Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Mar 1.
Published in final edited form as: Birth Defects Res A Clin Mol Teratol. 2014 Feb 27;100(3):189–201. doi: 10.1002/bdra.23220

POSTNATAL INFLAMMATION IN THE PATHOGENESIS OF BRONCHOPULMONARY DYSPLASIA

Vineet Bhandari 1
PMCID: PMC4023567  NIHMSID: NIHMS572008  PMID: 24578018

Abstract

Exposure to hyperoxia, invasive mechanical ventilation and systemic/local sepsis are important antecedents of postnatal inflammation in the pathogenesis of bronchopulmonary dysplasia (BPD). This review will summarize information obtained from animal (baboon, lamb/sheep, rat and mouse) models that pertain to the specific inflammatory agents and signaling molecules that predispose a premature infant to BPD.

Keywords: invasive ventilation, infection, hyperoxia, lung, newborn

Bronchopulmonary Dysplasia (BPD)

BPD is the most common chronic respiratory disease in infants and is a devastating condition that disrupts the developmental program of the lung secondary to preterm birth. BPD occurs as a result of interactions between genetic and environmental factors (invasive mechanical ventilation, sepsis, and hyperoxia) (Bhandari and Bhandari, 2009; Bhandari and Bhandari, 2011). Although the definition of BPD has evolved over the past decade, it is currently defined as the need for O2 supplementation for 28 days of life and a “physiologic” assessment of the O2 requirement at 36 weeks postmenstrual age (Bhandari and Bhandari, 2011; Trembath and Laughon, 2012). It is estimated that 10,000–15,000 new cases of BPD occur each year in the United States, and significantly, 97% of all BPD cases occur in infants with a birth weight <1250 grams (Bhandari and Bhandari, 2011). Despite many advances in neonatal ventilation techniques, widespread use of surfactant and antenatal corticosteroids, as well as aggressive fluid management, the incidence of BPD has remained the same (Smith et al., 2005) or even increased slightly (Bhandari and Bhandari, 2011; Trembath and Laughon, 2012). BPD is associated with significant pulmonary and neurodevelopmental sequelae that continue to have health ramifications into adulthood (Anderson and Doyle, 2006; Bhandari and Panitch, 2006; Bhandari and Bhandari, 2011).

Stages of Lung Development

Five distinct stages of lung development have been recognized: embryonic, pseudoglandular, canalicular, saccular and alveolar. These stages tend to be conserved across species but differences do exist with respect to the duration of each stage and its temporal relationship to gestational age (Table 1) (Thurlbeck, 1975; Kotecha, 2000; Hislop, 2002; Maeda et al., 2007; Schittny and Burri, 2008; Manisalco and Bhandari, 2010; Meller and Bhandari, 2012). In the present context, the human preterm babies most predisposed to develop BPD tend to be born with their lungs in the late canalicular (characterized by formation of primitive alveoli, differentiation of Type I and Type II pneumocytes and formation of alveolar capillary barrier) and early saccular (initiation of surfactant production, enlargement of terminal airways, pulmonary vascularization) stages (Kotecha, 2000; Joshi and Kotecha, 2007; Kramer et al., 2007; Maeda et al., 2007). It is important to mention that for neonatal rodents, the saccular stage begins in the uterus and continues through post natal day 5 (PN5), while in sheep/baboons /humans, this occurs in utero (Kramer et al., 2007; Maeda et al., 2007). The distinction between the timing/ex or in utero location of developmental stages of the lung in animal models versus humans assumes critical importance when designing/assessing experimental BPD models.

Table 1.

Stages of lung development in various animal models and humans.

Developmental
Stage of Lung
Sheep
(days)
Rat
(days)
Mouse
(days)
Baboon
(days)
Human
(weeks)
Embryonic E17 – 30 E11 – 13 E9 – 11.5 <E42 3 – 7
Pseudoglandular E30 – 85 E13 – 18.5 E11.5 – 16.5 E57 – 80 5 – 17
Canalicular E85 – 110 E18.5 – 20 E16.5 – 17.5 E80 – 140 16 – 26
Saccular E110 – 120 E20 – PN4 E17.5 – PN5 E140 – 168 24 – 38
Alveolar E120 – 145 PN4 – 28 PN5 – 28 E168 – 185 32 weeks - PN 3 years*

E: embryonic; PN: postnatal.

*

Some data suggest that this occurs up to 8 years of age.

Postnatal factors and Inflammation in the Pathogenesis of BPD

Inflammation is a cornerstone in the pathogenesis of BPD. While a variety of clinical factors have been associated with BPD (Akram Khan et al., 2006), the critical ones that have been linked to inflammation-induced cell signaling pathways include antenatal and postnatal factors (Bhandari and Bhandari, 2009; Bhandari and Bhandari, 2011). In the context of a genetic predisposition in an immature lung, a combination of pre- and post-natal factors initiate an inflammatory process that is mediated by a variety of molecular mediators, including cytokines. Damage to the developing lung by the activation of the cell death pathways is followed by resolution of injury to close to normal lung architecture, or repair. The latter state is characterized by the pulmonary phenotype of “new” BPD as evidenced by fewer and larger simplified alveoli, along with dysmorphic vasculature, leading to the description of impaired alveolarization and dysregulated vascularization (Bhandari and Bhandari, 2009; Bhandari, 2010; Bhandari and Bhandari, 2011).

The contributory role of the major prenatal factor – chorioamnionitis – has been covered by recent reviews (Gien and Kinsella, 2011; Viscardi, 2012; Thomas and Speer, 2013) and elsewhere in this issue. Among the postnatal factors, the 3 most important ones: invasive mechanical ventilation, postnatal local/systemic sepsis and hyperoxia, will be discussed in this review.

Invasive Ventilation: Animal Models

Early studies using a chronically-ventilated (3–4 weeks) preterm lamb model of BPD showed evidence of non-uniform inflation patterns and impaired alveolar formation with an abnormal abundance of elastin (ELN) (Albertine et al., 1999). Inflammation was evident by the presence of inflammatory cells, namely alveolar macrophages, neutrophils, and mononuclear cells and edema (Albertine et al., 1999). In this model, there was also reduced lung expression of growth factors that regulate alveolarization and differential alteration of matrix proteins that regulate ELN assembly (Bland et al., 2007). Conversely, a non-invasive (nasal) ventilation approach preserved alveolar architecture (Reyburn et al., 2008) and had a positive effect on parathyroid hormone-related protein-peroxisome proliferator-activated receptor-gamma (PTHrP-PPARγ)-driven alveolar homeostatic epithelial-mesenchymal signaling (Rehan et al., 2011).

More recently, even short-term stretch injury (15 minutes) secondary to invasive ventilation in preterm fetal sheep led to increased levels of proinflammatory cytokines interleukin-1beta (IL-1β), IL-6, monocyte chemoattractant protein (MCP)-1, and MCP-2 mRNA by 1 hour (h) (Hillman et al., 2011). This was accompanied by increased presence of inflammatory cells in the bronchoalveolar lavage fluid (BALF) with initial increases in neutrophils and monocytes by 1h and a transition to macrophages by 24h (Hillman et al., 2011).

The preterm ventilated baboon model of BPD [delivered at 125 days (d) – at 68% of gestation] showed evidence of alveolar hypoplasia and dysmorphic vasculature, akin to that seen in human BPD (Coalson et al., 1999). Importantly, there were significant elevations of tumor necrosis factor-alpha (TNF-α), IL-6, IL-8 levels, but not of IL-1β and IL-10, in tracheal aspirate fluids at various times during the period of ventilatory support, supporting a role for inflammation (Coalson et al., 1999). In addition, increased matrix metalloproteinase-9 (MMP-9) levels were associated with lung inflammation and edema seen in this invasive ventilation model (Tambunting et al., 2005). Alteration of vascular growth factors (vascular endothelial growth factor or VEGF) was also noted in the lungs of various baboon models (Maniscalco et al., 2002; Tambunting et al., 2005). Bombesin is a 14-amino acid peptide, initially detected in amphibian skin, but immunoreactive studies have shown the presence of bombesin-like peptides (BLP) in multiple organ systems in mammals (Ganter and Pittet, 2006). In the lung, BLP have been shown to be released by pulmonary neuroendocrine cells (Ganter and Pittet, 2006). BLP blockade improved alveolar septation and angiogenesis in the preterm baboon models (Sunday et al., 1998; Subramaniam et al., 2007).

In the 125d baboon model, treatment with early nasal continuous positive airway pressure (NCPAP) for 28d led to a pulmonary phenotype similar to 156d gestational control lungs, suggesting that this non-invasive approach could minimize lung injury (Thomson et al., 2004). In the same model, delayed extubation (till 5 days) versus early extubation to NCPAP at 24h, led to significantly increased BALF IL-6, IL-8, MCP-1, macrophage inflammatory protein-1 alpha (MIP-1α), and growth regulated oncogene-alpha (GRO-α) in the delayed NCPAP group (Thomson et al., 2006).

In neonatal rats (7–14d old – in the alveolar phase of lung development), high tidal volume ventilation increased IL-6 mRNA and upregulation of the transforming growth factor-beta (TGF-β) signaling molecule, connective tissue growth factor (CTGF) mRNA and protein expression, compared to controls (Wu et al., 2008). In an 8d old rat ventilation model, high tidal volumes increased the neutrophilic and inflammatory cytokine mRNA and/or protein expression (IL-1β, IL-6, CXCL-1 and 2) response (Kroon et al., 2010). In a 7d rat model, exposure to mechanical ventilation for 24h in room air led to cell cycle arrest (Kroon et al., 2011), suggesting a possible prelude to impaired alveolarization, the hallmark of BPD.

In an invasive ventilation model in 2-week old mice (well into the alveolar phase of lung development) for 1h, IL-6 lung levels were increased in the high tidal volume ventilation group (Cannizzaro et al., 2008). Studies conducted in 2–6d old mice (late saccular to early alveolar phase of lung development) ventilated for 8–24h with room air or 40% O2 revealed dysregulated elastin assembly, a 3–5 fold increase in cell death, TGF-β activation, and a decrease in VEGF-receptor 2 (VEGF-R2) expression (Bland et al., 2008; Mokres et al., 2010). Inhibiting lung elastase activity by using recombinant human elafin or genetically-modified mice that expressed elafin in the vascular endothelium was protective of the lung injury (Hilgendorff et al., 2011; Hilgendorff et al., 2012).

Interestingly, transcriptomic analyses of lung samples from ventilated premature baboons, mouse and rat models of BPD and humans resulted in a final “cross-species” data set which included 90 healthy and 24 ventilated samples of various ages, four species, and both sexes and identified several highly conserved genes in response to mechanical ventilation (Kompass et al., 2010). These included ELN, gastrin-releasing polypeptide (GRP, which is a BLP), and CTGF (Kompass et al., 2010).

To summarize, while the lamb/sheep/baboon ventilation models are in the saccular stage (akin to the human premature babies who are at most risk for BPD at birth), the rat/mouse ventilation models are in the alveolar phase of lung development. However, it is quite obvious that mechanical stretch injury generates an inflammatory response (mostly neutrophils, IL-1β, IL-6, CXCL-1 / -2, TGF-β signaling), along with alterations in matrix proteins (ELN, MMP-9) and vascular growth factors (VEGF). In addition, there is increased cell death and cell cycle arrest. Thus, it appears that an initial inflammatory cascade triggers the signaling of additional molecular mediators that lead to dysregulated vascularization and impaired alveolarization. Interestingly, non-invasive (nasal) ventilation approaches were protective of these responses.

Postnatal Sepsis: Animal Models

In a preterm lamb model treated with CPAP or mechanical ventilation up to 3h, mRNA expression of inflammatory cytokines was increased, compared to unventilated controls (Polglase et al., 2009). Lipopolysaccharide (LPS) administration increased inflammatory cells and enhanced the responses of IL-1β, IL-6 and IL-8 mRNA expression; however, cytokine markers of lung injury from intratracheal LPS were not decreased in the CPAP versus the ventilated group (Polglase et al., 2009).

In a systemic Gram-negative sepsis (Salmonella endotoxin) neonatal (10d) rat model, there was increased pulmonary neutrophil recruitment at 1h and expression of cytokine-induced neutrophil chemoattractant (CINC) at 2h, but with no overt lung injury (up to 16h) (Tillema et al., 2000). However, in a 6d neonatal rat model intratracheally exposed to LPS, there was increased neutrophil accumulation in the lungs at 24h, which changed to mostly macrophages at 15d (Franco et al., 2002). The latter time-point (postnatal day 21 or PN21) was also associated with enlarged peripheral air spaces and fewer alveoli and increased 92-kDa gelatinase activity, compared to controls. Interestingly, doxycycline, a non-specific MMP-inhibitor, worsened LPS-induced alveolarization abnormalities despite partly inhibiting the gelatinase overactivity (Franco et al., 2002).

Neonatal mice injected with intraperitoneal LPS demonstrated reduced lung inflammation and apoptosis after 24h as compared to adults, and this was associated with activation of the transcription factor nuclear factor-kappa B (NF-κB) (Alvira et al., 2007). Inhibition of NF-κB resulted in increased cell death and alveolar simplification and disruption of angiogenesis via VEGF-R2 (Iosef et al., 2012). Independent work by others using a targeted deletion of NF-κB signaling (using a lung epithelium-specific deletion of IKKβ – which is a known activating kinase upstream of NF-κB) in a mouse model revealed alveolar hypoplasia with decreased VEGF expression (Londhe et al., 2011).

Viral infections of epithelial cells are characterized by the generation of the pro-inflammatory molecule double-stranded RNA (dsRNA) during intracellular replication of viruses, and has been shown to induce activation of the neutrophil chemoattractant, IL-8. Hence, there is a possibility that inflammatory processes secondary to viral infections could also lead to a BPD pulmonary phenotype. This was tested by intratracheal delivery of dsRNA into a 10d-old mouse model, which resulted in neutrophil accumulation, increased lung injury and decreased alveolarization (Londhe et al., 2005). In addition, there was increased expression of CXCL-1, as well as its receptor CXCR2. Pre-treatment with CXCR2-neutralizing antibody was able to reverse the effects in the developing lung (Londhe et al., 2005).

Taken together, it would suggest that both bacterial and viral agents in the PN developing lung stimulate a strong inflammatory response, which leads to altered alveolar architecture, suggestive of the BPD pulmonary phenotype. Interestingly, infection-initiated inflammatory response in the developing lung appears not to be dampened by using a non-invasive ventilation approach.

Hyperoxia Exposure: Animal Models

Hyperoxia exposure to preterm rabbits led to impaired alveolarization (Mascaretti et al., 2009). In neonatal lambs, cimetidine (Hazinski et al., 1989) as well inhaled nitric oxide (iNO) (Cotton et al., 2006) have been reported to reduce hyperoxia-induced lung injury.

In hyperoxia-exposed neonatal rats, a variety of agents have been used as therapeutic agents to ameliorate hyperoxia-induced lung inflammation/injury and/or the BPD phenotype. These include keratinocyte growth factor (KGF) (Frank, 2003; Franco-Montoya et al., 2009), VEGF (Kunig et al., 2005; Thebaud et al., 2005; Kunig et al., 2006), iNO(ter Horst et al., 2007), inhaled ethyl nitrite (Auten et al., 2007), sildenafil (Ladha et al., 2005; Park et al., 2013), L-citrulline (Vadivel et al., 2010), leukotriene inhibition (Funk et al., 2007), azithromycin (Ballard et al., 2007), apelin – a potent vasodilator and angiogenic factor (Visser et al., 2010), phsophodiestrase-4 (PDE-4) inhibition (de Visser et al., 2008; de Visser et al., 2012), curcumin(Sakurai et al., 2011), colchicine (Ozdemir et al., 2012), pentoxifylline (Almario et al., 2012), Nigella sativa oil (Tayman et al., 2013), calcitonin-gene related peptide (Dang et al., 2012), caffeine (Weichelt et al., 2013), inhibition of Wnt-signaling (Alapati et al., 2013; Hummler et al., 2013), and administration of cytidine 5’-diphosphosphocholine (Cetinkaya et al., 2013). A variety of stem cells delivered to hyperoxia-exposed neonatal rat lungs have improved alveolar and vascular growth (van Haaften et al., 2009; Zhang et al., 2012; Baker et al., 2013; Pierro et al., 2013).

Prolonged exposure to hyperoxia to the neonatal mouse, for 14d or longer (Warner et al., 1998; Woyda et al., 2009; Velten et al., 2010; Zhang et al., 2011; Zhang et al., 2012; Tibboel et al., 2013), does result in lung injury, but the phenotype is more reminiscent of “old” rather than “new” BPD (Bhandari and Bhandari, 2011) – the major differentiating feature being the increased fibrosis in the former condition.

To replicate human BPD, exposure to hyperoxia during the critical saccular stage of lung development is sufficient to create the pulmonary phenotype, with the effects being not only dose-dependent on the fraction of inspired oxygen (FiO2) concentration, but lasting life-long and mimicking the increased susceptibility to respiratory tract infections (O'Reilly et al., 2008; Yee et al., 2009; Li et al., 2011 Dec. 15 ; Buczynski et al., 2012; Choo-Wing et al., 2013), which has also been well-documented in former premature human infants with BPD (Bhandari and Bhandari, 2003).

The specific role of individual inflammatory molecular mediators in the pathogenesis of BPD has been particularly well-illustrated by utilizing lung-targeted overexpressing transgenic models, in room air, resulting in pulmonary phenotypes reminiscent of human BPD. These include IL-1β (Bry et al., 2007; Backstrom et al., 2011), and interferon-gamma (IFN-γ) (Harijith et al., 2011; Choo-Wing et al., 2013). In the case of IL-1β transgenic mice, absence of the beta6 integrin subunit was protective of the BPD phenotype (Hogmalm et al., 2010). Interestingly, inhibition of cyclooxygenase-2 (Cox-2) ameliorated the BPD phenotype in the hyperoxia-induced as well as the IFN-γ lung overexpressing transgenic mouse model in room air. A recent paper has reported that increased Cox-2 activity may contribute to proinflammatory responses in hyperoxia-exposed developing mouse lungs (Britt et al., 2013).

Increased TGF-β signaling has been implicated in resulting in the BPD phenotype (Alejandre-Alcazar et al., 2007), with neutralizing antibodies against it improving hyperoxia-induced lung injury (Nakanishi et al., 2007). This has been corroborated by TGF-β1 overexpression models (Gauldie et al., 2003; Vicencio et al., 2004; Li et al., 2011 Dec. 15). Another molecule that has been implicated as a downstream mediator of TGF-β1 signaling in the newborn lung is CTGF (Li et al., 2011 Dec. 15). Targeted overexpression of CTGF in the developing mouse lung has been reported to have a BPD phenotype (Wu et al., 2010; Chen et al., 2011), with CTGF antibody treatment attenuating hyperoxia-induced lung injury in neonatal rats (Alapati et al., 2011). Periostin, a secreted extracellular matrix protein involved in TGF-β signaling, also appears to be playing a role, as periostin null mutant mice were protected from some features of hyperoxia-induced lung injury (Bozyk et al., 2012).

The importance of just the “right amount” of a molecular mediator (the “Goldilocks effect”) has also been made obvious with such a genetic approach in the case of macrophage migration inhibition factor (MIF) (Sun et al., 2013; Sun et al., 2013).

Besides inflammation, hyperoxia exposure has been well-recognized to cause cell death (McGrath-Morrow and Stahl, 2001; Bhandari, 2010) in mouse lungs, and not surprisingly, BPD models (hyperoxia-exposed or transgenic in room air) do reflect this (Choo-Wing et al., 2007; Harijith et al., 2011; Choo-Wing et al., 2013). Activation of the prosurvival factor, Akt, was protective of hyperoxia-induced lung injury and BPD phenotype in neonatal rats (Alphonse et al., 2011). Another important feature is cell cycle arrest and upregulation of the cyclin-dependent kinase inhibitor, p21 (Londhe et al., 2011). p21 null mutant neonatal mice exposed to hyperoxia had decreased survival and larger alveoli (McGrath-Morrow et al., 2004). Among matrix proteins, cathepsin-S deficiency (Hirakawa et al., 2007) and MMP-9 lack and/or deficiency (Chetty et al., 2008; Harijith et al., 2011) was protective of the BPD phenotype in mice models. Regarding anti-oxidants, the transcription factor Nrf2, appeared to have a role as lack of Nrf2 enhanced hyperoxia-induced lung injury, and was associated with increased IL-6 expression (McGrath-Morrow et al., 2009). While a blockade of neutrophil influx in hyperoxia-exposed rat lungs has been shown to be protective of the BPD phenotype (Auten et al., 2001), this effect was enhanced by combining with overexpression of extracellular superoxide dismutase in mice (Min et al., 2012).

Recently, newborn sphingosine kinase 1-deficient mice were reported to be protected from hyperoxia-induced lung injury, including decreased IL-6 BALF concentrations (Harijith et al., 2013). Another report noted that neonatal mice with dysfunctional aryl hydrocarbon receptor were more susceptible to hyperoxia-induced alveolar simplification and inflammation (Shivanna et al., 2013).

A variety of potential therapeutic agents have been used in hyperoxia-exposed mice models that have been shown to decrease inflammation and/or attenuate other parameters of lung injury/BPD phenotype. These include rosiglitazone (Dasgupta et al., 2009; Takeda et al., 2009), hepatocyte growth factor (HGF) (Ohki et al., 2009), B-naphthoflavone (Couroucli et al., 2011), arginyl-glutamine as well as docosahexaenoic acid (Ma et al., 2012), and a combination of vitamin A and retinoic acid (James et al., 2013). Treatment with human amnion epithelial cells attenuated some parameters of hyperoxia-induced inflammatory lung injury (mRNA expression of IL-1α, IL-6, TGF-β, platelet-derived growth factor-beta or PDGF-β, mean linear intercept, and septal crest density), but not other aspects, for example, alveolar airspace volume, collagen content or leukocyte infiltration in neonatal mice (Vosdoganes et al., 2013).

To summarize, while variable initiation and duration of exposure to hyperoxia animal models have been reported as models of human BPD, exposure to hyperoxia for a relatively-short (PN1-4) duration in mice, which is at the critical saccular stage of lung development, can result in an inflammatory response sufficient to create the BPD pulmonary phenotype. This can be recapitulated using transgenic mice models of the inflammatory mediators, but kept in room air. Importantly, exposure to 0.4, 0.6, >0.8 FiO2 can mimic mild, moderate and severe BPD, respectively. A vast array of therapeutic agents has been reported to be effective in improving alveolar and/or vascular architecture of the hyperoxia-exposed neonatal lung in lambs, rats and mice. While hyperoxia-exposure is a good starting point for testing the efficacy of potential therapeutic agents, it is important to be able to delineate the responsible molecule/signaling pathway in developmentally-appropriate room air models and confirm the results in preventing/ameliorating the BPD phenotype. This would avoid the confounding variable of hyperoxia-induced alterations in multiple other molecular mediators, allowing delineation of targeted molecules in specific signaling pathways for maximal potential therapeutic relevance. Among the inflammatory mediators of hyperoxia-induced lung injury that can mimic the BPD phenotype in room air, the well-defined ones are IL-1β, TGF-β1, CTGF, IFN-γ, and MIF. In addition, there has been limited translation of therapeutic targeting of hyperoxia-exposure models to premature human neonates to prevent/improve BPD. In some cases, where this has been done, the results have been quite disappointing, for example, with iNO(Cole et al., 2011). Hence, it would be important to attempt to translate some of the newer targets in specific signaling pathways that have been recently reported, for example, inhibition of Cox-2 (Britt et al., 2013; Choo-Wing et al., 2013) as a potential therapeutic option for prevention/amelioration of BPD.

Combination Models

Short-term (6h) invasive ventilation in preterm lambs increased mRNA expression of the acute phase reactant serum amyloid A (which is known to be induced upon exposure to pro-inflammatory cytokines), which was further enhanced by concomitant exposure to intratracheal endotoxin (Wilson et al., 2005). In another short-term (4h) study, invasive ventilation combined with a recombinant Clara Cell secretory protein (CC10) decreased systemic and/or lung inflammation markers such as TNF-α, IL-6 and IL-8 (Shashikant et al., 2005).

There was increased TNF-α production from macrophages obtained from the BALF of neonatal rats exposed to hyperoxia and LPS in vitro, compared to room air animals and LPS-exposed cells (Lindsay et al., 2000).

In an interesting study design, neonatal rats (PN6-7) were administered LPS 24h prior, and then ventilated for 8h, with assessments being made soon after or delayed for 48h (Trummer-Menzi et al., 2012). Results revealed the inflammatory reaction was significant with mechanical ventilation plus O2 exposure (FiO2: 0.6) leading to increased pro-inflammatory gene expression for MIP-2 (CXCL2, the rodent homolog for IL-8) and a trend towards up-regulation of IL-1β, IL-6, and TNF-α, mostly at the earlier time point. Lung histology was abnormal at both time points, but MMP-9 expression was significantly increased at the 48h time-point (Trummer-Menzi et al., 2012). In 8d old rats being ventilated, the inflammatory response was modified by pre-exposure to systemic LPS combined with low tidal volume ventilation, but enhanced by exposure to 50% O2(Kroon et al., 2010).

Intra-amniotic exposure to 2 different doses of LPS at embryonic day (E)20, followed by hyperoxia-exposure (FiO2: 0.85) at birth for 1 or 2 weeks in neonatal rats, led to amplification of lung injury at 1 week (Choi et al., 2009). However, the authors noted that 2-weeks of hyperoxia caused extensive lung injury, perhaps, masking the effect of LPS (Choi et al., 2009). Similarly, enhancement of lung injury has been reported by other investigators, by postnatal hyperoxia exposure, with intra-amniotic priming to LPS (Kim et al., 2010; Lee et al., 2010; Velten et al., 2010), with attenuation of these effects by antenatal betamethasone (Yoo et al., 2013).

Another animal model combined antenatal hypoxia-induced growth restriction (FiO2 0.1 from E14 to 18) with PN hyperoxia (FiO2 0.7 for 2 weeks) to generate a double-hit mouse model of BPD, which was associated with increased expression of IL-1β (Gortner et al., 2013).

In a recent report, hyperoxia was noted to differentially contribute to macrophage polarization by enhancing LPS-induced M1 and inhibiting the IL-4-induced M2 phenotype (Syed and Bhandari, 2013). Breast regression protein -39 (BRP-39) absence led to further enhancement of the hyperoxia- and LPS-induced M1 phenotype. In addition, neonatal BRP-39 null mutant mice were significantly more sensitive to LPS plus hyperoxia-induced lung injury and mortality, compared to wild-type mice. This was evident by abnormal histology, abnormal lung morphometry, increased BALF total cell counts and increased BALF protein concentrations, compared to controls. IL-6 and IL-1β were significantly increased in neonatal BRP-39 null mutant mouse lungs exposed to LPS plus hyperoxia, compared to controls (Syed and Bhandari, 2013).

Overall, experimental data from combination models suggests that exposure to endotoxin antenatally or after birth enhances the inflammatory response to PN invasive ventilation and/or hyperoxia. In a neonatal mouse model, lack of BRP-39 appears to be important for this response, possibly via alteration of the macrophage polarization status (Syed and Bhandari, 2013).

Relevant Human Studies

A large variety of biomarkers have been detected and associated with the development of BPD in tracheal aspirates, as well as blood and urine samples of premature infants (Bhandari and Bhandari, 2009; Bhandari and Bhandari, 2013). The ones that have been implicated in animal models (discussed above) include inflammatory cytokines (IL-1β, IL-8, IL-10, IFN-γ, MCP-1, MIF, TGF-β, TNF-α), matrix proteins (MMP-9), growth (BLP, HGF, KGF, PTHrP) and vascular factors (VEGF, VEGF-R2). Some inflammatory markers or their downstream targets have been shown to be increased in BPD lung specimens. These include endoglin, CTGF and periostin (TGF-β signaling) (De Paepe et al., 2008; Alapati et al., 2011; Bozyk et al., 2012), IP-9 (CXCL11), Cox-2 and C/EBP homologous protein (CHOP) (IFN-γ signaling) (Harijith et al., 2011; Choo-Wing et al., 2013). VEGF immunostaining was reported to be either decreased (Bhatt et al., 2001) or increased (Lassus et al., 2001), probably reflecting the temporal sequence of VEGF release/production in the neonatal human lung (Meller and Bhandari, 2012). BLP – which is elevated in BPD – has been shown to increase mast cells in the lung (Subramaniam et al., 2003). This may have a correlation with a recent publication of increased mast cells in human lungs with BPD (Bhattacharya et al., 2012). Lower tracheal aspirate concentrations of YKL-40, the human homolog of BRP-39, have been reported with development of BPD in premature neonates (Sohn et al., 2010). Others, such as Angiopoietin 2, have been implicated in human studies (Bhandari et al., 2006; Aghai et al., 2008), but developmentally-appropriate animal modeling data is lacking, to date. Table 2 lists selective mediators that have a role in the pathogenesis of BPD in animal models that have been associated with preterm human lungs (tracheal aspirates and/or tissue) with BPD.

Table 2.

Selective mediators involved in the pathogenesis of BPD in animal models and associated with human preterm lungs with BPD.

Mediators Major effects in relation to
BPD
References
Bombesin-like peptide Increased levels associated with BPD (Sunday et al., 1998; Subramaniam et al., 2007)
BRP-39 Decreased expression associated with BPD (Sohn et al., 2010; Syed and Bhandari, 2013)
CTGF Increased expression associated with BPD (Wu et al., 2008; Wu et al., 2010; Alapati et al., 2011; Chen et al., 2011)
Interferon gamma Increased levels leads to the BPD pulmonary phenotype (Harijith et al., 2011; Aghai et al., 2012; Choo-Wing et al., 2013)
IL-1β Increased expression leads to the BPD pulmonary phenotype (Bry et al., 2007; Backstrom et al., 2011; Bhandari and Bhandari, 2013)
IL-6 Increased expression associated with BPD (Choo-Wing et al., 2007; Bhandari and Bhandari, 2013)
Keratinocyte growth factor Lower levels associated with BPD (Frank, 2003; Franco-Montoya et al., 2009; Bhandari and Bhandari, 2013)
MIF Lower levels associated with BPD (Kevill et al., 2008; Sun et al., 2013)
MMP-9 Increased levels associated with BPD (Tambunting et al., 2005; Chetty et al., 2008; Harijith et al., 2011)
Nitric oxide Inhaled nitric oxide leads to improvement in pulmonary phenotype of BPD* (ter Horst et al., 2007; Cole et al., 2011)
Nuclear factor kappa-B Disruption of signaling pathway associated with BPD (Alvira et al., 2007; Londhe et al., 2011; Iosef et al., 2012; Bhandari and Bhandari, 2013)
Perisostin Increased expression associated with BPD (Bozyk et al., 2012)
PTHrP Disruption of signaling pathway associated with BPD (Rehan and Torday, 2006; Rehan et al., 2011; Bhandari and Bhandari, 2013)
TGF-β Increased levels leads to the BPD pulmonary phenotype (Gauldie et al., 2003; Vicencio et al., 2004; Alejandre-Alcazar et al., 2007; Nakanishi et al., 2007; Li et al., 2011; Bhandari and Bhandari, 2013)
VEGF Altered ligand and VEGF receptor 2 levels associated with BPD (Maniscalco et al., 2002; Kunig et al., 2005; Tambunting et al., 2005; Thebaud et al., 2005; Kunig et al., 2006; Bland et al., 2008; Mokres et al., 2010; Meller and Bhandari, 2012; Bhandari and Bhandari, 2013)

BRP: breast regression protein; CTGF: connective tissue growth factor; IL: interleukin; MIF: macrophage migration inhibitory factor; MMP: matrix metallo proteinase; PTHrP: parathyroid hormone related protein; TGF: transforming growth factor; VEGF: vascular endothelial growth factor.

*

Not shown consistently in human trials.

Summary and Conclusions

While the significantly different responses between adult and neonatal lungs to the postnatal factors discussed here – invasive ventilation (Naik et al., 2001; Ikegami et al., 2003; Copland et al., 2004; Kornecki et al., 2005), local/systemic sepsis (Martin et al., 1995; Lee et al., 2000; Lee et al., 2001; Franco et al., 2002; Alvira et al., 2007), and hyperoxia (Bhandari, 2002; Bhandari and Elias, 2006; Choo-Wing et al., 2007; Bhandari et al., 2008)– are well established, it is also important to be cognizant of the stages of lung development, when comparing animal data for relevance to humans. After all, it is the preterm lung in the late canalicular/ saccular phase of development that is most predisposed to BPD, on exposure to the postnatal factors. This is best exemplified by studies that highlight the differential responses in the various stages of lung development (mostly, saccular vs. alveolar) in animal models (Backstrom et al., 2011).

Figure 1 shows a schema of the relative contribution of 3 major postnatal factors in the inflammation-induced effects that are involved in the pathogenesis of BPD. The animal data would suggest that avoiding postnatal local/systemic infection is more important than avoiding invasive mechanical ventilation to decrease the inflammatory response in developing lungs. In other words, one could speculate that controlling postnatal lung/systemic infection (and thus, inflammation) would be likely to result in a more dramatic decrease in BPD. Once infection rates are relatively low, non-invasive ventilation approaches would provide an additional boost to decrease BPD. Hyperoxia exposure (>40% O2) appears to exacerbate both infection and invasive ventilation-induced inflammatory responses. Thus, among the environmental factors, modifying the O2 concentration and/or duration of exposure, in the context of a non-invasive ventilation approach, in conjunction with effective control of PN sepsis would offer the ideal combination to decrease inflammation-induced lung injury and BPD.

Figure 1. Postnatal inflammation in the pathogenesis of bronchopulmonary dysplasia (BPD).

Figure 1

The 3 major postnatal factors act on the immature lung initiating an inflammatory cascade involving specific cells and cytokines. Local/systemic sepsis is most prominent among the 3 to cause inflammation, followed by hyperoxia and invasive ventilation (as noted by the thickness of the arrows). These, in turn, instigate effects promoting cell death, cell cycle arrest, and abnormal production of growth factors, matrix proteins and vascular factors. Mediators that have been consistently reported to have the above noted effects have been listed in the figure. The net result is impaired alveolarization and dysregulated vascularization culminating in the pulmonary phenotype of BPD. Ang-2: Angiopoietin 2. For additional abbreviations and details, please see text.

Acknowledgments

Funded by NIH grant HL 085103

References

  1. Aghai ZH, Faqiri S, Saslow JG, et al. Angiopoietin 2 concentrations in infants developing bronchopulmonary dysplasia: attenuation by dexamethasone. J Perinatol. 2008;28:149–155. doi: 10.1038/sj.jp.7211886. [DOI] [PubMed] [Google Scholar]
  2. Aghai ZH, Saslow JG, Mody K, et al. IFN-gamma and IP-10 in tracheal aspirates from premature infants: Relationship with bronchopulmonary dysplasia. Pediatr Pulmonol. 2012 doi: 10.1002/ppul.22540. [DOI] [PubMed] [Google Scholar]
  3. Akram Khan M, Kuzma-O'Reilly B, Brodsky NL, et al. Site-specific characteristics of infants developing bronchopulmonary dysplasia. J Perinatol. 2006;26:428–435. doi: 10.1038/sj.jp.7211538. [DOI] [PubMed] [Google Scholar]
  4. Alapati D, Rong M, Chen S, et al. Connective tissue growth factor antibody therapy attenuates hyperoxia-induced lung injury in neonatal rats. Am J Respir Cell Mol Biol. 2011;45:1169–1177. doi: 10.1165/rcmb.2011-0023OC. [DOI] [PubMed] [Google Scholar]
  5. Alapati D, Rong M, Chen S, et al. Inhibition of LRP5/6-mediated Wnt/beta-catenin signaling by Mesd attenuates hyperoxia-induced pulmonary hypertension in neonatal rats. Pediatr Res. 2013;73:719–725. doi: 10.1038/pr.2013.42. [DOI] [PubMed] [Google Scholar]
  6. Albertine KH, Jones GP, Starcher BC, et al. Chronic lung injury in preterm lambs. Disordered respiratory tract development. Am J Respir Crit Care Med. 1999;159:945–958. doi: 10.1164/ajrccm.159.3.9804027. [DOI] [PubMed] [Google Scholar]
  7. Alejandre-Alcazar MA, Kwapiszewska G, Reiss I, et al. Hyperoxia modulates TGF-beta/BMP signaling in a mouse model of bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol. 2007;292:L537–L549. doi: 10.1152/ajplung.00050.2006. [DOI] [PubMed] [Google Scholar]
  8. Almario B, Wu S, Peng J, et al. Pentoxifylline and prevention of hyperoxia-induced lung -injury in neonatal rats. Pediatr Res. 2012;71:583–589. doi: 10.1038/pr.2012.14. [DOI] [PubMed] [Google Scholar]
  9. Alphonse RS, Vadivel A, Coltan L, et al. Activation of Akt protects alveoli from neonatal oxygen-induced lung injury. Am J Respir Cell Mol Biol. 2011;44:146–154. doi: 10.1165/rcmb.2009-0182OC. [DOI] [PubMed] [Google Scholar]
  10. Alvira CM, Abate A, Yang G, et al. Nuclear factor-kappaB activation in neonatal mouse lung protects against lipopolysaccharide-induced inflammation. Am J Respir Crit Care Med. 2007;175:805–815. doi: 10.1164/rccm.200608-1162OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Anderson PJ, Doyle LW. Neurodevelopmental outcome of bronchopulmonary dysplasia. Semin Perinatol. 2006;30:227–232. doi: 10.1053/j.semperi.2006.05.010. [DOI] [PubMed] [Google Scholar]
  12. Auten RL, Jr, Mason SN, Tanaka DT, et al. Anti-neutrophil chemokine preserves alveolar development in hyperoxia-exposed newborn rats. Am J Physiol Lung Cell Mol Physiol. 2001;281:L336–L344. doi: 10.1152/ajplung.2001.281.2.L336. [DOI] [PubMed] [Google Scholar]
  13. Auten RL, Mason SN, Whorton MH, et al. Inhaled ethyl nitrite prevents hyperoxia-impaired postnatal alveolar development in newborn rats. Am J Respir Crit Care Med. 2007;176:291–299. doi: 10.1164/rccm.200605-662OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Backstrom E, Hogmalm A, Lappalainen U, et al. Developmental stage is a major determinant of lung injury in a murine model of bronchopulmonary dysplasia. Pediatr Res. 2011;69:312–318. doi: 10.1203/PDR.0b013e31820bcb2a. [DOI] [PubMed] [Google Scholar]
  15. Baker CD, Seedorf GJ, Wisniewski BL, et al. Endothelial colony-forming cell conditioned media promote angiogenesis in vitro and prevent pulmonary hypertension in experimental bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol. 2013;305:L73–L81. doi: 10.1152/ajplung.00400.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Ballard HO, Bernard P, Qualls J, et al. Azithromycin protects against hyperoxic lung injury in neonatal rats. J Investig Med. 2007;55:299–305. doi: 10.2310/6650.2007.00011. [DOI] [PubMed] [Google Scholar]
  17. Bhandari A and Bhandari V. Pathogenesis, pathology and pathophysiology of pulmonary sequelae of bronchopulmonary dysplasia in premature infants. Front Biosci. 2003;8:e370–e380. doi: 10.2741/1060. [DOI] [PubMed] [Google Scholar]
  18. Bhandari A, Bhandari V. Pitfalls, problems, and progress in bronchopulmonary dysplasia. Pediatrics. 2009;123:1562–1573. doi: 10.1542/peds.2008-1962. [DOI] [PubMed] [Google Scholar]
  19. Bhandari A, Bhandari V. “New” bronchopulmonary dysplasia - A clinical review. Clin Pulm Med. 2011;18:137–143. [Google Scholar]
  20. Bhandari A, Bhandari V. Biomarkers in bronchopulmonary dysplasia. Paediatr Respir Rev. 2013;14:173–179. doi: 10.1016/j.prrv.2013.02.008. [DOI] [PubMed] [Google Scholar]
  21. Bhandari A, Panitch HB. Pulmonary outcomes in bronchopulmonary dysplasia. Semin Perinatol. 2006;30:219–226. doi: 10.1053/j.semperi.2006.05.009. [DOI] [PubMed] [Google Scholar]
  22. Bhandari V. Developmental differences in the role of interleukins in hyperoxic lung injury in animal models. Front Biosci. 2002;7:d1624–d1633. doi: 10.2741/A866. [DOI] [PubMed] [Google Scholar]
  23. Bhandari V. Hyperoxia-derived lung damage in preterm infants. Semin Fetal Neonatal Med. 2010;15:223–229. doi: 10.1016/j.siny.2010.03.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Bhandari V, Choo-Wing R, Lee CG, et al. Developmental regulation of NO-mediated VEGF-induced effects in the lung. Am J Respir Cell Mol Biol. 2008;39:420–430. doi: 10.1165/rcmb.2007-0024OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Bhandari V, Choo-Wing R, Lee CG, et al. Hyperoxia causes angiopoietin 2-mediated acute lung injury and necrotic cell death. Nat Med. 2006;12:1286–1293. doi: 10.1038/nm1494. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Bhandari V, Elias JA. Cytokines in tolerance to hyperoxia-induced injury in the developing and adult lung. Free Radic Biol Med. 2006;41:4–18. doi: 10.1016/j.freeradbiomed.2006.01.027. [DOI] [PubMed] [Google Scholar]
  27. Bhatt AJ, Pryhuber GS, Huyck H, et al. Disrupted pulmonary vasculature and decreased vascular endothelial growth factor, Flt-1, and TIE-2 in human infants dying with bronchopulmonary dysplasia. Am J Respir Crit Care Med. 2001;164:1971–1980. doi: 10.1164/ajrccm.164.10.2101140. [DOI] [PubMed] [Google Scholar]
  28. Bhattacharya S, Go D, Krenitsky DL, et al. Genome-wide transcriptional profiling reveals connective tissue mast cell accumulation in bronchopulmonary dysplasia. Am J Respir Crit Care Med. 2012;186:349–358. doi: 10.1164/rccm.201203-0406OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Bland RD, Ertsey R, Mokres LM, et al. Mechanical ventilation uncouples synthesis and assembly of elastin and increases apoptosis in lungs of newborn mice. Prelude to defective alveolar septation during lung development. Am J Physiol Lung Cell Mol Physiol. 2008;294:L3–L14. doi: 10.1152/ajplung.00362.2007. [DOI] [PubMed] [Google Scholar]
  30. Bland RD, Xu L, Ertsey R, et al. Dysregulation of pulmonary elastin synthesis and assembly in preterm lambs with chronic lung disease. Am J Physiol Lung Cell Mol Physiol. 2007;292:L1370–L1384. doi: 10.1152/ajplung.00367.2006. [DOI] [PubMed] [Google Scholar]
  31. Bozyk PD, Bentley JK, Popova AP, et al. Neonatal periostin knockout mice are protected from hyperoxia-induced alveolar simplication. PLoS One. 2012;7:e31336. doi: 10.1371/journal.pone.0031336. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Britt RD, Jr, Velten M, Tipple TE, et al. Cyclooxygenase-2 in newborn hyperoxic lung injury. Free Radic Biol Med. 2013;61C:502–511. doi: 10.1016/j.freeradbiomed.2013.04.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Bry K, Whitsett JA, Lappalainen U. IL-1beta disrupts postnatal lung morphogenesis in the mouse. