HHS Public Access Author manuscript Author Manuscript

J Immunol. Author manuscript; available in PMC 2017 June 01. Published in final edited form as: J Immunol. 2016 June 1; 196(11): 4692–4705. doi:10.4049/jimmunol.1501116.

Hyperoxic exposure of immature mice increases the inflammatory response to subsequent rhinovirus infection: Association with danger signals Tracy X. Cui*, Bhargavi Maheshwer*, Jun Y. Hong*, Adam M. Goldsmith*, J. Kelley Bentley*, and Antonia P. Popova*

Author Manuscript

*Department

of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI USA

Abstract Infants with a history of prematurity and bronchopulmonary dysplasia (BPD) have a high risk of asthma and viral-induced exacerbations later in life. We hypothesized that hyperoxic exposure, a predisposing factor to BPD, modulates the innate immune response, producing an exaggerated pro-inflammatory reaction to viral infection.

Author Manuscript

Two-to-3 day-old C57BL/6J mice were exposed to air or 75% oxygen for 14 days. Mice were infected intranasally with rhinovirus (RV) immediately after O2 exposure. Lung mRNA and protein expression, histology, dendritic cells (DCs) and airways responsiveness were assessed 1-12 days after infection. Tracheal aspirates from premature human infants were collected for mRNA detection. Hyperoxia increased lung IL-12 expression which persisted up to 12 days post-exposure. Hyperoxia-exposed RV-infected mice showed further increases in IL-12 and increased expression of IFN-γ, TNF-α, CCL2, CCL3 and CCL4, as well as increased airway inflammation and responsiveness. In RV-infected, air-exposed mice the response was not significant. Induced IL-12 expression in hyperoxia-exposed, RV-infected mice was associated with increased IL-12producing CD103+ lung DCs. Hyperoxia also increased expression of Clec9a, a CD103+ DCspecific damaged cell-recognition molecule. Hyperoxia increased levels of ATP metabolites and expression of adenosine receptor A1, further evidence of cell damage and related signaling. In human preterm infants, tracheal aspirate Clec9a expression positively correlated with the level of prematurity.

Author Manuscript

Hyperoxic exposure increases the activation of CD103+, Clec9a+ DCs, leading to increased inflammation and airway hyperresponsiveness upon RV infection. In premature infants, danger signal-induced DC activation may promote pro-inflammatory airway responses, thereby increasing respiratory morbidity.

Address correspondence to: Antonia P. Popova, M.D., University of Michigan Medical School, 1150 W. Medical Center Dr., Room 3570, MSRBII, Box 5688, Ann Arbor, MI USA 48109-5688. Phone: (734) 936-4200; FAX: (734) 764-3200; ; Email: [email protected].

Cui et al.

Page 2

Author Manuscript

INTRODUCTION Premature birth is associated with the development of recurrent wheezing and asthma (1, 2). Infants with bronchopulmonary dysplasia (BPD), the most common chronic pulmonary disease following premature birth (3), have increased respiratory morbidity and healthcare utilization, including increased medication use, wheezing and hospital readmissions (4–6). Infants with BPD are at particular risk for severe disease and hospitalization following respiratory viral infections including respiratory syncytial virus (RSV) (7) and rhinovirus (RV), the most common childhood viral pathogen (8–11).

Author Manuscript

The precise mechanisms leading to higher respiratory morbidity and abnormal airway function in prematurely-born children are not well understood. While early reports of lung structure in BPD demonstrated bronchial submucosal fibrosis and increased smooth muscle mass as well as hypoalveolarization and alveolar septal fibrosis (12, 13), lung structural abnormalities in the “new,” surfactant-treated BPD are relatively mild, with hypoalveolarization, little or no alveolar septal fibrosis and an absence of airway structural changes (14–16). At first blush, these structural abnormalities do not seem to completely explain the increased wheezing, airways obstruction, respiratory morbidity or susceptibility to viral infections observed in premature infants.

Author Manuscript

Instead, it is conceivable that early-life exposures may modulate the immune response, leading to asthma development. For example, neonatal hyperoxia exposure, a model of BPD, enhanced the subsequent inflammatory response to influenza A virus infection in adult mice (17). We hypothesize that neonatal hyperoxic exposure modulates the neonatal innate immune system, producing an exaggerated pro-inflammatory response to viral infection, leading to increased respiratory morbidity and airways hyperresponsiveness. To test this, we examined the effects of hyperoxic exposure on the inflammatory response to infection with RV, the most common respiratory tract infection. We found that hyperoxia increases IL-12 production from lung dendritic cells (DCs), leading to exaggerated pro-inflammatory responses to subsequent RV infection. Hyperoxic exposure also increased expression of the DC-specific damaged cell-recognition molecule Clec9a and the adenosine receptor Adora1, evidence of cell damage-related signaling.

METHODS Ethics statement

Author Manuscript

The animal experiments were performed in strict accordance with the NIH Guide for the Care and Use of Laboratory Animals recommendations. The protocol was approved by the University of Michigan Committee on Use and Care of Animals. Generation of RV RV1B or RV2 (both from American Type Culture Collection, Manassas, VA) were grown in HeLa cells, concentrated, partially purified, and titered as described (18). Viral titers were measured by plaque assay (19).

J Immunol. Author manuscript; available in PMC 2017 June 01.

Cui et al.

Page 3

Animal model

Author Manuscript

Two-to-three day-old C57BL/6J mice (Jackson Laboratories, Bar Harbor, ME) were exposed to air or 75% oxygen for up to 14 days using a polypropylene chamber coupled to an oxygen controller and sensor (BioSpherix, Lacona, NY) (20). Dams were exchanged between air and hyperoxia daily. On day 14, hyperoxic exposure was discontinued and mice were treated with either 30 μl of intranasal poly(I:C) (1 μg/μl), RV1B (3 × 108 PFU/ml), or respective control PBS or sham infection (HeLa cell lysate). Mice were sacrificed for analysis 1, 3, 5 or 12 days after poly(I:C) treatment or RV infection. Finally, selected mice were infected with RV 7 days after hyperoxic exposure (day 21 of the protocol), and lungs harvested 2 days after infection. Quantitative real-time PCR

Author Manuscript

Mouse whole-lung RNA was prepared using TRIzol (Invitrogen, Carlsbad, CA). Viral RNA and host mRNAs (IL-12p40, IFN-γ, TNF-α, CCL2, CCL3, CCL4, Clec9a, Adora1, Adora2a, Adora2b, Adora3, CD73, CD39, IFN-α, IFN-β, IFN-λ2, IL-4 and IL-13) were quantified using SYBR green technology. The primers used are listed in Table I. The level of gene expression was normalized to mRNA of β-actin using the 2−ΔCT algorithm. For graphic representation the mRNAs for the target genes relative to β-actin were plotted on the y-axis multiplied by 10−3. Measurement of lung cytokines

Author Manuscript

Measurement of AMP, ADP, ATP and adenosine

Author Manuscript

Whole lung homogenates in PBS were centrifuged and supernatants analyzed for proinflammatory cytokines. IL-12p40, IL-12p70 and TNF-α were measured by ELISA (eBioscience, San Diego, CA and R&D Systems, Minneapolis, MN) and CCL2, CCL3, CCL4, CXCL1 were detected by multiplex assay (Bio-Rad, Hercules, CA). Bronchoalveolar lavage (BAL) was performed using 0.5 ml PBS aliquots. BAL fluid was spun for 15 minutes at 1500 g, and the supernatants were analyzed.

Lung histology

Plasma and BAL fluid were obtained following 14 days of hyperoxic exposure and assayed for AMP, ADP, ATP and adenosine using mass spectroscopy. To stabilize ATP metabolite concentrations a modified stop solution with multiple enzyme inhibitors including 4.15mM Ethylenediaminetetraacetic acid, 5nM nitrobenzylthioinosine, 10μM forskolin, 100μM 3isobutyl-1-methylxanthine, 40μM dipyridamole, 10μM erythro-9-(2-hydroxy-3nonyl)adenine and 10μM 5-iodotubericidin in 40mM tricine buffer with 118mM NaCl and 5mM KCl was used (21). BAL was performed using 0.3 ml stop solution aliquots. BALF was centrifuged to remove cells and the supernatant was stored at −80°C. Plasma was obtained by collecting blood by intracardiac puncture and immediately mixing with equal volume stop solution. The samples were centrifuged to separate plasma and stored at −80°C.

