ArticlePDF Available

Maternal Tn Immunization Attenuates Hyperoxia-Induced Lung Injury in Neonatal Rats Through Suppression of Oxidative Stress and Inflammation

Frontiers
Frontiers in Immunology
Authors:

Abstract and Figures

Hyperoxia therapy is often required to treat newborns with respiratory disorders. Prolonged hyperoxia exposure increases oxidative stress and arrests alveolar development in newborn rats. Tn antigen is N-acetylgalactosamine residue that is one of the most remarkable tumor-associated carbohydrate antigens. Tn immunization increases the serum anti-Tn antibody titers and attenuates hyperoxia-induced lung injury in adult mice. We hypothesized that maternal Tn immunizations would attenuate hyperoxia-induced lung injury through the suppression of oxidative stress in neonatal rats. Female Sprague–Dawley rats (6 weeks old) were intraperitoneally immunized five times with Tn (50 μg/dose) or carrier protein at biweekly intervals on 8, 6, 4, 2, and 0 weeks before the day of delivery. The pups were reared in room air (RA) or 2 weeks of 85% O2, creating the four study groups: carrier protein + RA, Tn vaccine + RA, carrier protein + O2, and Tn vaccine + O2. The lungs were excised for oxidative stress, cytokine, vascular endothelial growth factor (VEGF) and platelet-derived growth factor (PDGF) expression, and histological analysis on postnatal day 14. Blood was withdrawn from dams and rat pups to check anti-Tn antibody using western blot. We observed that neonatal hyperoxia exposure reduced the body weight, increased 8-hydroxy-2-deoxyguanosine (8-OHdG) expression and lung cytokine (interleukin-4), increased mean linear intercept (MLI) values, and decreased vascular density and VEGF and PDGF-B expressions. By contrast, Tn immunization increased maternal and neonatal serum anti-Tn antibody titers on postnatal day 14, reduced MLI, and increased vascular density and VEGF and PDGF-B expressions to normoxic levels. Furthermore, the alleviation of lung injury was accompanied by a reduction in lung cytokine and 8-OHdG expression. Therefore, we propose that maternal Tn immunization attenuates hyperoxia-induced lung injury in neonatal rats through the suppression of oxidative stress and inflammation.
This content is subject to copyright.
ORIGINAL RESEARCH
published: 04 April 2019
doi: 10.3389/fimmu.2019.00681
Frontiers in Immunology | www.frontiersin.org 1April 2019 | Volume 10 | Article 681
Edited by:
Pedro A. Reche,
Complutense University of Madrid,
Spain
Reviewed by:
Silvia Sánchez-Ramón,
Complutense University of Madrid,
Spain
Javier Carbone,
Hospital General Universitario
Gregorio Marañón, Spain
Francisco Javier Cubero,
Complutense University of Madrid,
Spain
*Correspondence:
Chung-Ming Chen
cmchen@tmu.edu.tw
Specialty section:
This article was submitted to
Vaccines and Molecular Therapeutics,
a section of the journal
Frontiers in Immunology
Received: 27 December 2018
Accepted: 12 March 2019
Published: 04 April 2019
Citation:
Chen C-M, Hwang J and Chou H-C
(2019) Maternal Tn Immunization
Attenuates Hyperoxia-Induced Lung
Injury in Neonatal Rats Through
Suppression of Oxidative Stress and
Inflammation. Front. Immunol. 10:681.
doi: 10.3389/fimmu.2019.00681
Maternal Tn Immunization
Attenuates Hyperoxia-Induced Lung
Injury in Neonatal Rats Through
Suppression of Oxidative Stress and
Inflammation
Chung-Ming Chen 1,2
*, Jaulang Hwang 3and Hsiu-Chu Chou 4
1Department of Pediatrics, Taipei Medical University Hospital, Taipei, Taiwan, 2Department of Pediatrics, School of Medicine,
College of Medicine, Taipei Medical University, Taipei, Taiwan, 3Taipei Cancer Center, Taipei Medical University, Taipei, Taiwan,
4Department of Anatomy and Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
Hyperoxia therapy is often required to treat newborns with respiratory disorders.
Prolonged hyperoxia exposure increases oxidative stress and arrests alveolar
development in newborn rats. Tn antigen is N-acetylgalactosamine residue that is
one of the most remarkable tumor-associated carbohydrate antigens. Tn immunization
increases the serum anti-Tn antibody titers and attenuates hyperoxia-induced lung
injury in adult mice. We hypothesized that maternal Tn immunizations would attenuate
hyperoxia-induced lung injury through the suppression of oxidative stress in neonatal
rats. Female Sprague–Dawley rats (6 weeks old) were intraperitoneally immunized five
times with Tn (50 µg/dose) or carrier protein at biweekly intervals on 8, 6, 4, 2, and 0
weeks before the day of delivery. The pups were reared in room air (RA) or 2 weeks
of 85% O2, creating the four study groups: carrier protein +RA, Tn vaccine +RA,
carrier protein +O2, and Tn vaccine +O2. The lungs were excised for oxidative
stress, cytokine, vascular endothelial growth factor (VEGF) and platelet-derived growth
factor (PDGF) expression, and histological analysis on postnatal day 14. Blood was
withdrawn from dams and rat pups to check anti-Tn antibody using western blot.
We observed that neonatal hyperoxia exposure reduced the body weight, increased
8-hydroxy-2-deoxyguanosine (8-OHdG) expression and lung cytokine (interleukin-4),
increased mean linear intercept (MLI) values, and decreased vascular density and
VEGF and PDGF-B expressions. By contrast, Tn immunization increased maternal and
neonatal serum anti-Tn antibody titers on postnatal day 14, reduced MLI, and increased
vascular density and VEGF and PDGF-B expressions to normoxic levels. Furthermore,
the alleviation of lung injury was accompanied by a reduction in lung cytokine and
8-OHdG expression. Therefore, we propose that maternal Tn immunization attenuates
hyperoxia-induced lung injury in neonatal rats through the suppression of oxidative stress
and inflammation.
Keywords: hyperoxia, vaccine, interleukin-4, 8-hydroxy-2-deoxyguanosine, mean linear intercept,
von Willebrand factor
Chen et al. Immunization and Hyperoxia-Induced Lung Injury
INTRODUCTION
Respiratory distress syndrome is a major cause of morbidity
and mortality in preterm neonates (1). Hyperoxia therapy is
often required to treat newborns with respiratory disorders.
However, supplemental oxygen administered to newborn infants
with respiratory failure increases oxidant stress and leads to
lung injury. The rat model is appropriate to study the effects of
hyperoxia on preterm infants with respiratory distress because
rats are born at the saccular stage, equivalent to an 30 week
human gestation (2). Prolonged exposure of neonatal rodents to
hyperoxia resulted in decreased alveolar septation and increased
terminal air space size, similar to human bronchopulmonary
dysplasia (3,4). Despite early surfactant therapy, optimal
ventilation strategies, and increased use of noninvasive positive
pressure ventilation, bronchopulmonary dysplasia remains a
major cause of morbidity and mortality during the first year
of life, and many infants experience significant respiratory
morbidity, including decreased response to acute hypoxia,
increased airway reactivity, and development of obstructive
airway disease throughout childhood (57). No effective clinical
therapy is currently available to prevent the development and
long-term pulmonary sequelae of bronchopulmonary dysplasia.
Tn antigen is N-acetylgalactosamine residue that is α-
linked to a serine or threonine residue, which is one of
the most remarkable tumor-associated carbohydrate antigens,
often offered to mucin-type carbohydrates (8). Studies have
demonstrated that inflammatory cytokines can promote glycan
epitope by regulating specific glycosyltransferases (9,10). Using
the linear array epitope technology, Chiang et al. developed
an anti-Tn vaccine that induces anti-Tn antibodies with high
specificity and high affinity in mice (11). These results suggest
that Tn may show immunogenicity and protection in preclinical
animal studies. Tn immunization attenuates hyperoxia-induced
lung injury in adult mice by inhibiting the nuclear factor-kappa
B (NF-κB) activity (12). The effects of Tn immunization on
neonatal hyperoxia-induced lung injury are unknown. Therefore,
we hypothesize that the maternal Tn immunizations would
attenuate hyperoxia-induced lung injury in neonatal rats. This
study investigated the protective effects and mechanisms of
Tn immunization on lung inflammation and development in
neonatal rats exposed to hyperoxia.
MATERIALS AND METHODS
Tn Vaccine Preparation
Tn vaccine was prepared by conjugating Tn to a novel carrier
protein as described previously (11). Tn was conjugated to
mFc(Cys42)Histag2 or GST(Cys6)Histag2 at a glycotope–carrier
protein weight ratio of 5:1. The conjugation was performed
in a buffer containing 20 mM sodium phosphate, pH 7.9,
8 M urea, 500 mM imidazole, and 0.2 mM tris(2-carboxyethyl)
phosphine (TCEP). After 48 h, the conjugate was refolded in
phosphate-buffered saline (PBS) with 0.2 mM TCEP. GST(Cys6)
was dialyzed against PBS with 0.2 mM TCEP. Different
glycotopes and a linker (N-succinimidyl-6-maleimidocaproate)
were conjugated to GST(Cys6) at 4C for 48 h.
Animal Model and Experimental Groups
Female Sprague–Dawley rats (6 weeks old) were obtained from
BioLASCO Taiwan Co., Ltd and were housed in individual
cages with 12-h light–dark cycles. Laboratory food and water
were available ad libitum. The female rats were randomly
assigned to the Tn immunization or control treatment groups
(Supplement Figure 1). The Tn immunization strategy consisted
of an intraperitoneal injection of Tn (50 µg/dose) in 0.5 mL
normal saline, and the control immunization consisted of the
intraperitoneal injection of the same volume of carrier protein.
The immunizations were administered five times at biweekly
intervals on 8, 6, 4, 2, and 0 weeks before the day of delivery.
Female rats in estrus or proestrus were placed in cages with adult
male rats (two females for each male) for 12 h. The following
morning, mating was confirmed by the presence of a vaginal
plug, which was considered day 0 of gestation. The dams were
allowed to deliver vaginally at term. Within 12 h of birth, litters
were pooled and randomly redistributed to the newly delivered
mothers, and the pups were then randomly assigned to room air
(RA) or oxygen-enriched atmosphere (O2) treatment. The pups
in the O2treatment subgroups were reared in an atmosphere
containing 85% O2from postnatal days 1 to 14. The pups in the
control subgroups were reared in normobaric RA for 14 days.
