in this area. Relevant data showed that, in
further evaluation. As shown in Figure 1, the level of IL-17 addition to Th1 and Th2 cells that participated in immune-
was downregulated in the serum of COPD patients treated pathological inflammation, Th17 cells and Treg cells, as
4 BioMed Research International

100 50 ⁎⁎ 60

80

TGF-𝛽 expression
40
in serum (pg/ml)

in serum (pg/ml)
in serum (pg/ml)
IL-17 expression

IL-10 expression
40
60 30
⁎⁎
40 20
20
20 10

0 0 0
Before After Before After Before After
30 15 150

TNF-a expression
in serum (pg/ml)

in serum (pg/ml)
in serum (pg/ml)
IL-𝛽 expression

IL-9 expression
20 10 100
⁎⁎ ⁎⁎ ⁎⁎
10 5 50

0 0 0
Before After Before After Before After
Control
Treatment

Figure 1: Oral NAC decreases the level of IL-17 and increases IL-10 in the serum of COPD patients. The level of serum IL-17 in the treatment
group increased after oral administration of NAC. The level of serum IL-10 increased in the treatment group after oral administration of
NAC. (∗ P < 0:05; ∗∗ P < 0:01; ∗∗∗ P < 0:001).

Control NAC
250K 250K 25

200K 200K 20
TH17/CD4+cells (%)

150K 150K 15
SSC-A

SSC-A

18.5 2.1
100K 100K 10

50K 50K 5 ⁎⁎⁎

0 0 0
0 102 103 104 105 0 102 103 104 105 Control NAC

IL 17 IL 17
(a) (b)
250K 250K 25 ⁎⁎⁎

200K 200K 20
Treg /CD4+cells (%)

150K 150K 15
SSC-A

SSC-A

100K 3.5 100K 20.5 10

50K 50K 5

0 0 0
0 102 103 104 105 0 102 103 104 105 Control NAC

Foxp3 Foxp3
(c) (d)

Figure 2: TH17 cell decreased while Treg increased after NAC treatment in the COPD patients. PBMC isolated from peripheral blood and
were stained for IL-17 and Foxp3 in order to measure the percentage of Th17 and Treg. (a, b) The percentage of Th17 cells significantly
decreased after oral administration of NAC. (c, d) The percentage of Treg cells significantly decreased after oral administration of NAC
(∗∗∗ P < 0:01).
BioMed Research International 5

6.0
Control
6 5.5

Treg/CD4+cells (%)
TH17/Treg
5.0
4
4.5
2
4.0
⁎⁎⁎
0 3.5
Control NAC 15 20 25 30

Th17/CD4+cells (%)
30
NAC

Treg/CD4+cells (%)
25

20

15
4.0 4.5 5.0 5.5 6.0

Th17/CD4+cells (%)
(a) (b)

Figure 3: Oral NAC rebalance Th17/Treg ratio in the COPD patients. (a) The Th17/Treg ratio decreased significantly after oral
administration of NAC compared with the control group. (b) Th17 cells had the negative correlation with Treg cells. (∗ P < 0:05; ∗∗ P <
0:01; ∗∗∗ P < 0:001).

Th17 cells
2.0


1.5
Control NAC
Relative expression

1.5 1.5
HIF-1 relative expression

p38 relative expression

p-p38
1.0
⁎ 1.0 1.0
p38
⁎⁎ ⁎⁎⁎
0.5
0.5 0.5
HIF1𝛼

0.0 0.0 0.0


GAPDH
TGF-𝛽
ATF

Control
NF-kB

cAMP/PKA
IL-6

MAPK/P38

MAPK/JNK

MAPK/ERK

PI3K/AKT
HIF

Control NAC NAC

Treg cells
2.0

⁎ Control NAC
1.5
Relative expression

1.5 1.5
HIF-1 relative expression

p-p38
p38 relative expression

1.0 1.0 1.0


p38

0.5 HIF1𝛼 0.5 0.5


⁎⁎ ⁎⁎⁎

0.0 GAPDH 0.0 0.0


Control NAC Control NAC
TGF-𝛽
ATF

NF-kB

cAMP/PKA
IL-6

MAPK/P38

MAPK/JNK

MAPK/ERK

PI3K/AKT
HIF

Control
NAC

Figure 4: NAC significantly decreased HIF1-α expression of Th17 and Treg cells isolated from peripheral blood of COPD patients.
Inflammation-pathway array, western-blot, and qRT-PCR revealed the decreased expression of HIF1-α in Th17 and Treg cells in the NAC
control group. (∗ P < 0:05; ∗∗ P < 0:01; ∗∗∗ P < 0:001).
6 BioMed Research International

subgroups of CD4+ T cells, also played an important role in Data Availability


balancing COPD patients’ immune status [5, 20]. Previous
researches showed that during the development and acute All data in this article are available from the corresponding
exacerbation period in patients with COPD, the ratio of Treg author upon reasonable request.
and Th17 cells in the peripheral blood has changed signifi-
cantly, as well as in bronchoalveolar lavage (BAL). The Conflicts of Interest
Th17 cells in the serum and Th17/Treg ratio showed a rising
trend, but Treg cells were constantly falling. The normal The authors declare no conflict of interest.
Th17/Treg balance was broken, which led to the appearance
of an inflammatory response. At the same time, the increase Authors’ Contributions
in Th17 cell level was negatively correlated with the decrease
Xiaopeng Liu and Haiying Zhou conceived and designed the
in Treg level, suggesting that the imbalance between these
experiments. Xiaopeng Liu performed the research, con-
two subgroups might be one of the causes of COPD [21,
ducted the data analyses, and wrote the manuscript. Xiao-
25]. Therefore, proper intervention in the Th17/Treg balance
peng Liu and Zhixiong Hu contributed to the clinical data
of COPD patients was important for the treatment of COPD.
collection. Haiying Zhou revised the manuscript and coordi-
Previous researches have confirmed that NAC had the
nated the research team. All authors have read and approved
capability to inhibit the accumulation of neutrophils in the
the final manuscript.
lungs of COPD patients, reducing inflammation, and had a
strong expectorant effect [7, 9, 10]. At the same time, NAC
had a regulatory effect on immune function and could Acknowledgments
weaken cytokine activity. Some recent studies had shown This work was supported by Scientific Research Youth Pro-
that oral NAC could regulate the Th1/Th2 ratio balance, ject of Jinshan District Health Commission of Shanghai city
improving the quality of life of COPD patients in groups C (JSKJ-KTQN-2018-04)-LiuXiaoPeng, Hospital-level project
and D, increasing lung function to a large extent [26]. Our of Jinshan Hospital Affiliated to Fudan University (2018-
study showed that the drug could increase the serum JSYYKT-01)-LiuXiaoPeng, and Scientific Research Youth
interleukin-10 level after oral NAC at a dosage of 1200 mg Project of Shanghai Municipal Health Commission
daily in the treatment group, while the proinflammation (20204Y0172)-LiuXiaoPeng. The authors thank the patients