Lungs were collected, fixed with 10% formaldehyde (Sigma-Aldrich, St. Louis, MO) and paraffin embedded. Five μm-thick paraffin sections were stained with Hematoxylin and eosin (H&E) (22).

J Immunol. Author manuscript; available in PMC 2017 June 01.

Cui et al.

Page 4

Flow cytometry

Author Manuscript Author Manuscript

Lungs were perfused with PBS containing EDTA (0.5 mM), minced, and digested in collagenase IV (5 mg/ml) and DNase I (23). Cells were filtered and washed with RBC lysis buffer (BD Biosciences, Franklin Lakes NJ) and kept on ice in media containing 10% serum. Dead cells were stained with Pac-Orange Live/Dead fixable dead staining dye (Invitrogen). Lung cells were then stained with fluorescent-labeled antibodies against various leukocyte surface markers (CD45, CD11b, CD11c, F4/80, CD103, MHCII, CD86, Clec9a, CD3ε, TCRβ, CD4, CD69, IFN-γ). Appropriate isotype-matched controls were used in all experiments. Antibodies were purchased from EBiosciences (San Diego, CA) or Biolegend (San Diego, CA). Cells were fixed and analyzed on a Canto2 (Becton-Dickinson, San Jose, CA) or FACSAria II (BD Biosciences) flow cytometer. Results were analyzed using FlowJo software (Tree Star, Ashland, OR). For analysis of intracellular IL-12 or IFN-γ, fresh aliquots of digested lung tissue were stimulated for 5 hours at 37°C, with Cell-stimulation Cocktail Buffer (40.5 μmol/L phorbol 12-myristate 13-acetate [PMA], 670 μmol/L ionomycin, 5.3 mmol/L brefeldin A, and 1 mmol/L monensin; eBioscience), fixed, permeabilized with Cell Permeabilization Buffer (eBioscience) and incubated with antimouse IL-12 clone C17.8 (eBioscience) or anti-mouse IFN-γ clone XMG1.2 (Biolegend). Airways responsiveness Airway cholinergic responsiveness was assessed by measuring changes in total respiratory system resistance in response to increasing doses of nebulized methacholine. Mice were anesthetized with sodium pentobarbital and a tracheostomy was performed. Mice were mechanically ventilated and total respiratory system resistance was measured plethysmographically with a Buxco FinePointe operating system (Buxco, Wilmington, NC).

Author Manuscript

Human tracheal aspirate collection and RNA preparation We examined tracheal aspirates from infants admitted to the C.S. Mott Children's Hospital Newborn Intensive Care Unit, as approved by the University of Michigan Institutional Review Board. Entry criteria included gestational age at birth ≤ 32 weeks, mechanical ventilation for respiratory distress, and age ≤ 7 days. Aspirates were collected during routine tracheal suctioning of mechanically ventilated premature infants as described (24). A 200 μl aliquot of the sample was stored in RLT buffer (Qiagen, Valencia, CA) at −80°C. Total RNA was extracted using the RNeasy Plus Mini kit (Qiagen). Statistical analysis

Author Manuscript

Data are represented as mean±SEM or median and interquartile range (IQR). Statistical significance was determined by unpaired t test, nonparametric rank-sum test, one-way or two-way ANOVA, as appropriate. Differences were pointed by Newman-Keuls' multiple comparisons test.

J Immunol. Author manuscript; available in PMC 2017 June 01.

Cui et al.

Page 5

Author Manuscript

RESULTS Hyperoxic exposure increases lung inflammatory cytokine and chemokine expression in response to poly(I:C) and RV

Author Manuscript

Two- to three-day old mice were exposed to hyperoxia or room air for 14 days. Upon discontinuing hyperoxic exposure the mice were treated with PBS, poly(I:C) or RV1B intranasally. Poly(I:C) is a synthetic analog of double-stranded RNA which is recognized by Toll-like receptor (TLR)-3 and mimics respiratory viral infection. Cytokine mRNA (Figure 1A) and protein responses (Figure 1B) were measured five days after treatment. Of the panel of inflammatory cytokines and chemokines examined, only IL-12p40 expression was increased in the PBS-treated hyperoxia-exposed mice. Poly(I:C) treatment of hyperoxiaexposed mice increased mRNA and protein expression of the type 1 cytokines IL-12p40, IFNγ, TNFα, and chemokines CCL2, CCL3 and CCL4, but had no effect on cytokine mRNA expression in air-exposed mice, except for induction of IL-12p40. The increase in IL-12p40 mRNA expression in hyperoxia-exposed, poly(I:C)-treated mouse lungs was significantly greater than that observed after poly(I:C) treatment of air-exposed animals. These results with poly(I:C) suggest the possibility that hyperoxia alters the inflammatory response to viral infection.

Author Manuscript

To test this, we examined the lung inflammatory response of hyperoxia-exposed mice to subsequent RV infection. For these experiments, mice were infected with RV immediately following 14 days of hyperoxic exposure and inflammatory cytokine and chemokine mRNA responses were measured 1, 3, 5 and 12 days after infection. As noted above, hyperoxic exposure caused a sustained increase in lung IL-12p40 mRNA expression which lasted up to 12 days after infection (Figure 2A). Similar to the responses to poly(I:C), hyperoxia-exposed mice infected with RV showed significantly higher IL-12p40, IFN-γ, CCL2, CCL3, and CCL4 levels up to 5 days after RV infection. In contrast, five days after RV infection, airexposed C57BL/6 mice did not show significant increases in cytokine mRNA expression. Cytokine protein levels tended to reflect the mRNA data (Figure 2B and C). Further, compared to air-exposed animals, RV induced mRNA expression of IL-4 and IL-13, type 2 cytokines typically associated with asthma, was decreased after hyperoxic exposure (Figure S1). Together, these results show that hyperoxic exposure amplifies the pro-inflammatory response to RV infection. To further examine the persistence of hyperoxia's effect on RV responses, we infected air- or O2-exposed mice seven days after hyperoxic exposure was discontinued. Compared to airexposed mice, hyperoxia-exposed mice showed exaggerated cytokine responses to RV infection (Figure 2D).

Author Manuscript

To define host antiviral responses, we measured mRNAs encoding IFN-α, IFN-β and IFNλ2. When compared to air exposure, hyperoxia-exposed mice showed equal induction of IFN mRNA levels 1 and 3 days after RV infection. However, IFN mRNA levels were significantly lower in hyperoxia-exposed mice at baseline and 1 and 3 days after RV infection (Figure 3A). There were no differences in IFN levels between air- and hyperoxiaexposed mice 5 days after RV infection.

J Immunol. Author manuscript; available in PMC 2017 June 01.

Cui et al.

Page 6

Author Manuscript

We next measured viral copy number in the lungs of air- and hyperoxia-exposed mice 1, 3 and 5 days after infection. The viral copy number was higher in the hyperoxia-exposed mice 1 day after infection, consistent with the lower interferon levels in this group and suggesting potential defective initial antiviral response (Figure 3B). By day 3 and 5 after infection, the viral copy number was similar in both groups, demonstrating that the augmented proinflammatory cytokine production in hyperoxia-exposed mice was not due to a sustained increase in viral load (Figure 3B). Hyperoxic exposure is associated with accumulation of inflammatory cell infiltrates in response to treatment with poly(I:C) or RV infection

Author Manuscript

To further define the effects of neonatal hyperoxic exposure on the response to subsequent viral infection, we examined mouse lung histology. Compared to control, air-exposed mouse lungs (Figure 4A), the lungs of control, hyperoxia-exposed mice showed fewer and larger alveoli up to 12 days after hyperoxia was discontinued. No other structural abnormalities or lung inflammation were noted on H&E stain. The lungs of hyperoxia-exposed, RV infected mice showed accumulation of small inflammatory cell infiltrates along the distal bronchovascular bundles both 5 and 12 days after RV infection (Figure 4A, black arrows). Lung histological evaluation 5 and 12 days after poly(I:C) treatment also showed small inflammatory cell infiltrates along the distal bronchovascular bundles of hyperoxia-exposed mice (Figure S2). In contrast, the lungs of air-exposed, poly(I:C) or RV infected mice were devoid of such infiltrates. RV infection of hyperoxia-exposed mice leads to increase in cholinergic airway responsiveness

Author Manuscript

Two-to three day old C57BL/6J mice were exposed to air or 75% oxygen for 14 days. On day of life 16, upon discontinuing hyperoxic exposure, mice were infected with RV1B. Airway cholinergic responsiveness was examined on day of life 30 (12 days post infection). Compared to control, air-exposed mice, control, hyperoxia-exposed mice had normal airways responses 12 days after exposure (Figure 4B). In contrast, there was a significant increase in airway cholinergic responsiveness in mice exposed to hyperoxia as neonates and subsequently infected with RV. Hyperoxic exposure increases the response of specific lung dendritic cell (DCs) populations to RV infection

Author Manuscript

To define the innate immune cells involved in the exaggerated and sustained cytokine and chemokine response of hyperoxia-exposed mice to RV infection, we examined lungs by flow cytometry, focusing on lung DCs, a potent source of IL-12 (25). Mouse lungs were collected 5 days after RV infection and lung DCs were differentiated from alveolar macrophages (AM) and interstitial macrophages (IM) based on F4/80 and CD11c expression (26). Compared to air-exposure, hyperoxic exposure was associated with increased lung DCs (CD45+, CD11c+, F4/80−) (Figure 5 panel D and G). In addition, we examined the two major subsets of lung CD11c+, F4/80− DCs, CD103+ and CD11bhi DCs (27). Compared to air-exposure, hyperoxic exposure increased lung CD103+ DCs (Figure 5 panel E and H). This subset of DCs expressed a high level of the activation markers MHCII and CD86 (Figure 5 panel F). Following treatment with RV, the subset of CD103+ DCs was further J Immunol. Author manuscript; available in PMC 2017 June 01.

Cui et al.

Page 7

Author Manuscript

increased in hyperoxia-exposed mice (Figure 5 panel E and H). The CD11bhi DCs were not affected by hyperoxic exposure alone, however RV infection of hyperoxia-exposed mice also increased a population of CD11bhi lung DCs. We investigated whether CD103+ DCs produce IL-12 after RV infection. RV infection after hyperoxic exposure increased the number and percent of IL-12-producing CD103+ DCs (Figure 6, panel A and B). Together, these results indicate that neonatal hyperoxic exposure increases the number of activated, lung-resident CD103+ DCs and this subset is further increased by RV infection. RV infection of hyperoxia-exposed mice was also associated with an increased in the number of CD11bhi DCs. Hyperoxic exposure induces lung CD4+ T cell activation and Th1 cytokine production

Author Manuscript

When derived from CD4+ T cells, IFN-γ is sufficient to mediate Th1 cell development (28). Th1 immune response induces airway hyperresponsiveness in severe asthma (29). Based on the increase in IFN-γ mRNA expression observed in hyperoxia-exposed mice in response to RV, we sought to evaluate the effect of prior hyperoxic exposure on lung CD4+ T cell activation and IFN-γ production in response to RV infection. Using flow cytometry we examined lungs from hyperoxia- or air-exposed mice 5 days after RV infection and quantified CD69, T cell activation marker, and IFN-γ – expressing CD4+ T cells. Hyperoxic exposure increased the population of IFN-γ+CD69+ CD4+ T cells in the lungs (Figure 7). Hyperoxia-exposed mice showed further increase in this population upon RV infection. These results imply that hyperoxic exposure induces lung CD4+ T cells activation and Th1 differentiation. Expression of Clec9a is increased during hyperoxic exposure

Author Manuscript

DCs may be activated by DAMPs (for danger-associated molecular patterns). The damaged cell-recognition molecule Clec9a, a C-type lectin, is selectively expressed on CD103+ DCs in both lymphoid and nonlymphoid organs (30, 31). We investigated whether increases in lung CD103+ DCs during hyperoxic exposure are associated with changes in Clec9a expression. We found that lung Clec9a expression was increased at 7 and 14 days of hyperoxic exposure, peaked between day 3 and 5 after the exposure was discontinued and remained increased until 12 days post exposure (Figure 8A). Using qPCR, we examined Clec9a mRNA expression of sorted lung CD103+ DCs and CD11bhi DCs. CD103+ DCs expressed higher levels of Clec9a (a small amount of Clec9a transcript was identified in CD11bhi DCs) (Figure 8B). Flow cytometry confirmed Clec9a expression by lung CD103+ DCs, and a higher number of Clec9a+ CD103+ DCs was observed in the lungs of hyperoxia exposed mice (Figure 8C). These data suggest that exposure to high oxygen tension induces expression of Clec9a, by way of increasing the number of Clec9a expressing CD103+ DCs.

Author Manuscript

Next, we examined CLEC9A mRNA expression in tracheal aspirates from human premature infants mechanically ventilated for respiratory distress syndrome in the first week of life. The degree of prematurity positively correlated with tracheal aspirate CLEC9A mRNA expression (Figure 8D), suggesting that extreme prematurity correlates with expression of the damaged cell-recognition molecule CLEC9A.

J Immunol. Author manuscript; available in PMC 2017 June 01.

Cui et al.

Page 8

Effect of hyperoxic exposure on adenosine metabolism

Author Manuscript Author Manuscript

Damaged cells release ATP and extracellular levels of ATP and its metabolites ADP, AMP and adenosine may each be increased following cellular damage induced by oxidative stress (32, 33). ATP and its metabolites represent another group of DAMP molecules capable of inducing DC activation. Extracellular adenosine acts as a signaling molecule and activates the G protein-coupled adenosine receptors A1 (Adora1), A2a (Adora2A), A2b (Adora2B) and A3 (Adora3) (34). AMP also serves as a ligand for Adora1 (35). Using qPCR, we measured whole lung expression of the four adenosine receptor subtypes, as well as CD73 (ecto-5'-nucleotidase, which converts AMP to adenosine) and CD39 (ectonucleotidase, which hydrolyzes ATP and ADP to AMP). Compared to lungs from air-exposed mice, lungs of hyperoxia-exposed mice showed increased expression of Adora1 and CD73 (Figure 9A) at 7 and 14 days of exposure. After hyperoxia was discontinued, Adora1 expression remained increased. Hyperoxia exposure decreased Adora2a mRNA expression on days 3, 7 and 14 of exposure, increased Adora2b expression at 7 days of exposure and decreased Adora3 at days 3 and 14 of exposure and had no effect on mRNA expression of CD39. We next examined plasma and BALF concentrations of adenosine, AMP, ADP and ATP by mass spectroscopy. Following 14 days of hyperoxic exposure, serum levels of AMP were significantly increased (Figure 9B). AMP:ATP and ADP:ATP ratios in plasma were also increased after 14 days of hyperoxic exposure (Figure 9C). A similar tendency was observed in BALF without reaching statistical significance. Together, these results suggest that hyperoxic exposure is associated with changes in ATP release and breakdown and, in particular, AMP/Adora1 signaling. These danger signals could play a role in DC activation and promotion of pro-inflammatory responses.

Author Manuscript

Finally, we analyzed the effect of hyperoxic exposure on Adora1 mRNA expression in response to RV infection. Adora1 mRNA expression was increased in hyperoxia-exposed mice 5 days after RV infection compared to both sham-infected hyperoxia-exposed mice and air-exposed RV-infected mice (Figure 10). These data indicate that hyperoxic exposure stimulates Adora1 signaling in response to RV infection.