Four study groups were obtained as follows: carrier protein +
RA, Tn vaccine +RA, carrier protein +O2, and Tn vaccine
+O2. To avoid oxygen toxicity in the nursing mothers, they
were rotated between the O2treatment and RA control litters
every 24 h. An oxygen-rich atmosphere was maintained in a
transparent 40 ×50 ×60-cm plexiglass chamber receiving
O2continuously at 4 L/min. Oxygen levels were monitored
using a ProOx P110 monitor (NexBiOxy, Hsinchu, Taiwan). On
postnatal day 14, pups from each group were deeply anesthetized
with an overdose of isoflurane, and body and lung weights were
noted. Blood was withdrawn from dams and rat pups to check
anti-Tn antibody using western blot. The study protocol was
approved by the Institutional Animal Care and Use Committee
of Taipei Medical University.
Western Blot Analysis of the Serum
Anti-Tn Antibody
Solubilized proteins were separated using SDS-PAGE and
were electrophoretically transferred to polyvinylidene difluoride
(PVDF) membranes. PVDF membranes were rinsed in TBS
buffer and blocked for 1 h with TBS buffer containing 5% skim
milk. After washing with TBST (Tris-buffered saline, 0.1% Tween
20), the PVDF membranes were incubated overnight with rat
serum (1:1000) dissolved in antibody buffer. After multiple
washing with TBST, the membranes were incubated for 45 min
with Jackson AffiniPure Donkey AntiRat IgG (H+L) (1:5000,
Jackson ImmunoResearch Laboratories, Inc., West Grove, PA,
USA). Membranes were washed, and immunoreactive proteins
were detected using Immun-Star assay kit (Bio-Rad) following
the manufacturer’s suggestions.
Lung Histology
To standardize analysis, sections were obtained from the right
middle lobe of the right lung. The lung tissue was immersed
Frontiers in Immunology | www.frontiersin.org 2April 2019 | Volume 10 | Article 681
Chen et al. Immunization and Hyperoxia-Induced Lung Injury
with 4% paraformaldehyde in 0.1 M phosphate buffer (pH 7.4)
at 4C for 24 h. The tissues were then dehydrated in alcohol,
cleared in xylene, and embedded in paraffin. Five-micrometer
sections were stained with hematoxylin and eosin, examined
using light microscopy, and assessed for lung morphometry.
Mean linear intercept (MLI), an indicator of mean alveolar
diameter, was assessed in 10 nonoverlapping fields (13). Vascular
density was determined with von Willebrand factor (vWF)
immunohistochemistry reaction (see below).
Immunohistochemistry of
8-Hydroxy-2-Deoxyguanosine, von
Willebrand Factor, Vascular Endothelial
Growth Factor, Platelet-Derived Growth
Factor-B, Inducible Nitric Oxide Synthase,
and YM-1
Immunohistochemical staining was performed on 5-µm paraffin
sections using immunoperoxidase visualization. After routine
deparaffinization, heat-induced epitope retrieval was performed
by immersing the slides in 0.01 M sodium citrate buffer
(pH 6.0). To block the endogenous peroxidase activity and
nonspecific binding of antibodies, the sections were preincubated
for 1 h at room temperature in 0.1 M PBS containing 10%
normal goat serum and 0.3% H2O2. The sections were then
incubated for 20 h at 4C with mouse monoclonal anti-8-
hydroxy-2-deoxyguanosine (8-OHdG) antibody (1:100; Abcam
Inc., Cambridge, MA, USA), rabbit polyclonal antivWF antibody
(1:100; Abcam), rabbit polyclonal antivascular endothelial
growth factor (VEGF) antibody (1:50; Santa Cruz Biotechnology,
Inc., CA, USA), rabbit polyclonal antiplatelet-derived growth
factor (PDGF)-B antibody (1:50; Santa Cruz Biotechnology, Inc.),
rabbit polyclonal anti-inducible nitric oxide synthase (iNOS)
(1:100; Thermo Fisher Scientific, Rackford, IL, USA), or rabbit
polyclonal anti-Ym-1 (1:25; STEMCELL Technologies Inc.,
Vancouver, Canada) as primary antibodies. The sections were
then treated for 1 h at 37C with biotinylated goat anti-mouse
or rabbit IgG (1:200, Jackson ImmunoResesarch Laboratories
Inc., PA, USA). Following the reaction produced using reagents
from an avidin–biotin complex kit (Vector Laboratories, Inc.,
CA, USA), the reaction products were visualized using a
diaminobenzidine substrate kit (Vector Laboratories, Inc.)
according to the recommendations of the manufacturer. The
8-OHdG staining was quantified by considering the positively
stained nuclei per high-power field. Positively stained cells
were counted in five fields randomly selected from each
section using a light microscope (magnification: ×400), and
results were expressed as the percentage of positively stained
nuclei to total cells. Microvessel density was determined by
counting the vessels with the positive vWF stained in an
unbiased manner and a minimum of four random lung fields
at ×400 magnifications (14). The automatic object counting and
measuring process was used to quantify the immunoreactivity in
the sections. This generated a percentage of positively stained
cells, and the values were expressed as a labeling index (%).
The positive immunostaining of lung parenchyma for iNOS and
Ym-1 were measured at ×400 magnification by the density of
immunostained chromogen (0.1 mm2) using the Image Pro Plus
(Media Cybernetics, Silver Spring, USA).
Cytokine Assay
Approximately 100 mg of lung tissue from each pup was
homogenized, sonicated, and centrifuged at 500 ×g for 20 min
at 4C to remove cellular debris according to the manufacturer’s
instructions. The levels of interleukin-4 (IL-4) in the supernatants
were determined using the enzyme-linked immunosorbent assay
kit (MyBioSource, San Diego, CA, USA) as IL-4 expression
was significantly increased in the lungs from hyperoxia-exposed
neonatal rats and mice (15,16).
Western Blot Analysis of Growth Factors
Lung tissues were homogenized in ice-cold buffer containing
50 mM Tris·HCl (pH 7.5), 1 mM EGTA, 1 mM EDTA, and
protease inhibitor cocktail (complete minitablets; Roche,
Mannheim, Germany). The samples were sonicated and then
centrifuged at 500 gfor 20 min at 4C to remove cellular debris.
Proteins (30 µg) were resolved on 12% SDS-PAGE under
reducing conditions and electroblotted to a PVDF membrane
(ImmobilonP, Millipore, Bedford, MA, USA). After blocking
with 5% nonfat dry milk, the membranes were incubated with
antibody against VEGF (1:1000; Santa Cruz Biotechnology,
Inc.), PDGF-B (1:1000; Santa Cruz Biotechnology, Inc.), or
anti-β-actin (1:20,000; Sigma-Aldrich, St. Louis, MO, USA)
and subsequently with horseradish peroxidase-conjugated
goat anti-rabbit IgG or anti-mouse IgG (Pierce Biotechnology,
Rockford, USA). Protein bands were detected using SuperSignal
Substrate from Pierce. Densitometric analysis was performed
to measure the intensity of VEGF, PDGF-B, and β-actin bands
using AIDA software.
Statistical Analysis
All data were presented as mean ±SD. Statistical analyses
were performed using a two-way analysis of variance with a
Bonferroni post hoc test for multiple group comparisons. The
survival rate was evaluated using the Kaplan–Meier method, and
log-rank test was used for intergroup comparisons. Differences
were considered statistically significant when p<0.05.
RESULTS
Three Tn immunization-treated and three carrier protein-treated
female rats were successfully mated with male rats. Six dams
gave birth to a total of 46 pups; 23 pups each were randomly
distributed to the RA and hyperoxia groups. A total of 11 and
12 pups received carrier protein and Tn immunization in the
RA groups, and 11 and 12 pups received carrier protein and Tn
immunization in the hyperoxia groups.
Western Blot Analysis of Serum
Anti-Tn Antibody
PE-(PC7)Tn recognized predominantly one major band
(anti-Tn antibody), which was not recognized by PE-(PC7)
(Figure 1). Mothers and pups receiving Tn immunization
exhibited a dense anti-Tn antibody band, whereas mothers
Frontiers in Immunology | www.frontiersin.org 3April 2019 | Volume 10 | Article 681
Chen et al. Immunization and Hyperoxia-Induced Lung Injury
FIGURE 1 | Western blot analysis of serum anti-Tn antibody in dams and rat pups on postnatal day 14. PE-(PC7)Tn predominantly recognized one major band
(anti-Tn antibody), which was not recognized by PE-(PC7). Mothers and pups who received Tn immunization exhibited a dense anti-Tn antibody band, whereas
mothers and pups who received carrier protein immunization did not exhibit anti-Tn antibody. The diagram illustrates representative data from three experiments.
and pups receiving carrier protein immunization did not
exhibit anti-Tn antibody.
Survival
The rats reared in the carrier protein +RA or Tn vaccine
+RA group all survived (Figure 2). The rats reared in
the carrier protein +O2or Tn vaccine +O2group
exhibited a lower survival rate after postnatal day 7.
On postnatal day 14, the survival rate between the rats
treated with the carrier protein or Tn immunization
were comparable.
Body Weight, Lung Weight, and
Lung-To-Body Weight Ratios
The rats in the carrier protein +O2or Tn vaccine +O2
group exhibited significantly lower body and lung weights
on postnatal day 14 than those reared in the carrier protein
+RA or Tn vaccine +RA group (Table 1). Maternal
Tn immunization increased the body weight on postnatal
day 14 in rats reared in RA or hyperoxia. The rats in
the carrier protein +O2group exhibited a significantly
higher lung-to-body weight ratio than those in the other
three groups.
Immunohistochemistry for 8-OHdG
To investigate whether maternal Tn immunization reduced
oxidative stress in neonatal hyperoxia-exposed rat lungs, we
used immunohistochemical assays for oxidative stress marker 8-
OHdG. The 8-OHdG immunoreactivity was primarily detected
in the epithelial cells (Figure 3A). The rats in the carrier protein
+O2group exhibited significantly higher number of positive 8-
OHdG cells than those in the carrier protein +RA or Tn vaccine
TABLE 1 | Body weights, lung weights, and lung-to-body weight ratios of rat
pups on postnatal day 14.
Treatment nBody weight (g) Lung weight (g) Lung-to-
body weight
ratio (%)
Carrier protein +RA 11 20.36 ±1.43 0.33 ±0.02 1.60 ±0.12
Carrier protein +O29 15.11 ±1.58a0.31 ±0.05 2.08 ±0.48c
Tn vaccine +RA 12 23.67 ±2.12 0.36 ±0.02 1.55 ±0.11
Tn vaccine +O210 19.35 ±0.82a,b 0.33 ±0.02 1.71 ±0.11
Values are mean ±SD.
ap<0.001 vs. carrier protein +RA and Tn vaccine +RA.
bp<0.001 vs. carrier protein +O2.
cp<0.05 vs. carrier protein +RA, Tn vaccine +RA, and Tn vaccine +O2.
