You are on page 1of 7

Gene 678 (2018) 177–183

Contents lists available at ScienceDirect

Gene
journal homepage: www.elsevier.com/locate/gene

Review

Oxidative stress and bronchopulmonary dysplasia☆ T



Junyi Wang, Wenbin Dong
Department of Newborn Medicine, The Affiliated Hospital of Southwest Medical University, 25 Taiping Road, Luzhou, Sichuan 646000, People's Republic of China

A R T I C LE I N FO A B S T R A C T

Keywords: With the progress of modern medicine, oxygen therapy has become a crucial measure for the treatment of
Neonatal premature infants. As an environmental stimulus, in the normal development of lungs, oxygen plays a very
BPD important regulatory role. However, the problem is that long-term exposure to hyperoxia can interfere with the
Oxidative stress development of lungs, leading to irreversible developmental abnormalities. Now, the incidence of broncho-
pulmonary dysplasia (BPD) is increasing year by year. The existing related research shows that although BPD is a
multi-factor triggered disease, its main risk factors are the premature exposure to hyperoxia and the role of
reactive oxygen species (ROS). As for premature infants, especially very premature babies and those with very
low birth weight, prolonged exposure to high oxygen can affect and alter the normal developmental trajectories
of lung tissue and vascular beds, triggering developmental disorders, such as BPD. In the relevant studies about
human BPD, a large number of them support that ROS is associated with impaired lung development. Neonates,
due to the damage in the development of alveolar, are specific to hyperoxia-induced inflammatory damage. This
review while focusing on the role of oxidative stress in the pathogenesis of BPD, suggests that antioxidant
measures may be effective to guard against BPD of preterm infants.

1. Introduction hypertension in preterm infants with BPD is also relatively high.


Although many pharmacological and non-pharmacological methods
Nowadays, thanks to the widespread use of oxygen therapy, the have been proposed, there is no clear protocol for limiting or treating
survival rate of premature infants is increasing. As an environmental BPD. Therefore, to explore the pathogenesis of BPD is an important
stimulus, in the normal development of lungs, oxygen plays a very guide for early prevention. The purpose of this review is to summarize
important regulatory role, which may be related to the interaction the research progress in the role of oxidative stress during the devel-
between oxygen and cells. However, the problem is that long-term opment of BPD, which proposes that anti-oxidative stress may be a key
exposure to hyperoxia can interfere with the development of lungs, link in the prevention and treatment of BPD.
leading to irreversible developmental abnormalities (Buczynski et al.,
2013; Saugstad et al., 2011). 2. BPD
Now, bronchopulmonary dysplasia (BPD) incidence is rising year by
year. Current research shows that (Carraro et al., 2013; O'Reilly and 2.1. The definition of BPD
Thébaud, 2013; Kotecha et al., 2013) although BPD is a multi-factor
disease, including premature lung development immature, infection, Northern (Jr and Rosan, 1968) first reported BPD in 1967, defining
nutrition, oxygen poisoning, volumetric injury during mechanical the typical BPD as a chronic lung injury caused by severe respiratory
ventilation, barotrauma and inflammatory factor response, etc., its distress syndrome. In the same year, BPD was reported to be closely
main risk factor is due to the exposure to hyperoxia and the role of related to hyperoxia (Northway Jr and R., 1967). With the widespread
reactive oxygen species (ROS) on premature infants. Previous studies use of new treatments such as exogenous surfactants, mild ventilation
have shown that children with BPD have a poor prognosis. They often strategies and prenatal steroids, the description of traditional BPD has
suffer repeated respiratory infections and re-hospitalization. In addi- been changed. A typical BPD characterized by wide-ranging and serious
tion, they would suffer gradual decline of lung function with their age inflammation in lung tissue and extensive fibrosis in various respiratory
increasing. The further risk of retinopathy, learning disabilities and tracts has slowly evolved into a new type of BPD: NICHD redefined BPD


This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.

Corresponding author.
E-mail address: dongwenbin2000@163.com (W. Dong).

https://doi.org/10.1016/j.gene.2018.08.031
Received 10 April 2018; Received in revised form 30 July 2018; Accepted 6 August 2018
Available online 09 August 2018
0378-1119/ © 2018 The Authors. Published by Elsevier B.V. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/BY-NC-ND/4.0/).
J. Wang, W. Dong Gene 678 (2018) 177–183