DISCUSSION

Author Manuscript

In this report, we show that hyperoxic exposure of neonatal mice causes an exaggerated proinflammatory immune response to subsequent RV infection, leading to persistent airway inflammation and hyperresponsiveness. Hyperoxic exposure alone caused sustained increased expression of the inflammatory type 1 cytokine IL-12p40 and transient increased expression of IFN-γ, TNF-α, CCL2, CCL3 and CCL4. Further, infection of hyperoxiaexposed mice with RV led to increased and sustained expression of these cytokines compared to RV-infected, air-exposed mice. Hyperoxic exposure increased the number and activation of lung-resident IL-12-producing CD103+ DCs, which were further increased by RV infection. Hyperoxic exposure increased lung CD4+ T cell activation and IFN-γ production and these were further increased in response to RV infection. Hyperoxic exposure was associated with increased expression of Clec9a, a damaged cell recognition molecule. Clec9a expression was localized to CD103+ DCs and a higher number of these cells was found in the lungs of hyperoxia exposed mice. Finally, hyperoxia increased levels J Immunol. Author manuscript; available in PMC 2017 June 01.

Cui et al.

Page 9

Author Manuscript

of ATP metabolites and expression of Adora1, further evidence of cell damage and related signaling. These data suggest that hyperoxia causes danger signal-induced DC activation, leading to exaggerated pro-inflammatory airway responses to subsequent viral infection.

Author Manuscript

IL-12p40 is a subunit of the IL-12 cytokine which is produced by activated DCs and directs Th1 cell development (25). The IL-12 cytokine is a heterodimer composed of two distinct subunits, p40 and p35. When these subunits are assembled together they form biologically active IL-12. In our study, hyperoxic exposure induced overexpression of IL-12p40 mRNA which was further increased by poly(I:C) treatment and RV infection. IL-12p40 and IL-12p70 protein levels were increased in lung and BAL from hyperoxia-exposed mice infected with RV. Hyperoxia-exposed, RV-infected mice also showed increased levels of the type 1 cytokines IFN-γ and TNF-α, as well as increased airway inflammation and hyperresponsiveness. Together these data suggest that, in our model, hyperoxia-induced IL-12 overexpression drives an exaggerated inflammatory response to subsequent respiratory viral infection. Development of a Th1 response and induction of IL-12 or IFN-γ have each been associated with resolution of symptoms in viral infections (36, 37). Although IL-12 appears to be protective during respiratory viral infections, overproduction of IL-12, as observed in sepsis or in the absence of anti-inflammatory IL-10 production, leads to hyperinflammatory responses (38, 39). Furthermore, hyperoxic exposure of mature mice induces IL-12 production and IFN-γ-mediated lung injury (40), consistent with the notion that, under selected conditions, IL-12 could favor an adverse exaggerated type 1 cytokine response to subsequent infections or other adverse stimuli. We believe that, in our model, overproduction of IL-12 leads to similar hyperinflammatory responses.

Author Manuscript Author Manuscript

Next, we endeavored to determine the source of IL-12 production. Mouse lung DCs contain two major subpopulations: CD103+ CD11blow (or CD11b-negative) DCs and CD103negative CD11bhigh DCs (41–43). CD103+ DCs produce IL-12 and IFN-γ (44–46) and predominantly elicit Th1 and Th17 responses (47), whereas CD11bhigh DCs primarily provoke a Th2 response (47, 48). We found that hyperoxic exposure was associated with an increase in lung CD103+, MHCIIhi, CD86hi DCs, a population of cells which was further increased upon viral infection. CD103+ DCs produced IL-12 when stimulated ex vivo. Together these data suggest that hyperoxia-induced CD103+ DCs are responsible for the exaggerated pro-inflammatory response to subsequent respiratory viral infection. The mechanisms responsible for the increase in lung CD103+ DCs in hyperoxia-exposed mice are not well understood. While it is possible that, under hyperoxic conditions, recruited CD103+ DC progenitors differentiate and become activated (49), we cannot exclude the possibility that the activated CD103+ DCs have an impaired capacity to migrate to the regional lymph nodes as observed during viral infections or in the setting of altered extracellular ATP gradients (50, 51). Lung DCs form a network of cells that interact with the outside environment and express pathogen-associated molecular pattern (PAMP) and damage-associated molecular pattern (DAMP) receptors that recognize microbes and danger signals. Engagement of PAMP and DAMP receptors triggers DC activation and maturation. Activated DCs upregulate MHC class II and the co-stimulatory molecules CD80 and CD86 and migrate to the regional lymph nodes, where they stimulate naïve CD4+ T cells and direct their differentiation to

J Immunol. Author manuscript; available in PMC 2017 June 01.

Cui et al.

Page 10

Author Manuscript

various T-cell lineages (52, 53). We speculate that hyperoxic exposure generates danger signals which activate CD103+ DCs. Hyperoxic exposure has been associated with lung epithelial cell apoptosis and necrosis (54) and dead cells stimulate DC maturation and migration of mature DCs into draining lymph nodes (55, 56). CD103+ DCs, unlike CD11b+ DCs, express receptors for apoptotic cells along with the machinery to cross-present phagocytosed dead cells (57). Prolonged hyperoxic exposure and resultant oxidative stress are strong inducers of various proinflammatory cytokines in airway cells and pulmonary tissue, and lead to influx of macrophages and neutrophils (58, 59). Here we propose a previously unrecognized mechanism for oxidant-associated inflammatory cell influx: hyperoxia-induced cell injury and death (60) leads to release of “danger signals” that activate the innate immune system and increase inflammatory responses.

Author Manuscript Author Manuscript

Clec9a, also referred to as DNGR-1, is a DC receptor that couples sensing of cell death to innate immune responses. Clec9a diverts dead-cell-associated antigens away from lysosomal compartments to allow retrieval of antigens for cross-presentation to T cells (30, 61, 62). Clec9a appears to recognize particular forms of filamentous actin, perhaps in association with particular actin-binding domains of cytoskeletal proteins (63). Our studies indicate that hyperoxia increases the lung expression of Clec9a, reflecting the increased number of Clec9a+ CD103+ DCs. Clec9a expression is sustained increased after hyperoxia is discontinued, suggesting persistence of a larger population of CD103+ DCs with activated danger signaling. Furthermore, an analysis of tracheal aspirates from premature human infants showed that the level of prematurity positively correlated with respiratory tract Clec9a mRNA expression. Since hyperoxic exposure causes respiratory epithelial cell death (64–66), we speculate that premature infants experience greater epithelial cell death upon exposure to high oxygen tensions relative to that in utero, leading to expansion of Clec9a+ CD103+ DCs.

Author Manuscript

Following tissue injury, damaged cells release ATP and ADP in the extracellular space, which are then dephosphorylated to AMP by the ectonucleoside triphosphate diphosphohydrolase CD39 (67). AMP is subsequently dephosphorylated to adenosine by the ecto-5'-nucleotidase CD73 (68). Extracellular adenosine has been linked to regulation of inflammation and tissue repair (69). Adenosine signals through one of four G-protein coupled, cell surface receptors (Adora1, Adora2a, Adora2b, Adora3) (70), all of which are expressed on DCs (71), macrophages (72) neutrophils (73) and respiratory epithelial cells (74). AMP also signals through Adora1 (35). Adora1 exerts a pro-inflammatory response, while engagement of Adora2a inhibits adherence and activation of leukocytes (75). Our studies identified increased Adora1 and CD73 expression at 7 and 14 days of hyperoxic exposure, while Adora2a expression was decreased. Adora1 expression remained increased after hyperoxia was discontinued, suggesting persistent activation of danger signaling. Hyperoxic exposure was associated with increased plasma levels of AMP, as well as increased AMP:ATP and ADP:ATP ratios. A similar tendency was observed in BALF. Together these data are consistent with the notion that neonatal hyperoxic exposure induces airway inflammation by increasing ATP metabolism and AMP-Adora1 signaling. Despite increased CD73 expression, adenosine levels were not increased, perhaps due to increased adenosine uptake. Furthermore, we observed significantly increased Adora1 mRNA

J Immunol. Author manuscript; available in PMC 2017 June 01.