+RA group. Maternal Tn immunization significantly decreased
the neonatal hyperoxia-induced increase in the number of
positive 8-OHdG cells (Figure 3B).
Cytokine Level
The rats in the carrier protein +O2group exhibited
a significantly higher lung IL-4 level than those in
the carrier protein +RA or Tn vaccine +RA group
(Figure 3C). Maternal Tn immunization significantly
decreased the neonatal hyperoxia-induced increase in the lung
IL-4 levels.
Histology Results
Representative lung sections stained with hematoxylin and
eosin and vWF from maternal Tn immunization and postnatal
hyperoxia-exposed rats on postnatal day 14 are shown in
Figures 4A,B, respectively. The rats in the carrier protein +
Frontiers in Immunology | www.frontiersin.org 4April 2019 | Volume 10 | Article 681
Chen et al. Immunization and Hyperoxia-Induced Lung Injury
FIGURE 2 | Effects of Tn immunization on the survival rate on postnatal day 14. The rats in the carrier protein +RA or Tn vaccine +RA group all survived. The rats in
the carrier protein +O2or Tn vaccine +O2group exhibited a lower survival rate after postnatal day 7. On postnatal day 14, the survival rate between the rats treated
with carrier protein or Tn immunization were comparable.
FIGURE 3 | (A) Representative immunohistochemistry of 8-hydroxy-2-deoxyguanosine (8-OHdG), (B) quantitative analysis of 8-OHdG-positive cells, and (C) lung
IL-4 in 14-day-old rats in the carrier protein +RA, Tn vaccine +RA, carrier protein +O2, or Tn vaccine +O2group. Positive staining is indicated in brown (arrow).
The rats in the carrier protein +O2group exhibited a significantly higher number of positive 8-OHdG cells and IL-4 levels than those in the carrier protein +RA or Tn
vaccine +RA group. Maternal Tn immunization significantly decreased the hyperoxia-induced increase in the number of positive 8-OHdG cells and IL-4 levels. *p<
0.05, ***p<0.001.
O2group exhibited a significantly higher MLI and lower
vascular density than those in the carrier protein +RA or Tn
vaccine +RA group. Maternal Tn immunization improved the
hyperoxia-induced alteration in the MLI and vascular density to
normoxic levels.
Immunohistochemistry and Western
Blotting of VEGF and PDGF-B
Representative immunohistochemistry of VEGF and PDGF-B
are shown in Figures 5A,B, respectively. The VEGF and PDGF-B
immunoreactivities were primarily detected in the endothelial
Frontiers in Immunology | www.frontiersin.org 5April 2019 | Volume 10 | Article 681
Chen et al. Immunization and Hyperoxia-Induced Lung Injury
FIGURE 4 | Representative H&E stained lung sections for histology observation and (A) mean linear intercept assessment and (B) immunohistochemistry of vWF and
semiquantitative analysis for vascular density in lung of 14-day-old rats in the carrier protein +RA, Tn vaccine +RA, carrier protein +O2, or Tn vaccine +O2group.
The rats in the carrier protein +O2group exhibited a significantly higher MLI and lower vascular density than those in the carrier protein +RA or Tn vaccine +RA
group. Maternal Tn immunization reversed the MLI and vascular density to normoxic levels. **p<0.01, ***p<0.001.
and epithelial cells. The rats in the carrier protein +O2
group exhibited significantly lower VEGF and PDGF-B protein
expression than those in the carrier protein +RA or Tn vaccine
+RA group. Maternal Tn immunization significantly increased
the hyperoxia-induced decrease in the VEGF and PDGF-B
protein expression.
M1/M2 Phenotype in Macrophages
Representative lung sections stained with iNOS (M1 macrophage
maker) and Ym1 (M2 macrophage maker) from maternal Tn
immunization and postnatal hyperoxia-exposed rats on postnatal
day 14 are shown in Figures 6A,B, respectively. The rats in the
carrier protein +O2group exhibited a significantly higher M1
phenotype macrophages and lower M2 phenotype macrophages
than those in the carrier protein +RA or Tn vaccine +
RA group (Figures 6C,D). Maternal Tn immunization reversed
the hyperoxia-induced M1/M2 macrophage polarization to
normoxia levels.
DISCUSSION
Our in vivo model revealed that maternal Tn immunization
increased maternal and neonatal serum antibody titers and
attenuated hyperoxia-induced lung injury in newborn rats, as
evidenced by reversing hyperoxia-induced increase in MLI and
decrease in vascular density and growth factors. The alleviation
of lung injury was associated with a reduction in cytokine
and 8-OHdG expression. Therefore, we proposed that maternal
Tn immunization attenuates hyperoxia-induced lung injury
in neonatal rats through the suppression of oxidative stress
and inflammation.
Tn antigen is a pan-carcinoma antigen, expressed on breast,
pancreas, colon, lung, and bladder carcinomas, being less
common in hematological malignancies (17,18). Tn is associated
with immune disorders in addition to cancers. Tn antigen can
be detected on chronic inflammatory tissues in patients with
rheumatoid arthritis and osteoarthritis (19). Tn can induce
tumor-specific IgG antibodies in mice and in nonhuman
primates under appropriate conditions (20). These findings
revealed that Tn might be an essential component in the
design of humoral-mediated vaccines and suggested that Tn
may show immunogenicity and protection in preclinical animal
studies. Tn immunization increased serum anti-Tn antibody
titers and protected against hyperoxia-induced lung injury in
adult mice through the inhibition of NF-κB activity (12).
Therefore, maternal Tn immunization may attenuate hyperoxia-
induced lung injury in neonatal mice through suppression
of inflammation.
Frontiers in Immunology | www.frontiersin.org 6April 2019 | Volume 10 | Article 681
Chen et al. Immunization and Hyperoxia-Induced Lung Injury
FIGURE 5 | Representative immunohistochemistry and representative western blots and quantitative data for (A) VEGF and (B) PDGF-B protein expression in
14-day-old rats in the carrier protein +RA, Tn vaccine +RA, carrier protein +O2, or Tn vaccine +O2group. The rats in the carrier protein +O2group exhibited
significantly lower vascular density and VEGF and PFGF-B expression than those in the carrier protein +RA or Tn vaccine +RA group. Maternal Tn immunization in
hyperoxia-exposed rats improved vascular density and VEGF and PDGF-B expression to normoxic levels. *p<0.05, **p<0.01.
Maternal immunization provides protection to the newborns
through the transfer of vaccine-induced IgG across the placenta.
IgG is the only antibody class that significantly crosses the human
placenta. Coder et al. maternally administered radiolabeled
humanized IgG2 and found humanized IgG2 in rat embryo/fetal
tissues as early as gestation day 11 with a >1,000-fold increase
in the amount of total IgG2 by gestation day 21 (21). The
concentration of IgG2 in rat embryo/fetal tissues generally
remained unchanged from gestation day 11–17 with a slight
increase from day 17–21. Moffat et al. compared IgG2X
embryonic exposure in rats and found that fetal IgG2X plasma
concentrations increased more than six-fold from gestation
days 16–21 (22). In this study, we immunized the female
rats three times before gestation, on gestation day 7, and
at delivery and observed that Tn immunization increased
serum anti-Tn antibody titers and protected against hyperoxia-
induced lung injury in neonatal rats. These results indicate
that maternal immunization is a potential strategy to prevent
and treat neonatal diseases. Further studies are needed to
determine total IgG and complement factors to elucidate the
immunological mechanisms that mediate the beneficial effects of
maternal immunization.
Hyperoxia exposure for 7 days increased oxidative stress
in the neonatal murine lungs (23,24). 8-OHdG is a DNA
base-modified product generated by reactive oxygen species as
a marker of oxidative DNA damage and its levels in target
tissues are correlated with other oxidative stress markers (25
27). The expression of 8-OHdG reflects the oxidative stress
level in the lung tissues and its expression was elevated in the
hyperoxia-exposed neonatal rat lung tissue and primary cultured
neonatal rat alveolar epithelial type II cells compared with the
normoxic controls (28). Positive signals for 8-OHdG increased
in the hyperoxia-exposed rats, and signals were mainly found
in the nuclei of epithelial cells. Maternal Tn immunization
significantly decreased the hyperoxia-induced increase in the
number of positive 8-OHdG cells. These results suggest that anti-
Tn antibody-suppressed oxidative stress formation and support
that anti-oxidant enzymes are effective in reducing hyperoxia-
induced neonatal lung injury (24,29).
VEGF is a potent endothelial cell mitogen that regulates
angiogenesis and alveolar development (30). PDGF is crucial
for alveolarization of the normally developing lung (31). We
determined VEGF and PDGF-B expression as their mRNA
and protein expression was decreased in the hyperoxia-exposed
Frontiers in Immunology | www.frontiersin.org 7April 2019 | Volume 10 | Article 681
Chen et al. Immunization and Hyperoxia-Induced Lung Injury
FIGURE 6 | (A,B) Representative immunohistochemistry and (C,D) quantitative analysis of iNOS and Ym1 positive cells in 14-day-old rats in the carrier protein +RA,
Tn vaccine +RA, carrier protein +O2, or Tn vaccine +O2group. The rats in the carrier protein +O2group exhibited a significantly higher M1 phenotype (iNOS)
macrophages (arrow) and lower M2 phenotype (Ym-1) macrophages than those in the carrier protein +RA or Tn vaccine +RA group. Maternal Tn immunization
reversed the hyperoxia-induced M1/M2 macrophage polarization to normoxia levels. **p<0.01, ***p<0.001.
neonatal mice and piglet lungs (32,33). In this study, we
demonstrated that rats in the carrier protein +O2group
exhibited significantly decreased VEGF and PDGF-B expression
than those in the carrier protein +RA or Tn vaccine +RA group.
Maternal Tn immunization significantly reversed the hyperoxia-
induced decrease in VEGF and PDGF-B to normoxic levels.
These results suggest that maternal Tn immunization enhanced
vascular and alveolar development through the induction of
growth factors in neonatal rats.
In addition to the traditional host defense, inflammation,
and scavenging functions, macrophages have broader
functions, including vital roles in tissue repair and organ
development (3436). Animal studies have demonstrated
that neonatal hyperoxia exposure increases macrophage
infiltration into alveolar airspaces (37,38). In this study, we used
immunohistochemistry to detect the macrophage infiltration
on the lung tissue sections. Macrophage phenotype was
assessed through immunostaining for iNOS (M1 macrophage
marker) and Ym1 (M2 macrophage marker) as hyperoxia
increases iNOS expression in macrophages and hyperoxia-
exposed murine lungs and inhibits the M2 phenotype in
macrophages (39,40). The rats in the carrier protein +O2
group significantly exhibited more M1 macrophages than those
in the carrier protein +RA or Tn vaccine +RA group. These
results indicate that hyperoxia promotes the M1 phenotype
in macrophages and suggest that the M1/M2 polarization
mediates the pulmonary effects of hyperoxia in the developing
lung. Our study increases the understanding of the role of
macrophage polarization in hyperoxia-induced injury to the
developing lungs.