and its indexing standards in 2000. In terms of the “new BPD” char- concentration in the atmosphere at birth is also relatively high in
acterized by alveolar and pulmonary microvascular dysplasia, there neonatal exposure. The relevant studies have shown that anti-oxidative
was no change in chest radiographs used as a basis for assessing BPD enzyme activity in the pre-natal level is significantly higher than in
severity (Pesce and Musi, 1996). Any newborn with oxygen dependence early pregnancy, indicating a clear upward trend (such as superoxide
(oxygen concentration > 21%) over 28 days can be diagnosed as “new dismutase (SOD), catalase (CAT) and Glutathione peroxidase (GP)).
BPD”. Its characteristics include impaired lung function, mainly mani- This change in regarding fetuses suggests that the defensive capacity of
fested as airway hyperresponsiveness and airway obstruction, as well as the lungs in the defense of the uterus in the fetus is actually increasing,
obstructive ventilation dysfunction (Niedermaier and Hilgendorff, which seems to be essential in the process of fetal maturation. The in-
2015). Compared with traditional BPD (Jobe, 1999), its pathological crease of anti-oxidative enzyme activity before birth of fetuses con-
manifestations include less smooth muscle hyperplasia, lighter fibrosis, tributes to the smooth adaptation of lungs to a new respiratory en-
rarely, severe squamous metaplasia, decreased alveolar number, in- vironment, ensuring that a neonatal respiratory function can be
creased alveolar volume, and simple structure, pulmonary micro- adapted at birth (Tsukahara, 2007). At the same time, the tolerance of
vascular dysplasia and increased elastic tissue. In terms of the new BPD, term infants to relatively high level of oxygen in air can be explained by
the process of normal lung development is altered, leading to the de- the level of high antioxidant enzyme activity in mature type II alveolar
velopment of lungs in different orders and orientations. cells. As the most important endothelial cells, type-II alveolar cells that
mediate the antioxidant capacity of the lungs are more likely to be
2.2. Hyperoxia with lung injury hypoplasia in preterm labor and have a higher likelihood of being
earlier. Therefore, as to premature children with weak resistance to
Previous studies have shown that (Dani et al., 2004) infants have a hyperoxia exposure, there is not enough anti-oxidation ability to pre-
partial increase in oxidative stress signs before birth and at birth, sug- vent exposure to hyperoxia damage (Frank and Groseclose, 1984).
gesting an increase in its ability to resist oxidative stress, which may be
aimed at adapting to the increased level of oxidation due to the spon- 3. Oxidative stress
taneous inhalation of oxygen at birth. However, as for preterm children,
especially very premature ones and those with very low birth weight do 3.1. ROS
not have such response. Furthermore, due to its pulmonary in-
sufficiency, lack of alveolar surfactant, endogenous antioxidant en- Free radicals are defined as “any chemical that contains one or more
zymes system defects, the anti-oxidative stress system is imperfect. unpaired electrons and can stand alone” (Ryter and Choi, 2013). Free
Therefore, it is more likely to be exposed to hyperoxia. A very short electrons produced under the normal aerobic metabolism of cells are
time of high oxygen exposure will trigger a cascade of oxidative stress, readily accepted by oxygen to produce ROS, such as singlet oxygen,
thus leading to oxidative damage (Maltepe and Saugstad, 2009; superoxide (O2%) and oxygen-containing groups (hydroxyl, HO%) and
Kaarteenahowiik and Kinnula, 2004). oxidant H2O2 (Winterbourn and Hampton, 2008). The role of ROS in
The lung injury caused by hyperoxia exposure mainly includes cell the body depends largely on the internal environment of the body in
necrosis or apoptosis in the alveolar epithelium and vascular en- which ROS exists, including location, time, concentration, proximity,
dothelium, resulting in the destruction of alveolar structure, increased etc. (Auten and Davis, 2009). Current research suggests that ROS and its
vascular permeability, and massive recruitment of inflammatory cells reaction products can mediate cell signaling as signaling intermediates,
(Balany and Bhandari, 2015), which would can delay the separation of thus participating in adaptive or maladaptive molecular responses, in-
cystic alveoli with alveoli, inducing the proliferation of stromal cells, cluding cell growth, differentiation, apoptosis and necrosis (Saugstad,
thus leading to the persistence of inflammation, while blocking the 2010). In general, antagonism of free radicals to the enzymes and an-
formation of capillaries in the distal lung tissue. That is how it can tioxidants in body at physiological states can maintain the balance of
eventually result in pulmonary hypertension (Warner et al., 1998). ROS production and clearance. In terms of the mechanism of oxidative
Inflammatory cells cause damage to lung tissue by inducing in- stress reaction, any oxidant can induce oxidative stress reaction in an
flammatory and vascular responses by producing cytokines and che- excessive environment. ROS is the most important oxidant to initiate
motaxis. The relevant studies have shown that hyperoxia exposure in- oxidative stress reaction. Therefore, an overproduction of ROS under-
duces focal endothelial cell damage in the early stages of lung tissue, mines the oxidation-anti-oxidant balance and triggers an oxidative
which will leading to necrosis of epithelial cells if continued exposure to stress response that can lead to inflammatory reaction and tissue da-
hyperoxia still exists (Bhandari, 2010; Shyanne et al., 2016). mage. ROS can cause damage to all biological molecules, while the
Hyperoxia would cause pulmonary dynamics changes, the destruc- main targets of ROS in cells are proteins, nucleic acids, carbohydrates
tion of alveolar capillary membrane would cause gas exchange dis- and membrane lipids (Torres-Cuevas et al., 2016; Aparici et al., 2013;
orders, and pulmonary edema would cause pulmonary capillary per- Wiegman et al., 2014).
meability. All of them would eventually result in alveolar endothelial
and epithelial cell death, increased alveolar vascular permeability. 3.2. ROS with damage
Moreover, ruptured capillaries would result in a loss of lung integrity.
However, pulmonary edema and concomitant inflammatory processes The role of ROS in body is related to the environment it exists (Rees
decrease even with continued exposure to hyperoxia, but chronic lung et al., 2008). Physiologically, ROS induces an increase in specific types
inflammation occurs after acute reactions. Hyperoxia lung injury of of apoptosis. Therefore, under normal conditions, the synthesis of ROS
neonates occurs during the developmental stage, and is influenced by a is essential for the normal development of embryo morphology. In
variety of cytokines during hyperoxia-induced lung injury, thus forming addition, ROS stimulates the growth factors of live cells, and eliminates
a characteristic pathological manifestation of BPD (Hellström et al., dysfunctional proteins through oxidation, playing a vital role in main-
1990). taining the normal physiological functions of various organelles (Zuo
et al., 2015). In cells, mitochondria are the most important “factory” for
2.3. Relationship between hyperoxia exposure and BPD ROS (Quinlan et al., 2012; Parinandi et al., 2003; Carnesecchi et al.,
2009a) mainly through uncoupling of electron transport chains and
Long-term hyperoxia exposure is the most important cause of BPD electron leaks. Other components that can produce ROS include (Auten
pathogenesis (Kolls, 2017). A fetus relies on the placenta and the fetal and Davis, 2009) cellular components, such as the plasma membrane
circulation in its mother's body to obtain oxygen for developing tissues surface, endoplasmic reticulum-binding enzymes, and the cytosolic
for growth maintenance in relatively hypoxic conditions. The oxygen enzyme system; activated multi-enzyme systems include inflammation-