cytokine IL-9, TGF-β, TNF-a, IL-1β, and IL-17 were all who participated in this research and Mitchell Arico from
downregulated dramatically. Moreover, the proportion of Liwen Bianji, Edanz Group China (http://www.liwenbianji
Th17 in CD4+ T cells was decreased, and the proportion of .cn/ac), for editing the English text of a draft of this
Treg in CD4+ T cells was increased. Therefore, the manuscript.
Th17/Treg ratio was reduced compared with that before
diagnosis and treatment. The results suggested that after
References
NAC treatment, the Th17 immune response decreased, the
Treg immune response increased, and Th17/Treg tended to [1] A. I. Ritchie and J. A. Wedzicha, “Definition, causes, pathogen-
back to a new balance, the immune status of COPD patients esis, and consequences of chronic obstructive pulmonary dis-
return to normal. To figure out the mechanism of this effect ease exacerbations,” Clinics in Chest Medicine, vol. 41, no. 3,
of NAC, we used inflammation-pathway microarray, western pp. 421–438, 2020.
blot, and qRT-PCR to verified that HIF1-α might be a key [2] F. Decrue, O. Gorlanova, J. Usemann, and U. Frey, “Lung
metabolic sensor. In this study, we found HIF-1a was truly functional development and asthma trajectories,” Seminars
significantly downregulated after treatment. Previous studies in Immunopathology, vol. 42, no. 1, pp. 17–27, 2020.
reported that HIF-1α could upregulate IL-17 expression [3] W. W. Labaki, L. M. Kimmig, G. M. Mutlu, M. L. K. Han, and
through RORγt; meanwhile, it also cooperated with RORγt S. P. Bhatt, “Update in chronic obstructive pulmonary disease
protein to regulate IL-17A-related genes during Th17 devel- 2018,” American Journal of Respiratory & Critical Care Medi-
opment, thus enhanced the development of Th17 [13, 22]. cine, vol. 199, no. 12, pp. 1462–1470, 2019.
On the other hand, reports showed that HIF-1 targeted [4] S. M. Chan, S. Selemidis, S. Bozinovski, and R. Vlahos, “Patho-
Foxp3 for ubiquitination and proteasomal degradation, using biological mechanisms underlying metabolic syndrome
(MetS) in chronic obstructive pulmonary disease (COPD):
the same ubiquitin ligase system that was responsible for the
clinical significance and therapeutic strategies,” Pharmacology
degradation of HIF-1α itself [24]. So, based on the experi- & Therapeutics, vol. 198, pp. 160–188, 2019.
mental data and result, we proposed a hypothesis that NAC
[5] Y. C. Su, F. Jalalvand, J. Thegerström, and K. Riesbeck, “The
could rebalance the Th17/Treg ratio and improve the interplay between immune response and bacterial infection
immune status of COPD patients through downregulating in COPD: focus upon non-typeable Haemophilus influenzae,”
HIF1-a. Frontiers in Immunology, vol. 9, p. 2530, 2018.
In summary, this study reveals that NAC could affect the [6] L. Wen, S. Krauss-Etschmann, F. Petersen, and X. Yu, “Auto-
Th17/Treg balance in the COPD patients and had a good antibodies in chronic obstructive pulmonary disease,” Fron-
therapeutic effect for the immune status of COPD patients tiers in Immunology, vol. 9, p. 66, 2018.
which might be through regulating the HIF1-α expression. [7] M. G. Matera, L. Calzetta, and M. Cazzola, “Oxidation path-
By studying the Th17/Treg balance, possible intervention way and exacerbations in COPD: the role of NAC,” Expert
targets may be further found and developed. Review of Respiratory Medicine, vol. 10, no. 1, pp. 89–97, 2016.
BioMed Research International 7