In conclusion, we observed that Tn immunization increases
serum anti-Tn antibody titers in mothers and neonates, inhibits
lung inflammation and oxidative stress, and enhances lung
development in the neonatal hyperoxia-exposed rats. These
findings indicate that Tn activation may be involved in the
mechanism of proinflammatory cytokine release and lung
injury and suggest that the Tn vaccine may be a promising
treatment modality against hyperoxia-induced lung injury in
neonates. Future studies are necessary to evaluate the direct
therapeutic effects of anti-Tn antibody on hyperoxia-induced
lung injury.
ETHICS STATEMENT
Animal care and experimental procedures were performed
in accordance with the guidelines of the Laboratory Animal
Care Committee of Taipei Medical University (LAC-2017-
0291). Sprague–Dawley rats (6 weeks old) were obtained
Frontiers in Immunology | www.frontiersin.org 8April 2019 | Volume 10 | Article 681
Chen et al. Immunization and Hyperoxia-Induced Lung Injury
from BioLASCO Taiwan Co., Ltd and were maintained
in a pathogen-free facility and air-conventional animal
housing on a 12-h light/dark cycle. The care and housing
of experimental animals were approved in accordance with the
guidelines of the Laboratory Animal Care Committee of Taipei
Medical University.
AUTHOR CONTRIBUTIONS
C-MC and JH: designed and performed the experiments; C-MC,
JH, and H-CC: analysis and interpretation of data and drafted
and approved the manuscript.
FUNDING
This study received grants from the Ministry of Science and
Technology in Taiwan (107-2314-B-038-060-MY2).
SUPPLEMENTARY MATERIAL
The Supplementary Material for this article can be found
online at: https://www.frontiersin.org/articles/10.3389/fimmu.
2019.00681/full#supplementary-material
Supplement Figure 1 | Experimental design of the study timeline and rat
treatment groups.
REFERENCES
1. Ramanathan R, Bhatia JJ, Sekar K, Ernst FR. Mortality in preterm
infants with respiratory distress syndrome treated with poractant alfa,
calfactant or beractant: a retrospective study. J Perinatol. (2013) 33:119–25.
doi: 10.1038/jp.2011.125
2. O’Reilly M, Thébaud B. Animal models of bronchopulmonary dysplasia. The
term rat models. Am J Physiol Lung Cell Mol Physiol. (2014) 307:L948e58.
doi: 10.1152/ajplung.00160.2014
3. Manji JS, O’Kelly CJ, Leung WI, Olson DM. Timing of hyperoxic
exposure during alveolarization influences damage mediated by
leukotrienes. Am J Physiol Lung Cell Mol Physiol. (2001) 281:L799–806.
doi: 10.1152/ajplung.2001.281.4.L799
4. Chen CM, Wang LF, Chou HC, Lan YD, Yi-Ping Lai. Up-regulation of
connective tissue growth factor in hyperoxia-induced lung fibrosis. Pediatr
Res. (2007) 62:128–33. doi: 10.1203/PDR.0b013e3180987202
5. Gien J, Kinsella JP. Pathogenesis and treatment of
bronchopulmonary dysplasia. Curr Opin Pediatr. (2011) 23:305–13.
doi: 10.1097/MOP.0b013e328346577f
6. Jacob SV, Coates AL, Lands LC, MacNeish CF, Riley SP, Hornby L, et al. Long-
term pulmonary sequelae of severe bronchopulmonary dysplasia. J Pediatr.
(1998) 133:193–200. doi: 10.1016/S0022-3476(98)70220-3
7. Mouradian GC Jr, Alvarez-Argote S, Gorzek R, Thuku G, Michkalkiewicz T,
Wong-Riley MTT, et al. Acute and chronic changes in the control of breathing
in a rat model of bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol
Physiol. (2019) 316:L506–L518. doi: 10.1152/ajplung.00086.2018
8. Zhang X, Issagholian A, Berg EA, Fishman JB, Nesburn AB, BenMohamed
L. Th-cytotoxic T-lymphocyte chimeric epitopes extended by Nepsilon-
palmitoyl lysines induce herpes simplex virus type 1-specific effector CD8+
Tc1 responses and protect against ocular infection. J Virol. (2005) 79:15289–
301. doi: 10.1128/JVI.79.24.15289-15301.2005
9. Bassaganas S, Allende H, Cobler L, Ortiz MR, Llop E, de Bolos C, et al.
Inflammatory cytokines regulate the expression of glycosyltransferases
involved in the biosynthesis of tumor-associated sialylated glycans
in pancreatic cancer cell lines. Cytokine. (2015) 75:197–206.
doi: 10.1016/j.cyto.2015.04.006
10. Padro M, Mejias-Luque R, Cobler L, Garrido M, Perez-Garay M, Puig S,
et al. Regulation of glycosyltransferases and Lewis antigens expression by IL-
1beta and IL-6 in human gastric cancer cells. Glycoconj J. (2011) 28:99–110.
doi: 10.1007/s10719-011-9327-4
11. Chiang HL, Lin CY, Jan FD, Lin YS, Hsu CT, Whang-Peng J, et al. A novel
synthetic bipartite carrier protein for developing glycotope-based vaccines.
Vaccine. (2012) 30:7573–81. doi: 10.1016/j.vaccine.2012.10.041
12. Chen CM, Hwang J, Chou HC, Shiah HS. Tn (N-acetyl-d-galactosamine-
O-serine/threonine) immunization protects against hyperoxia-induced lung
injury in adult mice through inhibition of the nuclear factor kappa B activity.
Int Immunopharmacol. (2018) 59:261–8. doi: 10.1016/j.intimp.2018.04.022
13. Chou HC, Li YT, Chen CM. Human mesenchymal stem cells attenuate
experimental bronchopulmonary dysplasia induced by perinatal
inflammation and hyperoxia. Am J Transl Res. (2016) 8:342–53.
14. Irwin D, Helm K, Campbell N, Imamura M, Fagan K, Harral J, et al. Neonatal
lung side population cells demonstrate endothelial potential and are altered
in response to hyperoxia-induced lung simplification. Am J Physiol Lung Cell
Mol Physiol. (2007) 293:L941–51. doi: 10.1152/ajplung.00054.2007
15. Liu DY, Jiang T, Wang S, Cao X. Effect of hyperoxia on pulmonary SIgA
and its components, IgA and SC. J Clin Immunol. (2013) 33:1009–17.
doi: 10.1007/s10875-013-9891-4
16. Cheon IS, Son YM, Jiang L, Goplen NP, Kaplan MH, Limper AH,
et al. Neonatal hyperoxia promotes asthma-like features through IL-33-
dependent ILC2 responses. J Allergy Clin Immunol. (2018) 142:1100–12.
doi: 10.1016/j.jaci.2017.11.025
17. Desai P. Immunoreactive T and Tn antigens in malignancy: role in carcinoma
diagnosis, prognosis, and immunotherapy. Transfus Med Rev. (2000) 14:312–
25. doi: 10.1053/tmrv.2000.16229
18. Springer GF. Immunoreactive T and Tn epitopes in cancer diagnosis,
prognosis, and immunotherapy. J Mol Med. (1997) 75:594–602.
doi: 10.1007/s001090050144
19. Ishino H, Kawahito Y, Hamaguchi M, Takeuchi N, Tokunaga D, Hojo T, et al.
Expression of Tn and sialyl Tn antigens in synovial tissues in rheumatoid
arthritis. Clin Exp Rheumatol. (2010) 28:246–9.
20. Grigalevicius S, Chierici S, Renaudet O, Lo-Man R, Dériaud E, Leclerc
C, et al. Chemoselective assembly and immunological evaluation of
multiepitopic glycoconjugates bearing clustered Tn antigen as synthetic
anticancer vaccines. Bioconjug Chem. (2005) 16:1149–59. doi: 10.1021/bc05
0010v
21. Coder PS, Thomas JA, Stedman DB, Bowman CJ. Placental transfer
of 125Iodinated humanized immunoglobulin G21a in the Sprague
Dawley rat. Reprod Toxicol. (2013) 38:37–46. doi: 10.1016/j.reprotox.2013.
02.007
22. Moffat GJ, Retter MW, Kwon G, Loomis M, Hock MB, Hall C, et al. Placental
transfer of a fully human IgG2 monoclonal antibody in the cynomolgus
monkey, rat, and rabbit: a comparative assessment from during organogenesis
to late gestation. Birth Defects Res B Dev Reprod Toxicol. (2014) 101:178–88.
doi: 10.1002/bdrb.21105
23. Bouch S, O’Reilly M, Harding R, Sozo F. Neonatal exposure to mild hyperoxia
causes persistent increases in oxidative stress and immune cells in the lungs
of mice without altering lung structure. Am J Physiol Lung Cell Mol Physiol.
(2015) 309:L488–96. doi: 10.1152/ajplung.00359.2014
24. ValenciaAM, Abrantes MA, Hasan J, Aranda JV, Beharry KD. Reactive oxygen
species, biomarkers of microvascular maturation and alveolarization, and
antioxidants in oxidative lung Injury. React Oxyg Species. (2018) 6:373–88.
doi: 10.20455/ros.2018.867
25. Yamagami K, Yamamoto Y, Kume M, Ishikawa Y, Yamaoka Y, Hiai H,
et al. Formation of 8-hydroxy-2’-deoxyguanosine and 4-hydroxy-2-nonenal-
modified proteins in rat liver after ischemia–reperfusion: distinct localization
of the two oxidatively modified products. Antioxid Redox Signal. (2000)
2:127–36. doi: 10.1089/ars.2000.2.1-127
26. Kasai H. Analysis of a form of oxidative DNA damage, 8-hydroxy-2-
deoxyguanosine, as a marker of cellular oxidative stress during carcinogenesis.