178
J. Wang, W. Dong Gene 678 (2018) 177–183

associated synthase (COX and LOX), NOS, cytochrome oxidase, and (Carnesecchi et al., 2011). Animal experiments has shown that O2 -
xanthine oxidoreductase. Hyperoxia, inflammation, ischemia-reperfu- increases with the increase of NADPH oxidase in lambs with endothelial
sion and antioxidant defensive decompensation/damage will lead to the dysfunction (Oishi et al., 2013). Similarly, endothelin-1 (ET-1) is an
oxidative stress response of body tissue. In the early stage, over- important inflammatory mediator that is released by excessive ROS and
production of ROS results in the destruction of oxidative and anti- mediates increased NADPH oxidase activity and elevated O2 levels in
oxidant balance in body and damage to tissue cells, especially the lung lung epithelial cells (Rafikova et al., 2013; Wedgwood et al., 2013).
tissue, which is mainly caused by inducing autotoxicity of lung epi- Alveolar epithelial type-II cells (AEC II), as the most important stem
thelial cells and endothelial cells concerning lung tissue damage cells in lung tissue play a crucial role in the regulation of lung tissue
(Chuang et al., 2014). growth, maturation, wound-repair process and lung fluid homeostasis.
ROS is a highly active oxidant that directly damages cellular Proliferation and differentiation of AECIIs are the major factors which
structures and can directly damage proteins, lipids and nucleic acids, as regulate the post-injury repair of alveolar epithelial structures and
well as cause cell death through a variety of pathways (Cadenas, 2018; functions. As a stem cell, AECII can be converted to AECI, and can se-
Richter et al., 2015; Sullivan and Chandel, 2014; Tann et al., 2011). crete a variety of bioactive substances to restore alveolar capillary
ROS can act on the biofilm to induce membrane lipid peroxidation. barrier integrity and maintain alveolar function, oxidative stress in-
Lipid peroxidation can activate sphingomyelinase and release large duced by hyperoxia induces excessive apoptosis of AECII and inhibits
amounts of ceramide to induce apoptosis (Tann et al., 2011; Sies, 2017; its proliferation, which is crucial for the development of BPD (Zhang
Burgoyne et al., 2013). ROS can initiate protein oxidation processes and et al., 2014; Liu et al., 2014; Hou et al., 2015; Wu et al., 2018). Pul-
nitrosylation processes, such as carbonylation, nitration and nitrotyr- monary remodeling and vascular remodeling induced by hyperoxia are
osine formation, thus affecting the biological activity and formation of the basis of BPD formation. Airway remodeling during lung develop-
various enzymes, ultimately leading to significant cellular dysfunction. ment is the result of the combination of pathological processes that
In addition, ROS can act directly on oxidized nucleic acid which will includes airway epithelial cells, smooth muscle cells, inflammatory cells
lead to DNA strand breaks, thereby inducing cell necrosis or apoptosis. and ECM components. The main causes of airway remodeling include
At the same time, ROS can activate multiple signal transduction path- airway epithelial damage, especially AECII repair abnormalities (Royce
ways in cells, promoting the transmission of cytokines, growth factors et al., 2014; Shimoda and Laurie, 2013).
and calcium signals, and eventually leading to cell death (Tiganis, Cells under various harmful or stressful conditions will induce ab-
2011; Barnes, 1990). normal post-translational modifications of protein, resulting in a lot of
Oxidative stress triggered by hyperoxia exposure regulates the an- unfolded protein accumulation in cell and destroying the normal phy-
tioxidant capacity in vivo, while the antioxidant enzyme system of siological function of cell, Thus, eukaryotic cells would evolve into an
premature infants is dysplasia. Compared with those term infants, unfolded protein response (UPR) process to relieve unfolded protein
premature infants suffer weak expression and activity of antioxidant burden, including increasing protein folding capacity, degrading mis-
enzymes under hypoxic conditions. Therefore, poor tolerance to hy- folded proteins and inhibiting protein translation (Lee et al., 2014;
peroxia of preterm infants is also susceptible to ROS-mediated injury Konsavage et al., 2012). During hyperoxia exposure, oxidative stress
and is more likely to cause lung injury and developmental block promotes the increase of UPR (non-folded protein) and induces stronger
(Kaarteenahowiik and Kinnula, 2004). Datta et al. (2015) believe that endoplasmic reticulum oxidative stress (ER) (Teng et al., 2017; Bai
the leakage of ROS in the matrix may be involved in the occurrence of et al., 2017), ER mediates apoptosis of AECII cells, oxidative stress, ROS
injury. Additionally, in the isolated pulmonary arterial smooth muscle production and amplification of inflammation, leading to the apoptosis
cells, it confirmed that the exposure to high oxygen concentration first in mitochondria-dependent and independent pathways, as well as the
increased the production of ROS in mitochondria, followed by an in- alveolar growth retardation, which would damage the development of
crease level of ROS in cytoplasm, eventually leading to oxidative stress lung morphology, and cause increased AECII apoptosis while mediating
damage caused by the overload of ROS in cells. ROS is associated with impaired angiogenesis and vasodilation. Flodby et al. (2016) found that
pathophysiological changes in many lung diseases, such as broncho- ER-deficient mice stunted alveolar development, perinatal lung sur-
pulmonary dysplasia (Djr et al., 2017; Domej et al., 2014; Hoffman factant protein and type-I cell markers reduce, contributing to the oc-
et al., 2015). In the relevant studies about of human BPD, a great currence of BPD.
number of researches have shown that ROS is associated with ab-
normalities and injuries during lung maturation. 3.3.2. Oxidative stress with vascular development
The process of normal lung maturation and the formation and ma-
3.3. Oxidative stress with BPD turation of blood vessels are benefit from the presence of many kinds of
growth factors, such as FGF, TGF, HGF and VEG (Jin et al., 2016; Chen
3.3.1. Oxidative stress with lung development et al., 2017b). VEGF has an effect in promoting endothelial growth and
Many enzymes are capable of producing reactive oxygen species remodeling lung tissue in endothelial cells, especially during normal
(ROS) in human cells, but the NOX family is currently the only enzyme alveolar formation and vascular growth. The relevant studies have
that has been confirmed to produce ROS as its main function (Crosas- shown that (Muramatsu et al., 2016) vascular endothelial growth factor
Molist and Fabregat, 2015). Single Nucleotide Polymorphisms (SNPs) in receptor 2 (VEGFR2) is very essential for maintaining the normal
the NADPH oxidase (NOX) family are closely associated with oxidative structure and function of alveoli. The reduce of VEGFR2 activation level
stress injury, especially in preterm infants (Huizing et al., 2017). The is closely related to the stagnation of lung maturation, which can also
mechanism by which the NOX enzyme catalyzes the production of su- induce the apoptosis activity of endothelial cells and mesenchymal
peroxide and free radicals is primarily operated by facilitating oxygen cells. VEGF in allowing angiogenesis and alveolarization is a pre-
molecules to receive electrons transferred from NADPH (Brandes et al., requisite for the remodeling and repair of lung injury during hyperoxia-
2014). NOX1, NOX3 and NOX5 produce superoxide, while NOX2 and induced injury of microvasculature. There is a decrease tendency for
NOX4 mainly produce hydrogen peroxide (Bedard and Krause, 2007; VEGF in human children with BPD and the use of VEGF antagonists in
Takac et al., 2011). The increase number and over-activation of Nox1 neonatal rats resulted in significant impaired alveolar development
during hyperoxia exposure is an important link leading to excessive (Jiménez et al., 2018; Lingappan et al., 2016). Salaets et al. (2015) have
ROS production and destruction of the alveolar capillary barrier, and is demonstrated the protective effect of VEGF on mesenchymal stem cells
a participant in hyperoxia-induced acute oxidative stress injury in a hyperoxia-induced ALI rat model. The decreased expression of
(Carnesecchi et al., 2009b). In contrast, Nox1 deficiency showed sig- VEGF results in the cessation of sprouting during development of lung
nificant protection against hyperoxia-induced lung injury of mice angiogenesis. Excessive ROS and nitrogenous substances can be