[8] S. Moitra, “N-Acetylcysteine (NAC) in COPD: Benefits Often in stroke,” Free Radical Biology and Medicine, vol. 68, pp. 8–
Lost in Trials,” QJM: An International Journal of Medicine, 21, 2014.
vol. 112, no. 5, pp. 387-388, 2019. [24] E. V. Dang, J. Barbi, H. Y. Yang et al., “Control of TH17/Treg
[9] V. T. Salve and J. S. Atram, “N-acetylcysteine combined with balance by hypoxia-inducible factor 1,” Cell, vol. 146, no. 5,
home based physical activity: effect on health related quality pp. 772–784, 2011.
of life in stable COPD patients-a randomised controlled trial,” [25] M. Zhang, Y. Wan, Y. Jin et al., “Cigarette smoking promotes
Journal of Clinical and Diagnostic Research, vol. 10, no. 12, pp. - inflammation in patients with COPD by affecting the polariza-
OC16–OC19, 2016. tion and survival of Th/Tregs through up-regulation of musca-
[10] C. M. Sanguinetti, “N-Acetylcysteine in COPD: Why, How, rinic receptor 3 and 5 expression,” PLoS One, vol. 9, no. 11,
and When?,” Multidisciplinary Respiratory Medicine, vol. 11, article e112350, 2014.
no. 1, article 8, 2015. [26] J. Zhang, J. Zhang, H. Liu et al., “Effect of N-acetylcysteine in
[11] Q. Zhang, Y. Ju, Y. Ma, and T. Wang, “N-acetylcysteine COPD patients with different microsomal epoxide hydrolase
improves oxidative stress and inflammatory response in genotypes,” International Journal of Chronic Obstructive Pul-
patients with community acquired pneumonia: A randomized monary Disease, vol. 10, no. 1, pp. 917–923, 2015.
controlled trial,” Medicine, vol. 97, no. 45, article e13087, 2018.
[12] F. Polverino, L. J. M. Seys, K. R. Bracke, and C. A. Owen, “B
cells in chronic obstructive pulmonary disease: moving to cen-
ter stage,” American Journal of Physiology-Lung Cellular and
Molecular Physiology, vol. 311, no. 4, pp. L687–L695, 2016.
[13] J. Barbi, D. M. Pardoll, and F. Pan, “Metabolic control of the
Treg/Th17 axis,” Immunological Reviews, vol. 252, no. 1,
pp. 52–77, 2013.
[14] D. C. Newcomb and R. S. Peebles, “Th17-mediated inflamma-
tion in asthma,” Current Opinion in Immunology, vol. 25,
no. 6, pp. 755–760, 2013.
[15] T. E. Kononova, O. I. Urazova, V. V. Novitskii et al., “Func-
tional activity of Th-17 lymphocytes in pulmonary tuberculo-
sis,” Bulletin of Experimental Biology and Medicine, vol. 156,
no. 6, pp. 743–745, 2014.
[16] M. Noack and P. Miossec, “Th17 and regulatory T cell balance
in autoimmune and inflammatory diseases,” Autoimmunity
Reviews, vol. 13, no. 6, pp. 668–677, 2014.
[17] C. Gu, Z. Huang, X. Chen et al., “TEAD4 promotes tumor
development in patients with lung adenocarcinoma via ERK
signaling pathway,” Biochimica et Biophysica Acta - Molecular
Basis of Disease, vol. 1866, no. 12, article 165921, 2020.
[18] J. Chen, C. Gu, X. Chen et al., “Clinicopathological and prog-
nostic analyses of 86 resected pulmonary
lymphoepithelioma-like carcinomas,” Journal of Surgical
Oncology, vol. 123, no. 2, pp. 544–552, 2021.
[19] C. Chen, X. Zhang, C. Gu et al., “Surgery performed at night by
continuously working surgeons contributes to a higher inci-
dence of intraoperative complications in video-assisted thora-
coscopic pulmonary resection: a large monocentric
retrospective study,” European Journal of Cardio-Thoracic
Surgery, vol. 57, no. 3, pp. 447–454, 2020.
[20] M. I. Vargas-Rojas, A. Ramírez-Venegas, L. Limón-Camacho,
L. Ochoa, R. Hernández-Zenteno, and R. H. Sansores,
“Increase of Th17 cells in peripheral blood of patients with
chronic obstructive pulmonary disease,” Respiratory Medicine,
vol. 105, no. 11, pp. 1648–1654, 2011.
[21] X. Li, X. Pan, and D. Qiu, “Imbalances of Th17 and Treg cells
and their respective cytokines in COPD patients by disease
stage,” International Journal of Clinical and Experimental
Medicine, vol. 7, no. 12, pp. 5324–5329, 2014.
[22] T. Suha, M. Asli, S. Aynur et al., “Effects of N-acetylcysteine
and ethyl pyruvate on ischemia-reperfusion injury in experi-
mental electrical burn model,” American Journal of Emergency
Medicine, vol. 34, no. 7, pp. 1217–1224, 2016.
[23] Z. Zhang, J. Yan, S. Taheri, K. J. Liu, and H. Shi, “Hypoxia-
inducible factor 1 contributes to N-acetylcysteine’s protection

You might also like