Mutat Res. (1997) 387:146–63. doi: 10.1016/S1383-5742(97)00035-5
Frontiers in Immunology | www.frontiersin.org 9April 2019 | Volume 10 | Article 681
Chen et al. Immunization and Hyperoxia-Induced Lung Injury
27. Pérez DD, Strobel P, Foncea R, Díez MS, Vásquez L, Urquiaga I, et al. Wine,
diet, antioxidant defenses, and oxidative damage. Ann N Y Acad Sci. (2002)
957:136–45. doi: 10.1111/j.1749-6632.2002.tb02912.x
28. Jin L, Yang H, Fu J, Xue X, Yao L, Qiao L. Association between oxidative
DNA damage and the expression of 8-oxoguanine DNA glycosylase 1 in lung
epithelial cells of neonatal rats exposed to hyperoxia. Mol Med Rep. (2015)
11:4079–86. doi: 10.3892/mmr.2015.3339
29. Pasha AB, Chen XQ, Zhou GP. Bronchopulmonary dysplasia: pathogenesis
and treatment. Exp Ther Med. (2018) 16:4315–21. doi: 10.3892/etm.2018.6780
30. Yun EJ, Lorizio W, Seedorf G, Abman SH, Vu TH. VEGF and
endothelium-derived retinoic acid regulate lung vascular and alveolar
development. Am J Physiol Lung Cell Mol Physiol. (2016)310:L287–98.
doi: 10.1152/ajplung.00229.2015
31. Lindahl P, Boström H, Karlsson L, Hellström M, Kalen M, Betsholtz C. Role
of platelet-derived growth factors in angiogenesis and alveogenesis. Curr Top
Pathol. (1999) 93: 27–33.
32. Perveen S, Patel H, Arif A, Younis S, Codipilly CN, Ahmed
M. Role of EC-SOD overexpression in preserving pulmonary
angiogenesis inhibited by oxidative stress. PLoS ONE. (2012) 7:e51945.
doi: 10.1371/journal.pone.0051945
33. Zhang X, Reinsvold P, Thibeault DW, Ekekezie II, Rezaiekhaligh M, Mabry
SM, et al. Responses of pulmonary platelet-derived growth factor peptides
and receptors to hyperoxia and nitric oxide in piglet lungs. Pediatr Res. (2005)
57:523–9. doi: 10.1203/01.PDR.0000155762.91748.8D
34. Ovchinnikov DA. Macrophages in the embryo and beyond: much more
than just giant phagocytes. Genesis. (2008) 46:447–62. doi: 10.1002/dvg.
20417
35. Fantin A, Vieira JM, Gestri G, Denti L, Schwarz Q, Prykhozhij S, et al. Tissue
macrophages act as cellular chaperones for vascular anastomosis downstream
of VEGF-mediated endothelial tip cell induction. Blood. (2010) 116:829–40.
doi: 10.1182/blood-2009-12-257832
36. Rae F, Woods K, Sasmono T, Campanale N, Taylor D, Ovchinnikov DA, et al.
Characterisation and trophic functions of murine embryonic macrophages
based upon the use of a Csf1r-EGFP transgene reporter. Dev Biol. (2007)
308:232–46. doi: 10.1016/j.ydbio.2007.05.027
37. Eldredge LC, Treuting PM, Manicone AM, Ziegler SF, Parks WC,
McGuire JK. CD11b(+) mononuclear cells mitigate hyperoxia-induced lung
injury in neonatal mice. Am J Respir Cell Mol Biol. (2016) 54:273–83.
doi: 10.1165/rcmb.2014-0395OC
38. Al-Rubaie A, Wise AF, Sozo F, De Matteo R, Samuel CS, Harding R, et al.
The therapeutic effect of mesenchymal stem cells on pulmonary myeloid cells
following neonatal hyperoxic lung injury in mice. Respir Res. (2018) 19:114.
doi: 10.1186/s12931-018-0816-x
39. Syed MA, Bhandari V. Hyperoxia exacerbates postnatal inflammation-
induced lung injury in neonatal BRP-39 null mutant mice promoting
the M1 macrophage phenotype. Mediators Inflamm. (2013) 2013:457189.
doi: 10.1155/2013/457189
40. Nagato AC, Bezerra FS, Talvani A, Aarestrup BJ, Aarestrup FM. Hyperoxia
promotes polarization of the immune response in ovalbumin-induced airway
inflammation, leading to a TH17 cell phenotype. Immun Inflamm Dis. (2015)
3:321–37. doi: 10.1002/iid3.71
Conflict of Interest Statement: The authors declare that the research was
conducted in the absence of any commercial or financial relationships that could
be construed as a potential conflict of interest.
Copyright © 2019 Chen, Hwang and Chou. This is an open-access article distributed
under the terms of the Creative Commons Attribution License (CC BY). The use,
distribution or reproduction in other forums is permitted, provided the original
author(s) and the copyright owner(s) are credited and that the original publication
in this journal is cited, in accordance with accepted academic practice. No use,
distribution or reproduction is permitted which does not comply with these terms.
Frontiers in Immunology | www.frontiersin.org 10 April 2019 | Volume 10 | Article 681
... Term-born rat models are appropriate for studying the effects of hyperoxia on preterm infants with respiratory distress because rats are born at the saccular stage, which is approximately equivalent to a human gestational age of 30 weeks [1]. The prolonged exposure of neonatal rats to hyperoxia results in a decrease in alveolarization and vascularization similar to human bronchopulmonary dysplasia (BPD) [2,3]. The pathogenesis of BPD is multifactorial, and oxygen toxicity plays a crucial role in the process of lung injury leading to BPD [4,5]. ...
... In this study, we determined the levels of VEGF, PDGF-A, and PDGF-B expression and elucidated the mechanisms that mediate the hUC-MSCs' effects because their mRNA and protein expression decreased in the lungs of newborn animals exposed to 14 days of hyperoxia [3,24,25]. VEGF is a potent endothelial cell mitogen that regulates angiogenesis and alveolar development [26]. PDGF is crucial to the alveolarization of normally developing lungs [27]. ...
... Treatment with surfactant and hUC-MSCs augmented the hyperoxia-induced decrease in the VEGF, PDGF-A, and PDGF-B protein expression levels. These results suggest that treatment with hUC-MSCs enhanced vascular Prolonged exposure to hyperoxia increases cytokine and induces inflammation in neonatal rat lungs [3]. Oxygen supplementation in preterm infants with respiratory distress syndrome increases oxidative stress and cytokines and the cytokine levels were increased in the tracheal aspirate of newborns with BPD [28]. ...
Article
Full-text available
Background Surfactant therapy is a standard of care for preterm infants with respiratory distress and reduces the incidence of death and bronchopulmonary dysplasia in these patients. Our previous study found that mesenchymal stem cells (MSCs) attenuated hyperoxia-induced lung injury and the combination therapy of surfactant and human umbilical cord-derived MSCs (hUC-MSCs) did not have additive effects on hyperoxia-induced lung injury in neonatal rats. The aim is to evaluate the effects of 2 consecutive days of intratracheal administration of surfactant and hUC-MSCs on hyperoxia-induced lung injury. Methods Neonatal Sprague Dawley rats were reared in either room air (RA) or hyperoxia (85% O 2 ) from postnatal days 1 to 14. On postnatal day 4, the rats received intratracheal injections of either 20 μL of normal saline (NS) or 20 μL of surfactant. On postnatal day 5, the rats reared in RA received intratracheal NS, and the rats reared in O 2 received intratracheal NS or hUC-MSCs (3 × 10 ⁴ or 3 × 10 ⁵ cells). Six study groups were examined: RA + NS + NS, RA + surfactant + NS, O 2 + NS + NS, O 2 + surfactant + NS, O 2 + surfactant + hUC-MSCs (3 × 10 ⁴ cells), and O 2 + surfactant + hUC-MSCs (3 × 10 ⁵ cells). The lungs were excised for histological, western blot, and cytokine analyses. Results The rats reared in hyperoxia and treated with NS yielded significantly higher mean linear intercepts (MLIs) and interleukin (IL)-1β and IL-6 levels and significantly lower vascular endothelial growth factors (VEGFs), platelet-derived growth factor protein expression, and vascular density than did those reared in RA and treated with NS or surfactant. The lowered MLIs and cytokines and the increased VEGF expression and vascular density indicated that the surfactant and surfactant + hUC-MSCs (3 × 10 ⁴ cells) treatment attenuated hyperoxia-induced lung injury. The surfactant + hUC-MSCs (3 × 10 ⁵ cells) group exhibited a significantly lower MLI and significantly higher VEGF expression and vascular density than the surfactant + hUC-MSCs (3 × 10 ⁴ cells) group did. Conclusions Consecutive daily administration of intratracheal surfactant and hUC-MSCs can be an effective regimen for treating hyperoxia-induced lung injury in neonates.
... We revealed that maternal immunization with Tn vaccine increased neonatal rat serum levels of anti-Tn antibody; reduced mean linear intercept (MLI), lung cytokine stress, and oxidative stress; and increased vascular density and growth factor expressions to normoxic levels in hyperoxia-injured neonatal rats (Chen et al., 2019). These findings suggest that anti-Tn antibody has therapeutic effects on rats with hyperoxia-induced lung injury. ...
... In our previous study, we found that maternal immunization with Tn vaccine increased maternal and neonatal serum anti-Tn antibody titers and attenuated hyperoxia-induced lung injury in newborn rats (Chen et al., 2019). However, immunizing mothers to treat premature birth-related lung diseases is impractical because prematurity is clinically unpredictable and unavoidable. ...
... In this study, we evaluated the protective effects of anti-Tn monoclonal antibody on alveolar and vascular development in newborn mice exposed to hyperoxia. We demonstrated that hyperoxia increased oxidative stress and inflammation and led to impaired alveolarization and angiogenesis in neonatal rat lungs (Chen et al., 2019). Consequently, we chose a comparable model and demonstrated hyperoxia affected mice angiogenesis in a similar way. ...
Article
Full-text available
Maternal immunization with Tn vaccine increases serum anti-Tn antibody titers and attenuates hyperoxia-induced lung injury in neonatal rats. This study determined whether anti-Tn monoclonal antibody can protect against hyperoxia-induced lung injury in neonatal mice. Newborn BALB/c mice were exposed to room air (RA) or normobaric hyperoxia (85% O2) for 1 week, creating four study groups as follows: RA + phosphate-buffered saline (PBS), RA + anti-Tn monoclonal antibody, O2 + PBS, and O2 + anti-Tn monoclonal antibody. The anti-Tn monoclonal antibody at 25 μg/g body weight in 50 μl PBS was intraperitoneally injected on postnatal days 2, 4, and 6. Hyperoxia reduced body weight and survival rate, increased mean linear intercept (MLI) and lung tumor necrosis factor-α, and decreased vascular endothelial growth factor (VEGF) expression and vascular density on postnatal day 7. Anti-Tn monoclonal antibody increased neonatal serum anti-Tn antibody titers, reduced MLI and cytokine, and increased VEGF expression and vascular density to normoxic levels. The attenuation of lung injury was accompanied by a reduction in lung oxidative stress and nuclear factor-κB activity. Anti-Tn monoclonal antibody improves alveolarization and angiogenesis in hyperoxia-injured newborn mice lungs through the suppression of oxidative stress and inflammation.
... 7,8 Previous studies have found that inflammatory cytokines can promote the glycan epitope (e.g., sialyl-Lewis[x] antigen) by regulating specific glycosyltransferases. 9,10 Chiang et al. 11 developed an anti-Tn vaccine by using the linear array epitope technology, which induces anti-Tn antibodies with high specificity and affinity in mice. Since in the previous study, we showed that maternal Tn immunization increases maternal and neonatal serum antibody titers and attenuates hyperoxia-induced lung injury in newborn rats by suppressing oxidative stress and inflammation, 12 these observations prompted us to examine the effects of Tn immunization on neonatal hyperoxia-induced kidney injury. ...
... Tn vaccine preparation Tn vaccine preparation was described in detail in our previous study. 12 Briefly, Tn was conjugated to mFc(Cys42)Histag2 or GST (Cys6)Histag2 at a glycotope-carrier protein with a weight ratio of 5:1. After 48 h, the conjugate was refolded in phosphate-buffered saline (PBS) with 0.2 mM tris (2-carboxyethyl) phosphine (TCEP). ...
... The kidney tissues used for these experiments were obtained from a previous study, which was designed to assess lung injury. 12 Histological examination The kidney was placed in 4% paraformaldehyde, washed in PBS, and serially dehydrated in increasing ethanol concentrations before embedding in paraffin. Tissue sections of 7 µm were stained with hematoxylin and eosin (H&E) and Masson trichrome, examined under light microscopy, and assessed for kidney morphology and fibrosis. ...
Article
Neonatal hyperoxia increases oxidative stress and adversely disturbs glomerular and tubular maturity. Maternal Tn immunization induces anti-Tn antibody titer and attenuates hyperoxia-induced lung injury in neonatal rats. We intraperitoneally immunized female Sprague–Dawley rats (6 weeks old) with Tn immunogen (50 μg/dose) or carrier protein five times at biweekly intervals on 8, 6, 4, 2, and 0 weeks before the delivery day. The pups were reared for 2 weeks in either room air (RA) or in 85% oxygen-enriched atmosphere (O2), thus generating four study groups, namely carrier protein + RA, Tn vaccine + RA, carrier protein + O2, and Tn vaccine + O2. On postnatal day 14, the kidneys were harvested for the oxidative stress marker 8-hydroxy-2’-deoxyguanosine (8-OHdG), nuclear factor-κB (NF-κB), and collagen expression and histological analyses. Hyperoxia reduced body weight, induced tubular and glomerular injuries, and increased 8-OHdG and NF-κB expression and collagen deposition in the kidneys. By contrast, maternal Tn immunization reduced kidney injury and collagen deposition in neonatal rats. Furthermore, kidney injury attenuation was accompanied by a reduction in 8-OHdG and NF-κB expression. Maternal Tn immunization protects against hyperoxia-induced kidney injury in neonatal rats by attenuating oxidative stress and NF-κB activity. Hyperoxia increased nuclear factor-κB (NF-κB) activity and collagen deposition in neonatal rat kidney. Maternal Tn immunization reduced kidney injury as well as collagen deposition in neonatal rats. Maternal Tn immunization reduced kidney injury and was associated with a reduction in 8-hydroxy-2′-deoxyguanosine and NF-κB activity. Tn vaccine can be a promising treatment modality against hyperoxia-induced kidney injury in neonates.
... impaired lung angiogenesis and development. 9,24,34 The hyperoxia-induced decrease in the expressions of HIF-1a, pmTOR, and eNOs indicated the inhibition of lung angiogenesis and the related signaling pathway. By contrast, enhanced HIF-1a expression achieved through the intraperitoneal administration of roxadustat stimulated lung angiogenesis and reversed hyperoxia-induced arrested alveolar growth (Fig. 6). ...
... The vWF expressions in IHC or immunofluorescence were used to determine the pulmonary angiogenesis of neonatal hyperoxia-induced lung injury in previous research. 24,35 The attenuation of pmTOR and subsequent suppression of HIF-1a after oxygen exposure had been presented in a previous BPD rat model. 36 In the present study, we found that HIF-1a Multiple studies have reported the pharmacological stabilization of HIF, demonstrating its therapeutic benefits in experimental BPD. ...
Article
Full-text available
Background Premature infants who require oxygen therapy for respiratory distress syndrome often develop bronchopulmonary dysplasia, a chronic lung disease characterized by interrupted alveologenesis. Disrupted angiogenesis inhibits alveologenesis; however, the mechanisms through which disrupted angiogenesis affects lung development are poorly understood. Hypoxia-inducible factors (HIFs) are transcription factors that activate multiple oxygen-sensitive genes, including those encoding for vascular endothelial growth factor (VEGF). However, the HIF modulation of angiogenesis in hyperoxia-induced lung injury is not fully understood. Therefore, we explored the effects of roxadustat, an HIF stabilizer that has been shown to promote angiogenesis, in regulating pulmonary angiogenesis upon hyperoxia exposure. Methods C57BL6 mice pups reared in room air and 85% O2 were injected with phosphate-buffered saline or 5 mg/kg or 10 mg/kg roxadustat. Their daily body weight and survival rate were recorded. Their lungs were excised for histology and angiogenic factor expression analyses on postnatal Day 7. Results Exposure to neonatal hyperoxia reduced body weight; survival rate; and expressions of von Willebrand factor, HIF-1α, phosphor mammalian target of rapamycin, VEGF, and endothelial nitric oxide synthase and increased the mean linear intercept values in the pups. Roxadustat administration reversed these effects. Conclusion Hyperoxia suppressed pulmonary vascular development and the expression of proangiogenic factors. Roxadustat promoted pulmonary angiogenesis upon hyperoxia exposure by stabilizing HIF-1α and upregulating the expression of proangiogenic factors, indicating its potential in clinical and therapeutic applications.
... 1 Prolonged exposure of neonatal rats to hyperoxia was reported to result in decreased alveolarization and vascularization similar to those in human bronchopulmonary dysplasia. 2,3 The pathogenesis of BPD is multifactorial, and oxygen toxicity is considered to play a crucial role in the lung injury process that leads to BPD development. 4,5 Currently, no effective therapy is clinically available for treating established lung injury induced by hyperoxia. ...
... The nursing mothers were rotated between the 85% O 2 and RA groups every 24 h to prevent oxygen toxicity in the mothers and eliminate differing maternal effects between groups. Oxygen exposure was performed in a transparent 60 × 50 × 40-cm 3 Plexiglas chamber into which oxygen was continuously delivered at a rate of 4 L/min. Oxygen levels were monitored using a Pro:ox Model 110 monitor (NexBiOxy, Hsinchu, 5 Taiwan). ...
Article
Full-text available
Background Hyperoxia increases Sonic hedgehog (Shh) expression in neonatal rat lungs. The effect of mesenchymal stem cells (MSCs) on the hedgehog signaling pathway in hyperoxia-induced lung injury is unknown. This study evaluated whether MSCs could inhibit hedgehog signaling and improve established hyperoxia-induced lung injury in newborn rats. Methods: Newborn rats were assigned to room air (RA) or hyperoxia (85% O2) groups from postnatal day 4 to 15, and some received intravenous injection of human MSCs (9 × 10⁵ cells) in 90 μL of normal saline (NS) through the tail vein on postnatal day 15. We obtained four study groups as follows: RA + NS, RA + MSCs, O2 + NS, and O2 + MSCs. Pups from each group were sacrificed on postnatal days 15 and 29, and lungs were removed for histological and Western blot analyses. Results Neonatal hyperoxia on postnatal days 4–15 reduced the body weight, increased the mean linear intercept, and decreased the vascular density of the rats, and these effects were associated with increased Shh and Smoothened (Smo) expression and decreased Patched expression. By contrast, the MSC-treated hyperoxic pups exhibited improved alveolarization, increased vascularization, and decreased Shh and Smo expression on postnatal day 29. Conclusion Human MSC treatment reversed established hyperoxia-induced lung injury in newborn rats, probably through suppression of the hedgehog pathway.
... Hyperoxia increased lung oxidative stress in neonatal rats [7]. Oxidative stress is associated with complications in preterm infants, including preterm retinopathy, bronchopulmonary dysplasia, and necrotizing enterocolitis [8,9]. ...
Article
Full-text available
High oxygen concentrations are often required to treat newborn infants with respiratory distress but have adverse effects, such as increased oxidative stress and ferroptosis and impaired alveolarization. Cathelicidins are a family of antimicrobial peptides that exhibit antioxidant activity, and they can reduce hyperoxia-induced oxidative stress. This study evaluated the effects of cathelicidin treatment on lung ferroptosis and alveolarization in hyperoxia-exposed newborn rats. Sprague Dawley rat pups were either reared in room air (RA) or hyperoxia (85% O2) and then randomly given cathelicidin (8 mg/kg) in 0.05 mL of normal saline (NS), or NS was administered intraperitoneally on postnatal days from 1–6. The four groups obtained were as follows: RA + NS, RA + cathelicidin, O2 + NS, and O2 + cathelicidin. On postnatal day 7, lungs were harvested for histological, biochemical, and Western blot analyses. The rats nurtured in hyperoxia and treated with NS exhibited significantly lower body weight and cathelicidin expression, higher Fe2+, malondialdehyde, iron deposition, mitochondrial damage (TOMM20), and interleukin-1β (IL-1β), and significantly lower glutathione, glutathione peroxidase 4, and radial alveolar count (RAC) compared to the rats kept in RA and treated with NS or cathelicidin. Cathelicidin treatment mitigated hyperoxia-induced lung injury, as demonstrated by higher RAC and lower TOMM20 and IL-1β levels. The attenuation of lung injury was accompanied by decreased ferroptosis. These findings indicated that cathelicidin mitigated hyperoxia-induced lung injury in the rats, most likely by inhibiting ferroptosis.
... In this study, we found that rat pups exposed to hyperoxic conditions showed decreased body weight and lung weight 19 TRIM72 expression in lung tissues under hyperoxic conditions was higher than that under normoxia on postnatal days 7 and 14. Furthermore, the attenuation or induction of TRIM72 expression in response to hyperoxia seemed to enhance or suppress the viability of cultured lung cells. ...