Cui et al.

Page 11

Author Manuscript

expression in hyperoxia-exposed mice 5 days after RV infection, suggesting that hyperoxic exposure promotes Adora1 signaling in response to RV infection.

Author Manuscript

Premature infants and babies with BPD have increased respiratory morbidity including wheezing, asthma, medication use and hospitalizations with respiratory viral infection (1– 11). Atopy has not been associated with increased respiratory symptoms in prematurely born infants and infants with BPD (76, 77). The precise mechanisms leading to higher respiratory morbidity and abnormal airway function in prematurely-born children are not well understood. Lung structural abnormalities in the “new,” surfactant-treated BPD are relatively mild, with only hypoalveolarization but no structural airway changes (14–16). These abnormalities do not seem to explain the increased wheezing, airways obstruction, respiratory morbidity or susceptibility to viral infections observed in premature infants. It is conceivable that hypoalveolarization predisposes to airway narrowing by reducing the number of alveolar attachments (78). Airway smooth muscle thickening has been noted in hyperoxia-exposed mice (79, 80) but this finding has not been verified in human infants. We therefore sought an alternative explanation for the observed respiratory morbidity, the details of which we describe above. Few animal studies have investigated the effect of neonatal hyperoxic exposure on response to respiratory viral infection. Short-term neonatal hyperoxic exposure increased the subsequent inflammatory response to influenza A virus infection in mice infected at 8–9 weeks of age (17). Adult mice with prior hyperoxia exposure as neonates showed increased leukocyte recruitment to the lungs and higher levels of CCL2 in BAL fluid up to 9 days after infection. We also found increased CCL2 mRNA expression in hyperoxia-exposed, RV-infected mice. However, we extend these previous results by providing specific cellular and molecular mechanisms responsible for the observed inflammatory response.

Author Manuscript

We examined the inflammatory response to respiratory viral infection in immature mice, thereby simulating the clinical experience of children with BPD becoming sick with respiratory viruses. Animal studies with neonatal rhinovirus infection demonstrate that depending on age at the time of RV infection, the inflammatory response can be skewed towards Type 2 (< 8 days of age) or Type 1 (> 8 days of age) (23). Our findings are consistent with the above study, as RV infection on day of life 16 induced Type 1 responses.

Author Manuscript

In our model, the initial priming of the innate immune system by hyperoxia-induced DAMP signaling increased IL-12 expression and directed subsequent hyperinflammatory Th1 responses. Further investigation will be required to determine the requirement and sufficiency of IL-12 for exaggerated airway inflammation and hyperresponsiveness to RV infection. Also, since IL-10 is a potent anti-inflammatory cytokine (81) and negative regulator of IL-12 production (82), future studies examining the capacity of IL-10 treatment to block the observed hyperinflammatory response to RV are warranted. In summary, early life hyperoxic exposure increased the activation of CD103+ DCs, leading to increased type 1 inflammation and airway hyperresponsiveness upon RV infection. The expression of different DAMP receptors and their ligands was increased during immature mice hyperoxic exposure. In premature infants with a history of BPD, danger signal-induced

J Immunol. Author manuscript; available in PMC 2017 June 01.

Cui et al.

Page 12

Author Manuscript

DC activation may promote pro-inflammatory airway responses, thereby increasing respiratory morbidity.

Supplementary Material Refer to Web version on PubMed Central for supplementary material.

ACKNOWLEDGEMENTS We thank Dr. Marc Hershenson from the University of Michigan Medical School for his constructive critique of this research. We also thank Dr. Scott Visovatti from the University of Michigan Medical School and Dr. Chunhai Ruan from the Michigan Regional Comprehensive Metabolomics Resource Core at the University of Michigan for their help with the metabolomic analysis. This work was supported by NIH grant K23 HL109149.

Author Manuscript

REFERENCES

Author Manuscript Author Manuscript

1. Been JV, Lugtenberg MJ, Smets E, van Schayck CP, Kramer BW, Mommers M, Sheikh A. Preterm birth and childhood wheezing disorders: a systematic review and meta-analysis. PLoS Med. 2014; 11:e1001596. [PubMed: 24492409] 2. Jaakkola JJ, Ahmed P, Ieromnimon A, Goepfert P, Laiou E, Quansah R, Jaakkola MS. Preterm delivery and asthma: a systematic review and meta-analysis. J. Allergy Clin. Immunol. 2006; 118:823–830. [PubMed: 17030233] 3. Stoll BJ, Hansen NI, Bell EF, Shankaran S, Laptook AR, Walsh MC, Hale EC, Newman NS, Schibler K, Carlo WA, Kennedy KA, Poindexter BB, Finer NN, Ehrenkranz RA, Duara S, Sanchez PJ, O'Shea TM, Goldberg RN, Van Meurs KP, Faix RG, Phelps DL, Frantz ID 3rd, Watterberg KL, Saha S, Das A, Higgins RD, H. Eunice Kennedy Shriver National Institute of Child, and N. Human Development Neonatal Research. Neonatal outcomes of extremely preterm infants from the NICHD Neonatal Research Network. Pediatrics. 2010; 126:443–456. [PubMed: 20732945] 4. Greenough A, Alexander J, Burgess S, Bytham J, Chetcuti PA, Hagan J, Lenney W, Melville S, Shaw NJ, Boorman J, Coles S, Pang F, Turner J. Preschool healthcare utilisation related to home oxygen status. Arch. Dis. Child. Fetal Neonatal Ed. 2006; 91:F337–341. [PubMed: 16705008] 5. Fawke J, Lum S, Kirkby J, Hennessy E, Marlow N, Rowell V, Thomas S, Stocks J. Lung function and respiratory symptoms at 11 years in children born extremely preterm: the EPICure study. Am. J. Respir. Crit. Care Med. 2010; 182:237–245. [PubMed: 20378729] 6. Ehrenkranz RA, Walsh MC, Vohr BR, Jobe AH, Wright LL, Fanaroff AA, Wrage LA, Poole K, H. National Institutes of Child, and N. Human Development Neonatal Research. Validation of the National Institutes of Health consensus definition of bronchopulmonary dysplasia. Pediatrics. 2005; 116:1353–1360. [PubMed: 16322158] 7. Broughton S, Roberts A, Fox G, Pollina E, Zuckerman M, Chaudhry S, Greenough A. Prospective study of healthcare utilisation and respiratory morbidity due to RSV infection in prematurely born infants. Thorax. 2005; 60:1039–1044. [PubMed: 16227330] 8. Bennett NJ, Tabarani CM, Bartholoma NM, Wang D, Huang D, Riddell SW, Kiska DL, Hingre R, Rosenberg HF, Domachowske JB. Unrecognized viral respiratory tract infections in premature infants during their birth hospitalization: a prospective surveillance study in two neonatal intensive care units. J. Pediatr. 2012; 161:814–818. [PubMed: 22694859] 9. Drysdale SB, Alcazar-Paris M, Wilson T, Smith M, Zuckerman M, Broughton S, Rafferty GF, Peacock JL, Johnston SL, Greenough A. Rhinovirus infection and healthcare utilisation in prematurely born infants. Eur. Respir. J. 2013; 42:1029–1036. [PubMed: 23563263] 10. Linder JE, Kraft DC, Mohamed Y, Lu Z, Heil L, Tollefson S, Saville BR, Wright PF, Williams JV, Miller EK. Human rhinovirus C: Age, season, and lower respiratory illness over the past 3 decades. J. Allergy Clin. Immunol. 2013; 131:69–77. e61–66. [PubMed: 23146382]

J Immunol. Author manuscript; available in PMC 2017 June 01.

Cui et al.

Page 13

Author Manuscript Author Manuscript Author Manuscript Author Manuscript

11. Costa LF, Queiroz DA, Lopes da Silveira H, Bernardino Neto M, de Paula NT, Oliveira TF, Tolardo AL, Yokosawa J. Human rhinovirus and disease severity in children. Pediatrics. 2014; 133:e312–321. [PubMed: 24420808] 12. Margraf LR, Tomashefski JF Jr. Bruce MC, Dahms BB. Morphometric analysis of the lung in bronchopulmonary dysplasia. Am. Rev. Respir. Dis. 1991; 143:391–400. [PubMed: 1990959] 13. Stocker JT. Pathologic features of long-standing “healed” bronchopulmonary dysplasia: a study of 28 3- to 40-month-old infants. Hum. Pathol. 1986; 17:943–961. [PubMed: 3639056] 14. Hussain NA, Siddiqui NH, Stocker JR. Pathology of arrested acinar development in postsurfactant bronchopulmonary dysplasia. Hum. Pathol. 1998; 29:710–717. [PubMed: 9670828] 15. Jobe AH, Bancalari E. Bronchopulmonary dysplasia. Am. J. Respir. Crit. Care Med. 2001; 163:1723–1729. [PubMed: 11401896] 16. Coalson JJ. Pathology of new bronchopulmonary dysplasia. Semin. Neonatol. 2003; 8:73–81. [PubMed: 12667832] 17. O'Reilly MA, Marr SH, Yee M, McGrath-Morrow SA, Lawrence BP. Neonatal hyperoxia enhances the inflammatory response in adult mice infected with influenza A virus. Am. J. Respir. Crit. Care Med. 2008; 177:1103–1110. [PubMed: 18292469] 18. Newcomb DC, Sajjan U, Nanua S, Jia Y, Goldsmith AM, Bentley JK, Hershenson MB. Phosphatidylinositol 3-Kinase Is Required for Rhinovirus-induced Airway Epithelial Cell Interleukin-8 Expression. J. Biol. Chem. 2005; 280:36952–36961. [PubMed: 16120607] 19. Martin S, Casasnovas JM, Staunton DE, Springer TA. Efficient neutralization and disruption of rhinovirus by chimeric ICAM-1/immunoglobulin molecules. J. Virol. 1993; 67:3561–3568. [PubMed: 8098781] 20. Popova AP, Bentley JK, Anyanwu AC, Richardson MN, Linn MJ, Lei J, Wong EJ, Goldsmith AM, Pryhuber GS, Hershenson MB. Glycogen synthase kinase-3beta/beta-catenin signaling regulates neonatal lung mesenchymal stromal cell myofibroblastic differentiation. Am. J. Physiol. Lung Cell. Mol. Physiol. 2012; 303:L439–448. [PubMed: 22773696] 21. Gorman MW, Marble DR, Ogimoto K, Feigl EO. Measurement of adenine nucleotides in plasma. Luminescence. 2003; 18:173–181. [PubMed: 12701093] 22. Bentley JK, Chen Q, Hong JY, Popova AP, Lei J, Moore BB, Hershenson MB. Periostin is required for maximal airways inflammation and hyperresponsiveness in mice. J. Allergy Clin. Immunol. 2014; 134:1433–1442. [PubMed: 24996263] 23. Hong JY, Bentley JK, Chung Y, Lei J, Steenrod JM, Chen Q, Sajjan US, Hershenson MB. Neonatal rhinovirus induces mucous metaplasia and airways hyperresponsiveness through IL-25 and type 2 innate lymphoid cells. J. Allergy Clin. Immunol. 2014; 134:429–439. [PubMed: 24910174] 24. Hennrick KT, Keeton AG, Nanua S, Kijek TG, Goldsmith AM, Sajjan US, Bentley JK, Lama VN, Moore BB, Schumacher RE, Thannickal VJ, Hershenson MB. Lung cells from neonates show a mesenchymal stem cell phenotype. Am. J. Respir. Crit. Care Med. 2007; 175:1158–1164. [PubMed: 17332484] 25. Macatonia SE, Hosken NA, Litton M, Vieira P, Hsieh CS, Culpepper JA, Wysocka M, Trinchieri G, Murphy KM, O'Garra A. Dendritic cells produce IL-12 and direct the development of Th1 cells from naive CD4+ T cells. J. Immunol. 1995; 154:5071–5079. [PubMed: 7730613] 26. Bedoret D, Wallemacq H, Marichal T, Desmet C, Quesada Calvo F, Henry E, Closset R, Dewals B, Thielen C, Gustin P, de Leval L, Van Rooijen N, Le Moine A, Vanderplasschen A, Cataldo D, Drion PV, Moser M, Lekeux P, Bureau F. Lung interstitial macrophages alter dendritic cell functions to prevent airway allergy in mice. J. Clin. Invest. 2009; 119:3723–3738. [PubMed: 19907079] 27. Nakano H, Free ME, Whitehead GS, Maruoka S, Wilson RH, Nakano K, Cook DN. Pulmonary CD103(+) dendritic cells prime Th2 responses to inhaled allergens. Mucosal Immunol. 2012; 5:53–65. [PubMed: 22012243] 28. Wakil AE, Wang ZE, Ryan JC, Fowell DJ, Locksley RM. Interferon gamma derived from CD4(+) T cells is sufficient to mediate T helper cell type 1 development. J. Exp. Med. 1998; 188:1651– 1656. [PubMed: 9802977]

J Immunol. Author manuscript; available in PMC 2017 June 01.

Cui et al.

Page 14

Author Manuscript Author Manuscript Author Manuscript Author Manuscript

29. Raundhal M, Morse C, Khare A, Oriss TB, Milosevic J, Trudeau J, Huff R, Pilewski J, Holguin F, Kolls J, Wenzel S, Ray P, Ray A. High IFN-gamma and low SLPI mark severe asthma in mice and humans. J. Clin. Invest. 2015; 125:3037–3050. [PubMed: 26121748] 30. Sancho D, Joffre OP, Keller AM, Rogers NC, Martinez D, Hernanz-Falcon P, Rosewell I, R. e. Sousa C. Identification of a dendritic cell receptor that couples sensing of necrosis to immunity. Nature. 2009; 458:899–903. [PubMed: 19219027] 31. Poulin LF, Reyal Y, Uronen-Hansson H, Schraml BU, Sancho D, Murphy KM, Hakansson UK, Moita LF, Agace WW, Bonnet D, Reis e Sousa C. DNGR-1 is a specific and universal marker of mouse and human Batf3-dependent dendritic cells in lymphoid and nonlymphoid tissues. Blood. 2012; 119:6052–6062. [PubMed: 22442345] 32. Ahmad S, Ahmad A, Ghosh M, Leslie CC, White CW. Extracellular ATP-mediated signaling for survival in hyperoxia-induced oxidative stress. J. Biol. Chem. 2004; 279:16317–16325. [PubMed: 14761947] 33. Davies J, Karmouty-Quintana H, Le TT, Chen NY, Weng T, Luo F, Molina J, Moorthy B, Blackburn MR. Adenosine promotes vascular barrier function in hyperoxic lung injury. Physiol. Rep. 2014; 2:e12155. [PubMed: 25263205] 34. Fredholm BB, AP IJ, Jacobson KA, Linden J, Muller CE. International Union of Basic and Clinical Pharmacology. LXXXI. Nomenclature and classification of adenosine receptors--an update. Pharmacol. Rev. 2011; 63:1–34. [PubMed: 21303899] 35. Rittiner JE, Korboukh I, Hull-Ryde EA, Jin J, Janzen WP, Frye SV, Zylka MJ. AMP is an adenosine A1 receptor agonist. J. Biol. Chem. 2012; 287:5301–5309. [PubMed: 22215671] 36. Tekkanat KK, Maassab H, Berlin AA, Lincoln PM, Evanoff HL, Kaplan MH, Lukacs NW. Role of interleukin-12 and stat-4 in the regulation of airway inflammation and hyperreactivity in respiratory syncytial virus infection. Am. J. Pathol. 2001; 159:631–638. [PubMed: 11485921] 37. Durbin JE, Johnson TR, Durbin RK, Mertz SE, Morotti RA, Peebles RS, Graham BS. The role of IFN in respiratory syncytial virus pathogenesis. J. Immunol. 2002; 168:2944–2952. [PubMed: 11884466] 38. Gazzinelli RT, Wysocka M, Hieny S, Scharton-Kersten T, Cheever A, Kuhn R, Muller W, Trinchieri G, Sher A. In the absence of endogenous IL-10, mice acutely infected with Toxoplasma gondii succumb to a lethal immune response dependent on CD4+ T cells and accompanied by overproduction of IL-12, IFN-gamma and TNF-alpha. J. Immunol. 1996; 157:798–805. [PubMed: 8752931] 39. Fukao T, Tanabe M, Terauchi Y, Ota T, Matsuda S, Asano T, Kadowaki T, Takeuchi T, Koyasu S. PI3K-mediated negative feedback regulation of IL-12 production in DCs. Nat. Immunol. 2002; 3:875–881. [PubMed: 12154357] 40. Yamada M, Kubo H, Kobayashi S, Ishizawa K, Sasaki H. Interferon-gamma: a key contributor to hyperoxia-induced lung injury in mice. Am. J. Physiol. Lung Cell. Mol. Physiol. 2004; 287:L1042–1047. [PubMed: 15257986] 41. Sung S-SJ, Fu SM, Rose CE, Gaskin F, Ju S-T, Beaty SR. A Major Lung CD103 (αE)-β7 IntegrinPositive Epithelial Dendritic Cell Population Expressing Langerin and Tight Junction Proteins. J. Immunol. 2006; 176:2161–2172. [PubMed: 16455972] 42. Valladeau J, Clair-Moninot V, Dezutter-Dambuyant C, Pin J-J, Kissenpfennig A, Mattéi M-G, AitYahia S, Bates EEM, Malissen B, Koch F, Fossiez F, Romani N, Lebecque S, Saeland S. Identification of Mouse Langerin/CD207 in Langerhans Cells and Some Dendritic Cells of Lymphoid Tissues. J. Immunol. 2002; 168:782–792. [PubMed: 11777972] 43. Vermaelen K, Pauwels R. Pulmonary dendritic cells. Am. J. Respir. Crit. Care Med. 2005; 172:530–551. [PubMed: 15879415] 44. Leepiyasakulchai C, Taher C, Chuquimia OD, Mazurek J, Soderberg-Naucler C, Fernandez C, Skold M. Infection rate and tissue localization of murine IL-12p40-producing monocyte-derived CD103(+) lung dendritic cells during pulmonary tuberculosis. PLoS One. 2013; 8:e69287. [PubMed: 23861965] 45. Lantier L, Lacroix-Lamande S, Potiron L, Metton C, Drouet F, Guesdon W, Gnahoui-David A, Le Vern Y, Deriaud E, Fenis A, Rabot S, Descamps A, Werts C, Laurent F. Intestinal CD103+

J Immunol. Author manuscript; available in PMC 2017 June 01.

Cui et al.

Page 15

Author Manuscript Author Manuscript Author Manuscript Author Manuscript

dendritic cells are key players in the innate immune control of Cryptosporidium parvum infection in neonatal mice. PLoS Pathog. 2013; 9:e1003801. [PubMed: 24367259] 46. Martinez-Lopez M, Iborra S, Conde-Garrosa R, Sancho D. Batf3-dependent CD103+ dendritic cells are major producers of IL-12 that drive local Th1 immunity against Leishmania major infection in mice. Eur. J. Immunol. 2015; 45:119–129. [PubMed: 25312824] 47. Furuhashi K, Suda T, Hasegawa H, Suzuki Y, Hashimoto D, Enomoto N, Fujisawa T, Nakamura Y, Inui N, Shibata K, Nakamura H, Chida K. Mouse lung CD103+ and CD11bhigh dendritic cells preferentially induce distinct CD4+ T-cell responses. Am. J. Respir. Cell Mol. Biol. 2012; 46:165– 172. [PubMed: 21908266] 48. Zhou Q, Ho AW, Schlitzer A, Tang Y, Wong KH, Wong FH, Chua YL, Angeli V, Mortellaro A, Ginhoux F, Kemeny DM. GM-CSF-licensed CD11b+ lung dendritic cells orchestrate Th2 immunity to Blomia tropicalis. J. Immunol. 2014; 193:496–509. [PubMed: 24943219] 49. Jakubzick C, Tacke F, Ginhoux F, Wagers AJ, van Rooijen N, Mack M, Merad M, Randolph GJ. Blood Monocyte Subsets Differentially Give Rise to CD103+ and CD103− Pulmonary Dendritic Cell Populations. J. Immunol. 2008; 180:3019–3027. [PubMed: 18292524] 50. Cirone M, Conte V, Farina A, Valia S, Trivedi P, Granato M, Santarelli R, Frati L, Faggioni A. HHV-8 reduces dendritic cell migration through down-regulation of cell-surface CCR6 and CCR7 and cytoskeleton reorganization. Virol. J. 2012; 9:92. [PubMed: 22583958] 51. Schnurr M, Toy T, Stoitzner P, Cameron P, Shin A, Beecroft T, Davis ID, Cebon J, Maraskovsky E. ATP gradients inhibit the migratory capacity of specific human dendritic cell types: implications for P2Y11 receptor signaling. Blood. 2003; 102:613–620. [PubMed: 12649135] 52. Mempel TR, Henrickson SE, von Andrian UH. T-cell priming by dendriticcells in lymph nodes occurs in three distinct phases. Nature. 2004; 427:154–159. [PubMed: 14712275] 53. Zhu J, Yamane H, Paul WE. Differentiation of effector CD4 T cell populations (*). Annu. Rev. Immunol. 2010; 28:445–489. [PubMed: 20192806] 54. Barazzone C, Horowitz S, Donati YR, Rodriguez I, Piguet PF. Oxygen toxicity in mouse lung: pathways to cell death. Am. J. Respir. Cell Mol. Biol. 1998; 19:573–581. [PubMed: 9761753] 55. Gallucci S, Lolkema M, Matzinger P. Natural adjuvants: endogenous activators of dendritic cells. Nat. Med. 1999; 5:1249–1255. [PubMed: 10545990] 56. Shi Y, Rock KL. Cell death releases endogenous adjuvants that selectively enhance immune surveillance of particulate antigens. Eur. J. Immunol. 2002; 32:155–162. [PubMed: 11754356] 57. Desch AN, Randolph GJ, Murphy K, Gautier EL, Kedl RM, Lahoud MH, Caminschi I, Shortman K, Henson PM, Jakubzick CV. CD103+ pulmonary dendritic cells preferentially acquire and present apoptotic cell-associated antigen. J. Exp. Med. 2011; 208:1789–1797. [PubMed: 21859845] 58. Wagenaar GT, ter Horst SA, van Gastelen MA, Leijser LM, Mauad T, van der Velden PA, de Heer E, Hiemstra PS, Poorthuis BJ, Walther FJ. Gene expression profile and histopathology of experimental bronchopulmonary dysplasia induced by prolonged oxidative stress. Free Radic. Biol. Med. 2004; 36:782–801. [PubMed: 14990357] 59. Rozycki HJ, Comber PG, Huff TF. Cytokines and oxygen radicals after hyperoxia in preterm and term alveolar macrophages. Am. J. Physiol. Lung Cell. Mol. Physiol. 2002; 282:L1222–1228. [PubMed: 12003777] 60. Tanaka A, Jin Y, Lee SJ, Zhang M, Kim HP, Stolz DB, Ryter SW, Choi AM. Hyperoxia-induced LC3B interacts with the Fas apoptotic pathway in epithelial cell death. Am. J. Respir. Cell Mol. Biol. 2012; 46:507–514. [PubMed: 22095627] 61. Poulin LF, Reyal Y, Uronen-Hansson H, Schraml BU, Sancho D, Murphy KM, Håkansson UK, Ferreira Moita L, Agace WW, Bonnet D, Reis e Sousa C. DNGR-1 is a specific and universal marker of mouse and human Batf3-dependent dendritic cells in lymphoid and nonlymphoid tissues. Blood. 2012; 119:6052–62. [PubMed: 22442345] 62. Zelenay S, Keller AM, Whitney PG, Schraml BU, Deddouche S, Rogers NC, Schulz O, Sancho D, Reis e Sousa C. The dendritic cell receptor DNGR-1 controls endocytic handling of necrotic cell antigens to favor cross-priming of CTLs in virus-infected mice. J. Clin. Invest. 2012; 122:1615– 1627. [PubMed: 22505458]

J Immunol. Author manuscript; available in PMC 2017 June 01.

Cui et al.

Page 16

Author Manuscript Author Manuscript Author Manuscript Author Manuscript

63. Zhang JG, Czabotar PE, Policheni AN, Caminschi I, Wan SS, Kitsoulis S, Tullett KM, Robin AY, Brammananth R, van Delft MF, Lu J, O'Reilly LA, Josefsson EC, Kile BT, Chin WJ, Mintern JD, Olshina MA, Wong W, Baum J, Wright MD, Huang DC, Mohandas N, Coppel RL, Colman PM, Nicola NA, Shortman K, Lahoud MH. The dendritic cell receptor Clec9A binds damaged cells via exposed actin filaments. Immunity. 2012; 36:646–657. [PubMed: 22483802] 64. Maniscalco WM, Watkins RH, Roper JM, Staversky R, O'Reilly MA. Hyperoxic ventilated premature baboons have increased p53, oxidant DNA damage and decreased VEGF expression. Pediatr. Res. 2005; 58:549–556. [PubMed: 16148072] 65. Franco-Montoya ML, Bourbon JR, Durrmeyer X, Lorotte S, Jarreau PH, Delacourt C. Pulmonary effects of keratinocyte growth factor in newborn rats exposed to hyperoxia. Am. J. Physiol. Lung Cell. Mol. Physiol. 2009; 297:L965–976. [PubMed: 19700645] 66. Choo-Wing R, Syed MA, Harijith A, Bowen B, Pryhuber G, Janer C, Andersson S, Homer RJ, Bhandari V. Hyperoxia and interferon-gamma-induced injury in developing lungs occur via cyclooxygenase-2 and the endoplasmic reticulum stress-dependent pathway. Am. J. Respir. Cell Mol. Biol. 2013; 48:749–757. [PubMed: 23470621] 67. Kaczmarek E, Koziak K, Sévigny J, Siegel JB, Anrather J, Beaudoin AR, Bach FH, Robson SC. Identification and Characterization of CD39/Vascular ATP Diphosphohydrolase. J. Biol. Chem. 1996; 271:33116–33122. [PubMed: 8955160] 68. Resta R, Yamashita Y, Thompson LF. Ecto-enzyme and signaling functions of lymphocyte CD73. Immunol. Rev. 1998; 161:95–109. [PubMed: 9553767] 69. Zhou Y, Schneider DJ, Blackburn MR. Adenosine signaling and the regulation of chronic lung disease. Pharmacol. Ther. 2009; 123:105–116. [PubMed: 19426761] 70. Poth JM, Brodsky K, Ehrentraut H, Grenz A, Eltzschig HK. Transcriptional control of adenosine signaling by hypoxia-inducible transcription factors during ischemic or inflammatory disease. J. Mol. Med. (Berl.). 2013; 91:183–193. [PubMed: 23263788] 71. Hasko G, Cronstein BN. Adenosine: an endogenous regulator of innate immunity. Trends Immunol. 2004; 25:33–39. [PubMed: 14698282] 72. Hasko G, Pacher P. Regulation of macrophage function by adenosine. Arterioscler. Thromb. Vasc. Biol. 2012; 32:865–869. [PubMed: 22423038] 73. Barletta KE, Ley K, Mehrad B. Regulation of neutrophil function by adenosine. Arterioscler. Thromb. Vasc. Biol. 2012; 32:856–864. [PubMed: 22423037] 74. Factor P, Mutlu GM, Chen L, Mohameed J, Akhmedov AT, Meng FJ, Jilling T, Lewis ER, Johnson MD, Xu A, Kass D, Martino JM, Bellmeyer A, Albazi JS, Emala C, Lee HT, Dobbs LG, Matalon S. Adenosine regulation of alveolar fluid clearance. Proc. Natl. Acad. Sci. U. S. A. 2007; 104:4083–4088. [PubMed: 17360481] 75. Nakav S, Chaimovitz C, Sufaro Y, Lewis EC, Shaked G, Czeiger D, Zlotnik M, Douvdevani A. Anti-inflammatory preconditioning by agonists of adenosine A1 receptor. PLoS One. 2008; 3:e2107. [PubMed: 18461129] 76. Broström EB, Thunqvist P, Adenfelt G, Borling E, Katz-Salamon M. Obstructive lung disease in children with mild to severe BPD. Respir. Med. 2010; 104:362–370. [PubMed: 19906521] 77. Siltanen M, Savilahti E, Pohjavuori M, Kajosaari M. Respiratory symptoms and lung function in relation to atopy in children born preterm. Pediatr. Pulmonol. 2004; 37:43–49. [PubMed: 14679488] 78. Hershenson MB, Aghili S, Punjabi N, Hernandez C, Ray DW, Garland A, Glagov S, Solway J. Hyperoxia-induced airway hyperresponsiveness and remodeling in immature rats. Am. J. Physiol. 1992; 262:L263–269. [PubMed: 1550249] 79. Wang H, Jafri A, Martin RJ, Nnanabu J, Farver C, Prakash YS, MacFarlane PM. Severity of neonatal hyperoxia determines structural and functional changes in developing mouse airway. Am. J. Physiol. Lung Cell. Mol. Physiol. 2014; 307:L295–301. [PubMed: 24951774] 80. O'Reilly M, Harding R, Sozo F. Altered small airways in aged mice following neonatal exposure to hyperoxic gas. Neonatology. 2014; 105:39–45. [PubMed: 24281398] 81. Bogdan C, Vodovotz Y, Nathan C. Macrophage deactivation by interleukin 10. J. Exp. Med. 1991; 174:1549–1555. [PubMed: 1744584]

J Immunol. Author manuscript; available in PMC 2017 June 01.

Cui et al.

Page 17

Author Manuscript

82. Berg DJ, Kuhn R, Rajewsky K, Muller W, Menon S, Davidson N, Grunig G, Rennick D. Interleukin-10 is a central regulator of the response to LPS in murine models of endotoxic shock and the Shwartzman reaction but not endotoxin tolerance. J. Clin. Invest. 1995; 96:2339–2347. [PubMed: 7593621]

Author Manuscript Author Manuscript Author Manuscript J Immunol. Author manuscript; available in PMC 2017 June 01.

Cui et al.

Page 18

Author Manuscript Author Manuscript

Figure 1. Hyperoxic exposure increases lung pro-inflammatory cytokine expression in response to poly(I:C)

Author Manuscript

Two-to-3 day-old mice were exposed to air or 75% oxygen for 14 days and treated intranasally with 30 μl of intranasal poly(I:C) (1 μg/μl) or PBS immediately after O2 exposure. Five days after treatment control hyperoxia-exposed mice showed increased IL-12p40 mRNA (A) and protein (B) expression. Compared to air-exposed, poly(I:C)treated mice, hyperoxia-exposed, poly(I:C)-treated mice showed increased mRNA (A) and protein (B) expression of the type 1 cytokines IL-12p40, IFNγ, TNFα, CCL2, CCL3 and CCL4 and increased CXCL1 protein expression (Figure 1B). *P

Hyperoxic Exposure of Immature Mice Increases the Inflammatory Response to Subsequent Rhinovirus Infection: Association with Danger Signals.

Infants with a history of prematurity and bronchopulmonary dysplasia have a high risk of asthma and viral-induced exacerbations later in life. We hypo...
2MB Sizes 0 Downloads 8 Views