179
J. Wang, W. Dong Gene 678 (2018) 177–183

induced by oxidative stress and lead to apoptosis (Berger and Bhandari, thus ultimately promoting apoptosis (Kawaguchi et al., 2017;
2014). Lingappan et al., 2016).
Inadequate angiogenesis, accompanied by hypoxia in local tissues, Previous studies have shown that (Misra et al., 2015) sustained high
leads to the dysplasia of blood vessels and alveoli and is an important oxygen exposure can up-regulate CINC-1 in lung tissue. Administration
component of the mechanism of BPD. In addition, Zhang and Lingappan of anti-CINC-1 significantly reduced the neutrophils and myeloperox-
(2017) have found that HUVECs (umbilical vein endothelial cells) of idase activities in the alveolar lavage fluid while preserving more al-
female mice showed a better cell viability and angiogenesis compared veolar volume and surface area. However, the inhibition of neutrophils
with male ones after hyperoxia exposure. In oxidative stress, male mice recruitment in lung tissue of neonatal rats exposed to hyperoxia has
produced more H2O2 with macrophages and neutrophil infiltration been confirmed as an element which can reduce DNA-oxidation-in-
increased more significantly. In addition, the number of pulmonary duced breakage and injury, and inhibit the HO% formation and O2%
vessels and the expression of angiogenic markers (PECAM1 and accumulation. At the same time, it can enhance the alveolar develop-
VEGFR2) were more severely damaged. ment. The LPA receptor (LPAR1-6), as a 7-transmembrane G protein-
coupled receptor, is associated with a variety of physiological and pa-
3.3.3. Oxidative stress with inflammation thological responses, including pulmonary fibrosis, LPS-related in-
During hyperoxia exposure, the recruitment of inflammatory cells in flammation, bronchospasm, and airway hyperresponsiveness. Recently,
lung, which is caused by a large number of chemokines produced by Shim et al. (2015) have demonstrated that compared with air control
monocytes/macrophages, lymphocytes, epithelial cells, endothelial group, young rat pups exposed to hyperoxia had an important increase
cells and stroma, is the major mechanism leading to lung injury in enzyme production and activity to produce LPA and an increase in
(Bhandari, 2010; Thomas and Speer, 2014). NO is essential for main- LPA receptor LPAR1 and LPAR3 expression. Such data indicate that in
taining lung homeostasis. However, nitrification stress is activated and neonatal rats, LPA synthesis and secretion and signal transduction de-
enhanced by excessive NO, thus exerting pro-inflammatory effects. In- pendent on LPA can be induced by hyperoxia. This process may be
flammatory stimuli, hypoxia and hyperoxia will lead to increased iNOS related to the progression of BPD. Chen et al. (2017a) have found that
expression in lungs. Reynolds et al. (2016) have found that Lung iNOS under hyperoxia exposure, the inhibition of LPAR1 has protective ef-
protein expression in the hyperoxia models of mouse can serve as a fects on lung injury and fibrosis associated with ROS and inflammation.
biomarker of inflammation, revealing that hyperoxia exposure leads to Sphingosine-1-phosphate (S1P) is a signaling molecule existing inside
potential lung inflammation. and outside a cell, whose level is regulated by the synthesis of SphKs 1
Many kinds of proinflammatory cytokines are activated during and 2. Hyperoxia exposure may cause an increase in SphK1. Natarajan
oxidative stress. Chemotactic inflammatory cells stimulate the synthesis et al. (2017) have demonstrated that a significant increase in S1P/
and secretion of inflammatory mediators, and enhance the in- SphK1 signaling in neonatal lungs is associated with hyperoxia-induced
flammatory response (Anita et al., 2016; Yeh et al., 2016), such as IL-1, apoptosis, promoting gene expression, inflammation, extracellular
IL-6, IL-17 (Lawrence et al., 2017), IL-8, CINC-1 and MCP-1. IL-1β was matrix remodeling, and cell proliferation arrest. LOOH (Lipid Hydrogen
demonstrated to be overexpressed in alveolar epithelial cells in the Peroxide) is a special substance produced during lipid peroxidation
model of neonatal mice with BPD (Rudloff et al., 2017). IL-6 was found widely distributing in the whole body. Its production is dependent on
to be involved in the inflammatory cascade (Matsumura et al., 2017), oxygen radicals peroxidation of unsaturated fatty acids (especially in
and was overexpressed in lung tissue exposed to hyperoxia. At the same membrane phospholipids). LOOH has a strong destructive effect on
time, the relevant studies have shown that high levels of IL-6 are as- many biological macromolecules in cells. Fabiano et al. (2016) have
sociated with more severe pulmonary edema and inflammatory re- found that the level of LOOH in the alveolar lavage of children with
sponse during early exposure (Hsiao et al., 2017). Monocyte Chemo- BPD increased, which may be related to ROS overload. At the same time
tactic Protein-1 (MCP-1), as a chemokine like CINC-1, has main target a large number of cytokines, proteins and enzymes are induced by the
cells of monocytes, lymphocytes and basophils (Kumar et al., 2016). oxidative stress of body and promote cell death, such as acetylated P53.
The level of MCP-1 is elevated in tracheal aspirates of hyperoxia-in- It has been reported that (Hori et al., 2013; Marcel et al., 2015) hy-
duced premature infants with BPD, which is related to BPD. In the peroxia exposure can increase the transcriptional level of P53 and the
animal model with BPD, the number of neutrophils in tracheal aspirate expression and activity of acetylated P53. HGF (hepatocyte growth
and bronchoalveolar lavage fluid increased with the increasing level of factor) (Seedorf et al., 2016) has been demonstrated to protect neonatal
IL-8 protein, which was associated with infection. IL-8 protein con- lung tissue against hyperoxic-related injury in animal models under
centrations in homogenates of pre-term explant culture of lung exposed hyperoxia exposure while improving the dysfunction and preventing
to hyperoxia were much higher than those of lungs of full-term babies the alveolar development. However, the HGF level of premature infants
(Leroy et al., 2017). Tumor necrosis factor-α (TNF-α) (Ehrhardt et al., is low. HGF can not be protected from being up-regulated under hy-
2016a; Ehrhardt et al., 2016b) can induce apoptosis due to the in- peroxia exposure. The anti-oxidative damage is weak and alveolar de-
flammation through death receptor signaling. It is mainly composed of velopment is more easily inhibited.
various activated immune cells, including macrophages, natural killer As for adults and newborns, there is a significant difference in the
(NK) cells, granulocytes, CD4 + lymphocytes, mast cells and eosino- type and timing of cytokine synthesis and secretion in lung tissue
phils. The high level of TNF-α may promote chronic inflammation during hyperoxia exposure. Lung has a complex developmental and
leading to the development of BPD. Experiments in neonatal rats by cause mechanism for lung injury by hyperoxia of newborn infants,
Oncel et al. (2015) have showed that the inhibition of TNF-α is bene- especially preterm infants. Various cytokines would produce different
ficial for alveolar and lung injury by reducing lung inflammation and pro-inflammatory/anti-inflammatory effects. Inflammation triggered
oxidative stress. Kaya et al. (2016) have indicate that the inhibition of by hyperoxia exposure erupts acutely in the early stages of acute injury,
NF-κB and TNF-α in the BPD model of neonatal rats can decrease the followed by a reduction in acute and inflammatory responses to chronic
MDA level while increase the GSH-PX and SOD levels, which is helpful inflammation depending on the acute lung response to hyperoxia.
for lung development and pulmonary vascularization. At the same time, Neonates, however, are specific to hyperoxia-induced inflammatory
TNF-α activates NOX to generate excess ROS. Mitochondria-generated damage occurring in the alveolar development (Wollen et al., 2013;
ROS plays a signaling role in enhancing TGF signaling and promoting Thomas and Speer, 2014; Wollen et al., 2014).
fibrosis (Gonzalez-Gonzalez et al., 2017). NF-κB can be activated by
oxidative stress (Yu et al., 2015). Activated NF-κB mediates the pro- 3.4. Oxidation with anti-oxidation
duction and activation of many inflammatory factors with pro-in-
flammatory effect, and mediates the translocation of nuclear proteins, Redox potential is thought to be the key to many metabolic

180
J. Wang, W. Dong Gene 678 (2018) 177–183

pathways, including cell proliferation, differentiation and apoptosis. to the calcitonin family, the normal physiological functions of en-
Abnormalities of such pathways can lead to a characteristic block of dothelial cells depends on the action of AM. Zhang et al. (2015) have
blood vessels and a stalled alveolus in BPD. Mohamed et al. (2015) have demonstrated that AM can reduce oxidative stress, inflammation and
found that the oxidative stress injury caused by the hyperoxia plays an cytotoxicity induced by hyperoxia by activating Akt or protein kinase B
important role in the development of BPD. Antioxidant enzymes are (Akt/PKB). Calcitonin gene-related peptide (CGRP) is a 37-amino acid
essential components of antioxidants in the whole body. The specific neuropeptide. It is an important biomarker for pain control and is
distribution of antioxidant enzymes in cells determines the sensitivity of closely related to the embryonic development and cell proliferation
tissues and cells to ROS-related injury, including superoxide dismutase, (Azuma et al., 2016; Dang et al., 2012). In lung tissue, AECII cells can
catalase, glutathione peroxidase and peroxiredoxin. The major non- secrete CGRP and mainly distribute in nerve fibers around vascular
enzymatic intracellular antioxidants are glutathione (GSH) (Sullivan smooth muscle and airway mucosa. Recent studies have shown (Fu
et al., 2013). The reduced glutathione can be oxidized by glutathione et al., 2010; Yu et al., 2015; Li and Wang, 2006) that CGRP can improve
peroxidase to reduce free radical activity and the production of H2O2 hyperoxia-induced oxidative stress and inflammatory response, thus
and lipid peroxidation products, or to stabilize electrophiles. Glu- preventing hyperoxia-induced apoptosis and destruction of lung struc-
tathione reductase subsequently is mediated to restore reduced glu- ture by inhibiting the production of ROS. The decompensation of en-
tathione by using electrons from NADPH. Immature and low activity of dogenous antioxidant system may lead to the redox function and induce
the antioxidant enzyme system of preterm infants is a causing me- the oxidative stress injury, which can induce the transcription of genes
chanism for lung injury by hyperoxia and lung development retardation that inhibiting the progression of cell cycle (Schieber and Chandel,
(Poggi and Dani, 2014). Extracellular superoxide dismutase (EC-SOD) is 2014). However, there has been some controversy in the recent use of
one of the essential antioxidant enzymes, which is highly expressed in antioxidants. The relevant literature has suggested that antioxidants
lung tissue and vascular system, and can dissociate superoxide into may play a dual role in promoting the survival rate of cancer cells.
hydrogen peroxide and oxygen. Studies conducted by Cassidy et al. Moreover, the efficacy of antioxidants is positively correlated with the
(2015) has showed that under oxidative stress, mice lacking the EC-SOD drug concentration in target organs. Therefore, targeted administration
system had more-pronounced obstructive alveolar development, pul- is also important in clinical application of antioxidants (Saeidnia and
monary hypertension, and pulmonary vascular remodeling. Nrf2, Abdollahi, 2013).
known as the “main helmsman” for antioxidant responses, stimulates In summary, the oxidative stress response plays an important role in
most of the gene expressions for antioxidant/detoxifying enzymes, with the causing mechanism regarding hyperoxia-induced BPD. The oxida-
antioxidants (SODs) and NQO1 induced (Loboda et al., 2016). NQO1 is tion/antioxidant balance can be broken by many factors. Oxidative
one of the important antioxidant enzymes through which the oxidative stress can induce an oxidative stress response, which can lead to cas-
stress induces NQO1 gene expression and regulates the synthesis of ROS cade reactions, inducing cell death and further resulting in acute lung
and other free radicals, thereby protecting cells from oxidative injury. injury. Under the condition of hyperoxia exposure, continuous oxida-
However, recently, Gavrili et al. (2015) found that the NQO1 variant tion can affect the process of lung development, alveolar development
genotype may increase the risk of BPD. and angiogenesis, eventually leading to BPD. In addition, inflammation,
Other antioxidants include NO, CO, AM, etc. During normal alveolar infection, genetic susceptibility and any other iatrogenic injuries and
development, endogenous NO signaling is enhanced, thereby sig- factors concerning the pathogenesis of BPD mechanism are still needed
nificantly improving the oxygenation response while reducing the lung further study. The study on the action mechanism of oxidative stress
oxidation (Davis, 2002; Tsukahara, 2014). CO is a gaseous signal mo- plays an important guiding role in the control of clinical use of oxygen
lecule that has a powerful protective activity on tissues and cells during therapy, early detection and prevention of oxidative injury and im-
oxidative stress (Anyanwu et al., 2014). Adrenomedullin (AM) belongs provement of the prognosis of BPD.

181
J. Wang, W. Dong Gene 678 (2018) 177–183

References Cell. Mol. Phys. 310 (10), L909.


Fabiano, A., Gavilanes, A.W.D., Zimmermann, L.J.I., et al., 2016. The development of
lung biochemical monitoring can play a key role in the early prediction of bronch-
Anita, B., Christopher, C., Vineet, B., 2016. BPD following preterm birth: a model for opulmonary dysplasia. Acta Paediatr. 105 (5), 535–541.
chronic lung disease and a substrate for ARDS in childhood. Front. Pediatr. 4(3). Flodby, P., Li, C., Liu, Y., et al., 2016. GRP78 regulates ER homeostasis and distal epi-
Anyanwu, A.C., Bentley, J.K., Popova, A.P., et al., 2014. Suppression of inflammatory cell thelial cell survival during lung development. Am. J. Respir. Cell Mol. Biol. 55 (1),
trafficking and alveolar simplification by the heme oxygenase-1 product carbon 135.
monoxide. Am. J. Physiol. Lung Cell. Mol. Physiol. 306 (8), L749. Frank, L., Groseclose, E.E., 1984. Preparation for birth into an O2-rich environment: the
Aparici, S., Martorell, L., Codoñer-Franch, P., 2013. Role of oxidative stress in preterm antioxidant enzymes in the developing rabbit lung. Pediatr. Res. 18 (3), 240–244.
infants with bronchopulmonary dysplasia after exposure to chorioamnionitis. J. Fu, H.M., Li, L., Wang, Y.J., et al., 2010. The proliferation-promoting effects of calcitonin
Pediatr. Biochem. 03 (03), 143–153. gene-related peptide on type II alveolar epithelial cell exposed to hyperoxia mediated
Auten, R.L., Davis, J.M., 2009. Oxygen toxicity and reactive oxygen species: the devil is in by protein kinase C alpha pathway. Zhongguo Wei Zhong Bing Ji Jiu Yi Xue 22 (5),
the details. Pediatr. Res. 66 (2), 121–127. 263–266.
Azuma, Y., Miwa, Y., Sato, I., 2016. Expression of CGRP in embryonic mouse masseter Gavrili, S., Zachaki, S., Daraki, A., et al., 2015. Association of C609T-inborn poly-
muscle. Ann. Anat. 206, 34–47. morphism of NAD(P)H: quinone oxidoreductase 1 with the risk of bronchopulmonary
Bai, Y.X., Fang, F., Jiang, J.L., 2017. Extrinsic calcitonin gene-related peptide inhibits dysplasia in preterm neonates. Am. J. Perinatol. 33 (06), 535–539.
hyperoxia-induced alveolar epithelial type II cells apoptosis, oxidative stress, and Gonzalez-Gonzalez, F.J., Chandel, N.S., Jain, M., 2017. Reactive oxygen species as sig-
reactive oxygen species (ROS) production by enhancing notch 1 and homocysteine- naling molecules in the development of lung fibrosis. Transl. Res. 190, 61–68.
induced endoplasmic reticulum protein (HERP) expression. Med. Sci. Monit. 23, https://doi.org/10.1016/j.trsl.2017.09.005.
5774–5782. Hellström, A., Smith, L.E., Dammann, O., 1990. Retinopathy of prematurity. Dev. Med.
Balany, J., Bhandari, V., 2015. Understanding the impact of infection, inflammation, and Child Neurol. 32 (5), 377–378.
their persistence in the pathogenesis of bronchopulmonary dysplasia. Front. Med. 2 Hoffman, S., Nolin, J., Mcmillan, D., et al., 2015. Thiol redox chemistry: role of protein
(3), 90. cysteine oxidation and altered redox homeostasis in allergic inflammation and
Barnes, P.J., 1990. Reactive oxygen species and airway inflammation. Free Radic. Biol. asthma. J. Cell. Biochem. 116 (6), 884.
Med. 9 (3), 235–243. Hori, Y.S., Kuno, A., Hosoda, R., et al., 2013. Regulation of FOXOs and p53 by SIRT1
Bedard, K., Krause, K.H., 2007. The NOX family of ROS-generating NADPH oxidases: modulators under oxidative stress. PLoS One 8 (9), e73875.
physiology and pathophysiology. Physiol. Rev. 87 (1), 245. Hou, A., Fu, J., Yang, H., et al., 2015. Hyperoxia stimulates the transdifferentiation of
Berger, J., Bhandari, V., 2014. Animal models of bronchopulmonary dysplasia. The term type II alveolar epithelial cells in newborn rats. Am. J. Physiol. Lung Cell. Mol.
mouse models. Am. J. Physiol. Lung Cell. Mol. Physiol. 307 (12), 936–947. Physiol. 308 (9), L861.
Bhandari, V., 2010. Hyperoxia-derived lung damage in preterm infants. Semin. Fetal Hsiao, C.C., Chang, J.C., Tsao, L.Y., et al., 2017. Correlates of elevated interleukin-6 and
Neonatal Med. 15 (4), 223. 8-hydroxy-2′-deoxyguanosine levels in tracheal aspirates from very low birth weight
Brandes, R.P., Norbert, W., Katrin, S., 2014. Nox family NADPH oxidases: molecular infants who develop bronchopulmonary dysplasia. Pediatr. Neonatol. 58 (1), 63–69.
mechanisms of activation. Free Radic. Biol. Med. 76, 208–226. https://doi.org/10.1016/j.pedneo.2016.01.004.
Buczynski, B.W., Maduekwe, E.T., O'Reilly, M.A., 2013. The role of hyperoxia in the Huizing, M.J., Cavallaro, G., Moonen, R.M., et al., 2017. Is the C242T polymorphism of
pathogenesis of experimental BPD. Semin. Perinatol. 37 (2), 69–78. the CYBA gene linked with oxidative stress-associated complications of prematurity?
Burgoyne, J.R., Oka, S., Aleagha, N., et al., 2013. Hydrogen peroxide sensing and sig- Antioxid. Redox Signal. 27, 1432–1438. https://doi.org/10.1089/ars.2017.7042.
naling by protein kinases in the cardiovascular system. Antioxid. Redox Signal. 18 Jiménez, J., Lesage, F., Richter, J., et al., 2018. Upregulation of vascular endothelial
(9), 1042. growth factor in amniotic fluid stem cells enhances their potential to attenuate lung
Cadenas, S., 2018. ROS and redox signaling in myocardial ischemia-reperfusion injury injury in a preterm rabbit model of bronchopulmonary dysplasia. Neonatology 113
and cardioprotection. Free Radic. Biol. Med. 117, 76–89. (3), 275–285.
Carnesecchi, S., Deffert, C., Pagano, A., et al., 2009a. NOX1 plays a crucial role in hy- Jin, Meihua, Lee, Juyoung, Lee, 等, Kyung-yup, 2016. Alteration of TGF-Î2-ALK-Smad
peroxia-induced acute lung injury in mice. Am. J. Respir. Crit. Care Med. 180 (10), signaling in hyperoxia-induced bronchopulmonary dysplasia model of newborn rats.
972–981. Exp. Lung Res. 42 (7), 1.
Carnesecchi, S., Deffert, C., Pagano, A., et al., 2009b. NADPH oxidase-1 plays a crucial Jobe, A.J., 1999. The new BPD: an arrest of lung development. Pediatr. Res. 46 (6),
role in hyperoxia-induced acute lung injury in mice. Am. J. Respir. Crit. Care Med. 641–643.
180 (10), 972–981. Jr, W.H.N., Rosan, R.C., 1968. Radiographic features of pulmonary oxygen toxicity in the
Carnesecchi, S., Dunandsauthier, I., Singovsky, G., et al., 2011. NADPH oxidase isoform 1 newborn: bronchopulmonary dysplasia1. Radiology 91 (1), 49–58.
is expressed in lung tissue of ARDS patients and decreases hyperoxia-induced ROS Kaarteenahowiik, R., Kinnula, V.L., 2004. Distribution of antioxidant enzymes in devel-
production and cell death in pulmonary type II epithelial cells. Eur. Respir. J. (Suppl. oping human lung, respiratory distress syndrome, and bronchopulmonary dysplasia.
55), 2942. J. Histochem. Cytochem. 52 (9), 1231–1240.
Carraro, S., Filippone, M., Da, D.L., et al., 2013. Bronchopulmonary dysplasia: the earliest Kawaguchi, T., Yanagihara, T., Yokoyama, T., et al., 2017. Probucol attenuates hyper-
and perhaps the longest lasting obstructive lung disease in humans. Early Hum. Dev. oxia-induced lung injury in mice. PLoS One 12 (4), e0175129.
89 (5), S3. Kaya, G., Saldir, M., Polat, A., et al., 2016. Evaluation of etanercept treatment in newborn
Cassidy, D., Wright, R.H., Tang, J.R., et al., 2015. Lack of EC-SOD worsens alveolar and rat model with hyperoxic lung injury. Pediatr. Pathol. 35 (5), 327–338.
vascular development in a neonatal mouse model of bleomycin-induced broncho- Kolls, Jay K., 2017. Commentary: understanding the impact of infection, inflammation
pulmonary dysplasia and pulmonary hypertension. Pediatr. Res. 78 (6), 634–640. and their persistence in the pathogenesis of bronchopulmonary dysplasia. Front.
Chen, X., Walther, F.J., Laghmani, E.H., et al., 2017a. Adult lysophosphatidic acid re- Med. 4.
ceptor 1-deficient rats with hyperoxia-induced neonatal chronic lung disease are Konsavage, W.M., Zhang, L., Wu, Y., et al., 2012. Hyperoxia-induced activation of the
protected against lipopolysaccharide-induced acute lung injury. Front. Physiol. 8. integrated stress response in the newborn rat lung. Am. J. Physiol. Lung Cell. Mol.
Chen, X.Q., Wu, S.H., Luo, Y.Y., et al., 2017b. Lipoxin A4 attenuates bronchopulmonary Physiol. 302 (1), L27.
dysplasia via upregulation of let-7c and downregulation of TGF-β1 signaling Kotecha, S.J., Edwards, M.O., Watkins, W.J., et al., 2013. Effect of preterm birth on later
pathway. Inflammation 40 (6), 1–15. FEV1: a systematic review and meta-analysis. Thorax 68 (8), 760–766.
Chuang, C.Y., Degendorfer, G., Davies, M.J., 2014. Oxidation and modification of ex- Kumar, V.H.S., Lakshminrusimha, S., Kishkurno, S., et al., 2016. Neonatal hyperoxia in-
tracellular matrix and its role in disease. Free Radic. Res. 48 (9), 970–989. creases airway reactivity and inflammation in adult mice. Pediatr. Pulmonol. 51 (11),
Crosas-Molist, E., Fabregat, I., 2015. Role of NADPH oxidases in the redox biology of liver 1131–1141.
fibrosis. Redox Biol. 6, 106–111. Lawrence, S.M., Ruoss, J.L., Wynn, J.L., 2017. IL-17 in neonatal health and disease. Am.
Dang, H., Yang, L., Wang, S., et al., 2012. Calcitonin gene-related peptide ameliorates J. Reprod. Immunol.(1 Pt 1).
hyperoxia-induced lung injury in neonatal rats. Tohoku J. Exp. Med. 227 (2), 129. Lee, D., Sun, S., Ho, A.S., et al., 2014. Hyperoxia resensitizes chemoresistant glioblastoma
Dani, C., Cecchi, A., Bertini, G., 2004. Role of oxidative stress as physiopathologic factor cells to temozolomide through unfolded protein response. Anticancer Res. 34 (6),
in the preterm infant. Minerva Pediatr. 56 (4), 381–394. 2957–2966.
Datta, A., Kim, G.A., Taylor, J.M., et al., 2015. Mouse lung development and NOX1 in- Leroy, S., Caumette, E., Waddington, C., et al., 2017. A time-based analysis of in-
duction during hyperoxia are developmentally regulated and mitochondrial ROS flammation in infants at risk of bronchopulmonary dysplasia. J. Pediatr. 192.
dependent. Am. J. Phys. Lung Cell. Mol. Phys. 309 (4), 369–377. Li, W.J., Wang, T.K., 2006. Calcitonin gene-related peptide inhibits interleukin-1beta-
Davis, J.M., 2002. Role of oxidant injury in the pathogenesis of neonatal lung disease. induced interleukin-8 secretion in human type II alveolar epithelial cells. Acta
Acta Paediatr. 91 (s437), 23–25. Pharmacol. Sin. 27 (10), 1340.
Djr, F., Li, X., Bordan, Z., et al., 2017. Reactive oxygen and nitrogen species in the de- Lingappan, K., Jiang, W., Wang, L., et al., 2016. Sex-specific differences in neonatal hy-
velopment of pulmonary hypertension. Antioxidants 6 (3), 54. peroxic lung injury. Am. J. Physiol. Lung Cell. Mol. Physiol. 311 (2), L481.
Domej, W., Oettl, K., Renner, W., 2014. Oxidative stress and free radicals in Liu, G., Zhang, J., Chen, H., et al., 2014. Effects and mechanisms of alveolar type II
COPD—implications and relevance for treatment. Int. J. Chron. Obstruct. Pulmon. epithelial cell apoptosis in severe pancreatitis-induced acute lung injury. Exp. Ther.
Dis. 9 (9), 1207–1224. Med. 7 (3), 565.
Ehrhardt, H., Pritzke, T., Oak, P., et al., 2016a. Absence of TNF-α enhances inflammatory Loboda, A., Damulewicz, M., Pyza, E., et al., 2016. Role of Nrf2/HO-1 system in devel-
response in the newborn lung undergoing mechanical ventilation. Am. J. Phys. Lung opment, oxidative stress response and diseases: an evolutionarily conserved me-
Cell. Mol. Phys. 310 (10), L909. chanism. Cell. Mol. Life Sci. 73 (17), 3221–3247.
Ehrhardt, H., Pritzke, T., Oak, P., et al., 2016b. Biomarkers in lung diseases: from pa- Maltepe, E., Saugstad, O.D., 2009. Oxygen in health and disease: regulation of oxygen
thogenesis to prediction to new therapies: absence of TNF-α enhances inflammatory homeostasis-clinical implications. Pediatr. Res. 65 (3), 261–268.
response in the newborn lung undergoing mechanical ventilation. Am. J. Phys. Lung Marcel, V., Catez, F., Diaz, J.J., 2015. p53, a translational regulator: contribution to its

182
J. Wang, W. Dong Gene 678 (2018) 177–183

tumour-suppressor activity. Oncogene 34 (44), 5513. Shim, G.H., Kim, H.S., Kim, E.S., et al., 2015. Expression of autotaxin and lysopho-
Matsumura, H., Ichiba, H., Ohnishi, S., et al., 2017. Histologic chorioamnionitis, amniotic sphatidic acid receptors 1 and 3 in the developing rat lung and in response to hy-
fluid interleukin 6, Krebs von den Lungen 6, and transforming growth factor β1 for peroxia. Free Radic. Res. 49 (11), 1362–1370.
the development of neonatal bronchopulmonary dysplasia. Jpn. Clin. Med. 8, Shimoda, L.A., Laurie, S.S., 2013. Vascular remodeling in pulmonary hypertension. J.
1179066017696076. Mol. Med. 91 (3), 297–309.
Misra, R.S., Shah, S., Fowell, D.J., et al., 2015. Preterm cord blood CD4+, T cells exhibit Shyanne, P., Alli, M., Al-Ahmad, A.J., 2016. Cerebral hypoxia/ischemia selectively dis-
increased IL-6 production in chorioamnionitis and decreased CD4+, T cells in rupts tight junctions complexes in stem cell-derived human brain microvascular en-
bronchopulmonary dysplasia. Hum. Immunol. 76 (5), 329–338. dothelial cells. Fluids Barriers Cns 13 (1), 16.
Mohamed, I., Elremaly, W., Rouleau, T., et al., 2015. Oxygen and parenteral nutrition two Sies, H., 2017. Hydrogen peroxide as a central redox signaling molecule in physiological
main oxidants for extremely preterm infants: ‘It all adds up’. J. Neonatal-Perinatal oxidative stress: oxidative eustress. Redox Biol. 11 (C), 613.
Med. 8 (3), 189–197. Sullivan, L.B., Chandel, N.S., 2014. Mitochondrial reactive oxygen species and cancer.
Muramatsu, Y., Ito, M., Oshima, T., et al., 2016. Hydrogen-rich water ameliorates Cardiovasc. Ultrasound 2 (1), 1–12 (2,1(2014-11-28)).
bronchopulmonary dysplasia (BPD) in newborn rats. Pediatr. Pulmonol. 51 (9), Sullivan, L.B., Martinez-Garcia, E., Nguyen, H., et al., 2013. The proto-oncometabolite
928–935. fumarate binds glutathione to amplify ROS-dependent signaling. Mol. Cell 51 (2),
Natarajan, V., Ha, A.W., Dong, Y., et al., 2017. Expression profiling of genes regulated by 236–248.
sphingosine kinase1 signaling in a murine model of hyperoxia induced neonatal Takac, I., Schröder, K., Zhang, L., et al., 2011. The E-loop is involved in hydrogen per-
bronchopulmonary dysplasia. BMC Genomics 18 (1), 664. oxide formation by the NADPH oxidase Nox4. J. Biol. Chem. 286 (15), 13304.
Niedermaier, S., Hilgendorff, A., 2015. Bronchopulmonary dysplasia - an overview about Tann, A.W., Boldogh, I., Meiss, G., et al., 2011. Apoptosis induced by persistent single-
pathophysiologic concepts. Mol. Cell. Pediatr. 2 (1), 1–7. strand breaks in mitochondrial genome. J. Biol. Chem. 286 (37), 31975–31983.
Northway Jr., W.H., R., C., 1967. Pulmonary disease following respiratory therapy of Teng, R.J., Jing, X., Michalkiewicz, T., et al., 2017. Attenuation of endoplasmic reticulum
hyaline-membrane disease. N. Engl. J. Med. 276, 357–368. stress by caffeine ameliorates hyperoxia-induced lung injury. Am. J. Physiol. Lung
Oishi, P.E., Sharma, S., Datar, S.A., et al., 2013. Rosiglitazone preserves pulmonary Cell. Mol. Physiol. 312 (5), L586.
vascular function in lambs with increased pulmonary blood flow. Pediatr. Res. 73 Thomas, W., Speer, C.P., 2014. Chorioamnionitis is essential in the evolution of
(1), 54. bronchopulmonary dysplasia – the case in favour. Paediatr. Respir. Rev. 15 (1),
Oncel, M.Y., Yurttutan, S., Alyamac, D.E., et al., 2015. Beneficial effect of etanercept on 49–52.
hyperoxic lung injury model in neonatal rats. J. Investig. Surg. 29 (1), 1–5. Tiganis, T., 2011. Reactive oxygen species and insulin resistance: the good, the bad and
O'Reilly, M., Thébaud, B., 2013. The promise of stem cells in bronchopulmonary dys- the ugly. Trends Pharmacol. Sci. 32 (2), 82–89.
plasia. [C]//seminars in perinatology. Semin. Perinatol. 79–84. Torres-Cuevas, I., Cernada, M., Nuñez, A., et al., 2016. Oxygen Modulation and
Parinandi, N.L., Kleinberg, M.A., Usatyuk, P.V., et al., 2003. Hyperoxia-induced NAD(P)H Bronchopulmonary Dysplasia: Delivery Room and Beyond[M]//Bronchopulmonary
oxidase activation and regulation by MAP kinases in human lung endothelial cells. Dysplasia. Springer International Publishing.
Am. J. Phys. Lung Cell. Mol. Phys. 284 (1), 26–38. Tsukahara, H., 2007. Biomarkers for oxidative stress: clinical application in pediatric
Pesce, C., Musi, L., 1996. Progress in the surgical treatment of congenital and acquired medicine. Curr. Med. Chem. 14 (3).
broncho-pulmonary pathology. Pediatr. Med. Chir. 18 (5), 451–461. Tsukahara, H., 2014. Oxidative Stress Biomarkers in Pediatric Medicine – A 2013 Update.
Poggi, C., Dani, C., 2014. Antioxidant strategies and respiratory disease of the preterm pp. 689–715.
newborn: an update. Oxidative Med. Cell. Longev. 2014 (5), 721043. Warner, B.B., Stuart, L.A., Papes, R.A., et al., 1998. Functional and pathological effects of
Quinlan, C.L., Orr, A.L., Perevoshchikova, I.V., et al., 2012. Mitochondrial complex II can prolonged hyperoxia in neonatal mice. Am. J. Phys. 275 (1), 110–117.
generate reactive oxygen species at high rates in both the forward and reverse re- Wedgwood, S., Lakshminrusimha, S., Czech, L., et al., 2013. Increased p22phox/Nox4
actions. J. Biol. Chem. 287 (32), 27255–27264. expression is involved in remodeling through hydrogen peroxide signaling in ex-
Rafikova, O., Rafikov, R., Kumar, S., et al., 2013. Bosentan inhibits oxidative and ni- perimental persistent pulmonary hypertension of the newborn. Antioxid. Redox
trosative stress and rescues occlusive pulmonary hypertension. Free Radic. Biol. Med. Signal. 18 (14), 1765.
56 (2), 28–43. Wiegman, C.H., Li, F., Clarke, C.J., et al., 2014. A comprehensive analysis of oxidative
Rees, M., Kennett, E.J., Davies, M., 2008. Oxidative damage to extracellular matrix and its stress in the ozone-induced lung inflammation mouse model. Clin. Sci. 126 (6),
role in human pathologies. Free Radic. Biol. Med. 44 (12), 1973–2001. 425–440.
Reynolds, C.L., Zhang, S., Shrestha, A.K., et al., 2016. Phenotypic assessment of pul- Winterbourn, C.C., Hampton, M.B., 2008. Thiol chemistry and specificity in redox sig-
monary hypertension using high-resolution echocardiography is feasible in neonatal naling. Free Radic. Biol. Med. 45 (5), 549–561.
mice with experimental bronchopulmonary dysplasia and pulmonary hypertension: a Wollen, E.J., Sejersted, Y., Wright, M.S., et al., 2013. Transcriptome profiling of the
step toward preventing chronic obstructive pulmonary disease. Int. J. Chron. newborn mouse lung after hypoxia and reoxygenation: hyperoxic reoxygenation af-
Obstruct. Pulmon. Dis. 11 (Issue 1), 1597. fects mTOR signaling pathway, DNA repair, and JNK-pathway regulation. Pediatr.
Richter, K., Konzack, A., Pihlajaniemi, T., et al., 2015. Redox-fibrosis: impact of TGFβ1 on Res. 74 (5), 536.
ROS generators, mediators and functional consequences. Redox Biol. 6, 344–352. Wollen, E.J., Sejersted, Y., Wright, M.S., et al., 2014. Transcriptome profiling of the
Royce, S.G., Li, X., Tortorella, S., et al., 2014. Mechanistic insights into the contribution of newborn mouse brain after hypoxia-reoxygenation: hyperoxic reoxygenation induces
epithelial damage to airway remodeling novel therapeutic targets for asthma. Am. J. inflammatory and energy failure responsive genes. Pediatr. Res. 75 (4), 517–526.
Respir. Cell Mol. Biol. 50 (1), 180–192. https://doi.org/10.1165/rcmb.2013- Wu, D., Liang, M., Dang, H., et al., 2018. Hydrogen protects against hyperoxia-induced
0008OC. apoptosis in type II alveolar epithelial cells via activation of PI3K/Akt/Foxo3a sig-
Rudloff, I., Cho, S.X., Bui, C.B., et al., 2017. Refining anti-inflammatory therapy strategies naling pathway. Biochem. Biophys. Res. Commun. 495 (2), 1620–1627.
for bronchopulmonary dysplasia. J. Cell. Mol. Med. 21 (6), 1128–1138. Yeh, T.F., Chen, C.M., Wu, S.Y., et al., 2016. Intratracheal administration of budesonide/
Ryter, S.W., Choi, A.M., 2013. Regulation of autophagy in oxygen-dependent cellular surfactant to prevent bronchopulmonary dysplasia. Am. J. Respir. Crit. Care Med.
stress. Curr. Pharm. Des. 19 (15), 2747–2756. 193 (1), 86–95.
Saeidnia, S., Abdollahi, M., 2013. Toxicological and pharmacological concerns on oxi- Yu, S., Shi, M., Liu, C., et al., 2015. Time course changes of oxidative stress and in-
dative stress and related diseases. Toxicol. Appl. Pharmacol. 273 (3), 442–455. flammation in hyperoxia-induced acute lung injury in rats. Iran. J. Basic Med. Sci. 18
Salaets, T., Richter, J., Brady, P., et al., 2015. Transcriptome analysis of the preterm (1), 98–103.
rabbit lung after seven days of hyperoxic exposure. PLoS One 10 (8), e0136569. Zhang, Y., Lingappan, K., 2017. Differential sex-specific effects of oxygen toxicity in
Saugstad, O.D., 2010. Oxygen and oxidative stress in bronchopulmonary dysplasia. J. human umbilical vein endothelial cells. Biochem. Biophys. Res. Commun. 486 (2),
Perinat. Med. 38 (6), 571–577. 431–437.
Saugstad, O.D., Speer, C.P., Halliday, H.L., 2011. Oxygen saturation in immature babies: Zhang, Q., Jianhua, F.U., Xue, X., 2014. Expression and function of aquaporin-1 in hy-
revisited with updated recommendations. Neonatology 100 (3), 217–218. peroxia-exposed alveolar epithelial type II cells. Exp. Ther. Med. 8 (2), 493–498.
Schieber, M., Chandel, N.S., 2014. ROS function in redox signaling and oxidative stress. Zhang, S., Patel, A., Moorthy, B., et al., 2015. Adrenomedullin deficiency potentiates
Curr. Biol. 24 (10), 453–462. hyperoxic injury in fetal human pulmonary microvascular endothelial cells. Biochem.
Seedorf, G.J., Metoxen, A.J., Rock, R., et al., 2016. Hepatocyte growth factor as a Biophys. Res. Commun. 464 (4), 1048.
downstream mediator of vascular endothelial growth factor-dependent preservation Zuo, L., Zhou, T., Pannell, B.K., et al., 2015. Biological and physiological role of reactive
of growth in the developing lung. Am. J. Physiol. Lung Cell. Mol. Physiol. 310 (11) oxygen species—the good, the bad and the ugly. Acta Physiol. 214 (3), 329–348.
(ajplung.00423.2015).

183

You might also like