Article
Full-text available
Background: Premature infants often require oxygen (O2) therapy for respiratory distress syndrome; however, excessive use of O2 can cause clinical conditions such as bronchopulmonary dysplasia (BPD). Although many treatment methods are currently available, they are not effective in preventing BPD. Herein, we explored the role of tripartite motif protein 72 (TRIM72), a factor involved in repairing alveolar epithelial wounds, in regulating alveolar cells upon hyperoxia exposure. Methods: In this in vivo study, we used Sprague-Dawley rat pups that were reared in room air or 85% O2 for 2 weeks after birth. The lungs were excised for histological analyses, and TRIM72 expression was assessed on postnatal days 7 and 14. For in vitro experiments, RLE-6TN cells (i.e., rat alveolar type II epithelial cells) and A549 cells (i.e., human lung carcinoma epithelial cells) were exposed to 85% O2 for 5 days. The cells were then analyzed for cell viability, and TRIM72 expression was determined. Results: Exposure to hyperoxia reduced body and lung weight, increased mean linear intercept values, and upregulated TRIM72 expression. In vitro study results revealed increased or decreased lung cell viability upon hyperoxia exposure depending on the suppression or overexpression of TRIM72, respectively. Conclusion: Hyperoxia upregulates TRIM72 expression in neonatal rat lung tissue; moreover, it initiates TRIM72-dependent alveolar epithelial cell death, leading to hyperoxia-induced lung injury.
... We evaluated the therapeutic effects of cathelicidin on lung and vascular development in hyperoxia-exposed newborn rats. We previously observed that hyperoxia exposure causes increased lung oxidative stress and inflammation and leads to alveolar and pulmonary vascular simplification in rats [30]. Thus, we used a similar model for this study and found that hyperoxia exposure affects rat lung vascularization in an identical pattern. ...
Article
Purpose: High concentrations of oxygen administered to newborn infants with respiratory failure increases oxidant stress and leads to lung injury, characterized by decreased alveolar and capillary development. Cathelicidin belongs to an important group of human antimicrobial peptides that exhibit antioxidant activity; its overexpression reduces hyperoxia-induced oxidative stress. This study evaluated the therapeutic effects of cathelicidin in hyperoxia-induced lung injury in newborn rats. Methods and materials: Sprague Dawley rat pups were reared in either room air (RA) or hyperoxia (85% O2) and then randomly treated with low-dose (4 mg/kg) and high-dose (8 mg/kg) cathelicidin in 0.05 mL of normal saline (NS) administered intraperitoneally on postnatal days 1-6. The following six groups were obtained: RA + NS, RA + low-dose cathelicidin, RA + high-dose cathelicidin, O2 + NS, O2 + low-dose cathelicidin, and O2 + high-dose cathelicidin. Lungs were harvested for Western blot and histological analyses on postnatal day 7. Results: Compared with the RA-reared rats, the hyperoxia-reared rats exhibited significantly lower body weights, higher mean linear intercept (MLI), lung injury score, interleukin-6, and oxidative stress marker 8-hydroxy-2'-deoxyguanosine (8-OHdG) expression but lower superoxide dismutase 1 (SOD1) and vascular endothelial growth factor (VEGF) protein expression and vascular density. Cathelicidin treatment attenuated hyperoxia-induced lung injury as demonstrated by lower MLI and injury score and higher VEGF expression and vascular density. Conclusions: Cathelicidin attenuated hyperoxia-induced lung injury and caused a decrease in 8-OHdG and SOD1 protein expression, most likely by inhibiting oxidative stress in the lung.
Article
Although oxygen supplementation is beneficial to support life in the clinic, excessive oxygen therapy also has been linked to damage to organs such as the lung or the eye. However, there is a lack of understanding of whether high oxygen therapy directly affects the kidney, leading to acute kidney injury, and what molecular mechanisms may be involved in this process. In this review, we revise our current understanding of the mechanisms by which hyperoxia leads to organ damage and highlight possible areas of investigation for the scientific community interested in novel mechanisms of kidney disease. Overall, we found a significant need for both animal and clinical studies evaluating the role of hyperoxia in inducing kidney damage. Thus, we urge the research community to further investigate oxygen therapy and its impact on kidney health with the goal of optimizing oxygen therapy guidelines and improving patient care.
Article
It has been reported that Dendrobium officinale polysaccharides (DOPS) could alleviate colitis in animal model and suppress the activation of NLRP3 inflammasome and β-arrestin1 in vitro. However, it remains unclear whether DOPS has effect on protecting against colitis-induced pulmonary injury. The purpose of this study was to explore the protective effect and mechanism of DOPS on colitis-induced lung injury. A dextran sodium sulfate (DSS)-induced mice colitis model and lipopolysaccharide (LPS)-stimulated BEAS-2B cells model were applied in this study. The results showed that DOPS treatment restored histopathological changes, reduced inflammatory cells infiltration, pro-inflammatory cytokines levels, reactive oxygen species (ROS) formation and MDA generation, and increased anti-oxidative enzymes activities including SOD and GSH-Px in colitis mice. Further investigation showed that DOPS significantly inhibited the protein expression of TLR4, and apparently up-regulated proteins expressions of nuclear-Nrf2, HO-1 and NQO-1 in lung tissues of colitis mice and in BEAS-2B cells. These results indicated that DOPS significantly inhibited inflammation and oxidative stress to alleviate colitis-induced secondary lung injury, and its mechanisms are closely related to the inhibition of TLR4 signaling pathway and the activation of Nrf2 signaling pathway. DOPS may be a promising drug for alleviating colitis-induced lung injury.
Article
Full-text available
The lungs of extremely low gestational age neonates (ELGANs) are deficient in pulmonary surfactant and are incapable of efficient gas exchange necessary for successful transition from a hypoxic intrauterine environment to ambient air. To improve gas exchange and survival, ELGANs often receive supplemental oxygen with mechanical ventilation which disrupts normal lung developmental processes, including microvascular maturation and alveolarization. Factors that regulate these developmental processes include vascular endothelial growth factor and matrix metalloproteinases, both of which are influenced by generation of oxygen byproducts, or reactive oxygen species (ROS). ELGANs are also deficient in antioxidants necessary to scavenge excessive ROS. Thus, the accumulation of ROS in the preterm lungs exposed to prolonged hyperoxia, results in inflammation and development of bronchopulmonary dysplasia (BPD), a form of chronic lung disease (CLD). Despite advances in neonatal care, BPD/CLD remains a major cause of neonatal morbidity and mortality. The underlying mechanisms are not completely understood, and the benefits of current therapeutic interventions are limited. The association between ROS and biomarkers of microvascular maturation and alveolarization, as well as antioxidant therapies in the setting of hyperoxia-induced neonatal lung injury are reviewed in this article.
Article
Full-text available
Background: Exposure to high levels of oxygen (hyperoxia) after birth leads to lung injury. Our aims were to investigate the modulation of myeloid cell sub-populations and the reduction of fibrosis in the lungs following administration of human mesenchymal stem cells (hMSC) to neonatal mice exposed to hyperoxia. Method: Newborn mice were exposed to 90% O2 (hyperoxia) or 21% O2 (normoxia) from postnatal days 0-4. A sub-group of hyperoxia mice were injected intratracheally with 2.5X105 hMSCs. Using flow cytometry we assessed pulmonary immune cells at postnatal days 0, 4, 7 and 14. The following markers were chosen to identify these cells: CD45+ (leukocytes), Ly6C+Ly6G+ (granulocytes), CD11b+CD11c+ (macrophages); macrophage polarisation was assessed by F4/80 and CD206 expression. hMSCs expressing enhanced green fluorescent protein (eGFP) and firefly luciferase (fluc) were administered via the trachea at day 4. Lung macrophages in all groups were profiled using next generation sequencing (NGS) to assess alterations in macrophage phenotype. Pulmonary collagen deposition and morphometry were assessed at days 14 and 56 respectively. Results: At day 4, hyperoxia increased the number of pulmonary Ly6C+Ly6G+ granulocytes and F4/80lowCD206low macrophages but decreased F4/80highCD206high macrophages. At days 7 and 14, hyperoxia increased numbers of CD45+ leukocytes, CD11b+CD11c+ alveolar macrophages and F4/80lowCD206low macrophages but decreased F4/80highCD206high macrophages. hMSCs administration ameliorated these effects of hyperoxia, notably reducing numbers of CD11b+CD11c+ and F4/80lowCD206low macrophages; in contrast, F4/80highCD206high macrophages were increased. Genes characteristic of anti-inflammatory 'M2' macrophages (Arg1, Stat6, Retnla, Mrc1, Il27ra, Chil3, and Il12b) were up-regulated, and pro-inflammatory 'M1' macrophages (Cd86, Stat1, Socs3, Slamf1, Tnf, Fcgr1, Il12b, Il6, Il1b, and Il27ra) were downregulated in isolated lung macrophages from hyperoxia-exposed mice administered hMSCs, compared to mice without hMSCs. Hydroxyproline assay at day 14 showed that the 2-fold increase in lung collagen following hyperoxia was reduced to control levels in mice administered hMSCs. By day 56 (early adulthood), hMSC administration had attenuated structural changes in hyperoxia-exposed lungs. Conclusions: Our findings suggest that hMSCs reduce neonatal lung injury caused by hyperoxia by modulation of macrophage phenotype. Not only did our cell-based therapy using hMSC induce structural repair, it limited the progression of pulmonary fibrosis.
Article
Full-text available
Background: Systemic maternal inflammation and neonatal hyperoxia arrest alveolarization in neonates. The aims were to test whether human mesenchymal stem cells (MSCs) reduce lung inflammation and improve lung development in perinatal inflammation- and hyperoxia-induced experimental bronchopulmonary dysplasia. Methods: Pregnant Sprague-Dawley rats were intraperitoneally injected with lipopolysaccharide (LPS, 0.5 mg/kg/day) on Gestational Days 20 and 21. Human MSCs (3×10(5) and 1×10(6) cells) in 0.03 ml normal saline (NS) were administered intratracheally on Postnatal Day 5. Pups were reared in room air (RA) or an oxygen-enriched atmosphere (O2) from Postnatal Days 1 to 14, and six study groups were obtained: LPS+RA+NS, LPS+RA+MSC (3×10(5) cells), LPS+RA+MSC (1×10(6) cells), LPS+O2+NS, LPS+O2+MSC (3×10(5) cells), and LPS+O2+MSC (1×10(6) cells). The lungs were excised for cytokine, vascular endothelial growth factor (VEGF) and connective tissue growth factor (CTGF) expression, and histological analyses on Postnatal Day 14. Results: Body weight was significantly lower in rats reared in hyperoxia than in those reared in RA. The LPS+O2+NS group exhibited a significantly higher mean linear intercept (MLI) and collagen density and a significantly lower vascular density than the LPS+RA+NS group did. Administering MSC to hyperoxia-exposed rats improved MLI and vascular density and reduced tumor necrosis factor-α and interleukin-6 levels and collagen density to normoxic levels. This improvement in lung development and fibrosis was accompanied by an increase and decrease in lung VEGF and CTGF expression, respectively. Conclusion: Human MSCs attenuated perinatal inflammation- and hyperoxia-induced defective alveolarization and angiogenesis and reduced lung fibrosis, likely through increased VEGF and decreased CTGF expression.
Article
Full-text available
Previous studies have demonstrated that hyperoxia-induced stress and oxidative damage to the lungs of mice lead to an increase in IL-6, TNF-α, and TGF-β expression. Together, IL-6 and TGF-β have been known to direct T cell differentiation toward the TH17 phenotype. In the current study, we tested the hypothesis that hyperoxia promotes the polarization of T cells to the TH17 cell phenotype in response to ovalbumin-induced acute airway inflammation. Airway inflammation was induced in female BALB/c mice by intraperitoneal sensitization and intranasal introduction of ovalbumin, followed by challenge methacholine. After the methacholine challenge, animals were exposed to hyperoxic conditions in an inhalation chamber for 24 h. The controls were subjected to normoxia or aluminum hydroxide dissolved in phosphate buffered saline. After 24 h of hyperoxia, the number of macrophages and lymphocytes decreased in animals with ovalbumin-induced airway inflammation, whereas the number of neutrophils increased after ovalbumin-induced airway inflammation. The results showed that expression of Nrf2, iNOS, T-bet and IL-17 increased after 24 of hyperoxia in both alveolar macrophages and in lung epithelial cells, compared with both animals that remained in room air, and animals with ovalbumin-induced airway inflammation. Hyperoxia alone without the induction of airway inflammation lead to increased levels of TNF-α and CCL5, whereas hyperoxia after inflammation lead to decreased CCL2 levels. Histological evidence of extravasation of inflammatory cells into the perivascular and peribronchial regions of the lungs was observed after pulmonary inflammation and hyperoxia. Hyperoxia promotes polarization of the immune response toward the TH17 phenotype, resulting in tissue damage associated with oxidative stress, and the migration of neutrophils to the lung and airways. Elucidating the effect of hyperoxia on ovalbumin-induced acute airway inflammation is relevant to preventing or treating asthmatic patients that require oxygen supplementation to reverse the hypoxemia.
Article
Full-text available
Bronchopulmonary dysplasia (BPD) is a common consequence of life-saving interventions for infants born with immature lungs. Resident tissue myeloid cells regulate lung pathology, but their role in BPD is poorly understood. To determine the role of lung interstitial myeloid cells in neonatal responses to lung injury, we exposed newborn mice to hyperoxia, a neonatal mouse lung injury model with features of human BPD. In newborn mice raised in normoxia, we identified a CD45+F4/80+CD11b+CD71+ population of cells in lungs of neonatal mice present in significantly greater percentages than in adult mice. In response to hyperoxia, surface marker and gene expression in whole lung macrophages/monocytes was biased to an alternatively activated phenotype. Partial depletion of these CD11b+ mononuclear cells using CD11b-diptheria toxin receptor (DTR) transgenic mice resulted in 60% mortality by 40 h of hyperoxia exposure with more severe lung injury, perivascular edema, alveolar hemorrhage, compared to DT-treated CD11b-DTR negative controls, which displayed no mortality. These results identify and anti-inflammatory population of CD11b+ mononuclear cells that are protective in hyperoxia-induced neonatal lung injury in mice and suggest that enhancing their beneficial functions may be a treatment strategy in infants at risk for BPD.
Article
Full-text available
Preterm infants often require supplemental oxygen due to lung immaturity, but hyperoxia can contribute to an increased risk of respiratory illness later in life. Our aim was to compare the effects of mild and moderate levels of neonatal hyperoxia on markers of pulmonary oxidative stress and inflammation, and on lung architecture; both immediate and persistent effects were assessed. Neonatal mice (C57BL6/J) were raised in either room air (21% O2), mild (40% O2), or moderate (65% O2) hyperoxia from birth until postnatal day 7 (P7d). The mice were killed at either P7d (immediate effects), or lived in air until adulthood (P56d, persistent effects). We enumerated macrophages in lung tissue at P7d and immune cells in bronchoalveolar lavage fluid (BALF) at P56d. At P7d and P56d, we assessed pulmonary oxidative stress (heme oxygenase-1 (HO-1) and nitrotyrosine staining) and lung architecture. The data were interrogated for sex differences. At P7d, HO-1 gene expression was greater in the 65% O2 group than in the 21% O2 group. At P56d, the area of nitrotyrosine staining and number of immune cells were greater in the 40% O2 and 65% O2 groups relative to the 21% O2 group. Exposure to 65% O2, but not 40% O2, led to larger alveoli and lower tissue fraction in the short-term and to persistently fewer bronchiolar-alveolar attachments. Exposure to 40% O2 or 65% O2 causes persistent increases in pulmonary oxidative stress and immune cells, suggesting chronic inflammation within the adult lung. Unlike 65% O2, 40% O2 does not affect lung architecture. Copyright © 2014, American Journal of Physiology - Lung Cellular and Molecular Physiology.
Article
Infants born very prematurely (<28 wk gestation) have immature lungs and often require supplemental oxygen. However, long-term hyperoxia exposure can arrest lung development, leading to bronchopulmonary dysplasia (BPD), which increases acute and long-term respiratory morbidity and mortality. The neural mechanisms controlling breathing are highly plastic during development. Whether the ventilatory control system adapts to pulmonary disease associated with hyperoxia exposure in infancy remains unclear. Here, we assessed potential age-dependent adaptations in the control of breathing in an established rat model of BPD associated with hyperoxia. Hyperoxia exposure ( FI O 2 ; 0.9 from 0 to 10 days of life) led to a BPD-like lung phenotype, including sustained reductions in alveolar surface area and counts, and modest increases in airway resistance. Hyperoxia exposure also led to chronic increases in room air and acute hypoxic minute ventilation (V̇e) and age-dependent changes in breath-to-breath variability. Hyperoxia-exposed rats had normal oxygen saturation ( S p O 2 ) in room air but greater reductions in S p O 2 during acute hypoxia (12% O2) that were likely due to lung injury. Moreover, acute ventilatory sensitivity was reduced at P12 to P14. Perinatal hyperoxia led to greater glial fibrillary acidic protein expression and an increase in neuron counts within six of eight or one of eight key brainstem regions, respectively, controlling breathing, suggesting astrocytic expansion. In conclusion, perinatal hyperoxia in rats induced a BPD-like phenotype and age-dependent adaptations in V̇e that may be mediated through changes to the neural architecture of the ventilatory control system. Our results suggest chronically altered ventilatory control in BPD.
Article
Prolonged hyperoxia exposure leads to inflammation and acute lung injury. Since hyperoxia activates nuclear factor kappa B (NF-κB) and proinflammatory mediators in lung fibroblasts and murine lungs, and proinflammatory cytokines upregulate Tn (N-acetyl-d-galactosamine-O-serine/threonine) expression in human gingival fibroblasts. We hypothesized connections exist between Tn expression and inflammation regulation. Thus, we immunized adult mice with Tn antigen to examine whether Tn vaccine can protect against hyperoxia-induced lung injury by inhibiting NF-κB activity and cytokine expression through the action of anti-Tn antibodies. Five-week-old female C57BL/6NCrlBltw mice were subcutaneously immunized with Tn antigen four times at biweekly intervals, and one additional immunization was performed at 1 week after the fourth immunization. Four days after the last immunization, mice were exposed to room air (RA) or hyperoxia (100% O2) for up to 96 h. Four study groups were examined: carrier protein + RA (n = 6), Tn vaccine + RA (n = 6), carrier protein + O2 (n = 6), and Tn vaccine + O2 (n = 5). We observed that hyperoxia exposure reduced body weight, increased alveolar protein and cytokine (interleukin-6 and tumor necrosis factor-α) levels, increased mean linear intercept (MLI) values and lung injury scores, and increased lung NF-κB activity. By contrast, Tn immunization increased serum anti-Tn antibody titers and reduced the cytokine levels, MLI values, and lung injury scores. Furthermore, the alleviation of lung injury was accompanied by a reduction in NF-κB activity. Therefore, we proposed that Tn immunization attenuates hyperoxia-induced lung injury in adult mice by inhibiting the NF-κB activity.
Article
Background: Premature infants often require oxygen supplementation and, therefore, are exposed to oxidative stress. Following oxygen exposure, preterm infants frequently develop chronic lung disease and have a significantly increased risk of asthma. Objective: We sought to identify the underlying mechanisms by which neonatal hyperoxia promotes asthma development. Methods: Mice were exposed to neonatal hyperoxia followed by a period room air recovery. A group of mice were also intranasally exposed to house dust mite antigen (HDM). Assessments were performed at various time points for evaluation of airway hyperresponsiveness (AHR), eosinophilia, mucus production, inflammatory gene expression, T helper (Th) and group 2 innate lymphoid cell (ILC2) responses. Sera from term- and preterm-born infants were also collected and the levels of IL-33 and type 2 cytokines were measured. Results: Neonatal hyperoxia induced asthma-like features including AHR, mucus hyperplasia, airway eosinophilia, and type 2 pulmonary inflammation. In addition, neonatal hyperoxia promoted allergic Th responses to HDM exposure. Elevated IL-33 levels and ILC2 responses were observed in the lungs most likely due to oxidative stress caused by neonatal hyperoxia. IL-33 receptor signaling and ILC2s were vital for the induction of asthma-like features following neonatal hyperoxia. Serum IL-33 levels correlated significantly with serum levels of IL-5 and IL-13, but not IL-4 in preterm infants. Conclusion: These data demonstrate that an axis involving IL-33 and ILC2s is important for the development of asthma-like features following neonatal hyperoxia and suggest therapeutic potential for targeting IL-33, ILC2s, and oxidative stress to prevent and/or treat asthma development related to prematurity.
Article
Prevention or treatment of lung diseases caused by the failure to form, or destruction of, existing alveoli, as observed in infants with bronchopulmonary dysplasia (BPD) and adults with emphysema, requires understanding of the molecular mechanisms of alveolar development. In addition to its critical role in gas exchange, the pulmonary circulation also contributes to alveolar morphogenesis and maintenance by the production of paracrine factors termed "angiocrines" that impact the development of surrounding tissue. To identify lung angiocrines that contribute to alveolar formation, we disrupted pulmonary vascular development by conditional inactivation of the Vegf-A gene during alveologenesis. This resulted in decreased pulmonary capillary and alveolar development and altered lung elastin and retinoic acid (RA) expression. We determined that RA is produced by pulmonary endothelial cells and regulates pulmonary angiogenesis and elastin synthesis by induction of VEGF-A and FGF18, respectively. Inhibition of RA synthesis in newborn mice decreased FGF18 and elastin expression and impaired alveolarization. Treatment with RA and vitamin A partially reversed the impaired vascular and alveolar development induced by VEGF inhibition. Thus, we identified RA as a lung angiocrine that regulates alveolarization through autocrine regulation of endothelial development and paracrine regulation of elastin synthesis via induction of FGF18 in mesenchymal cells.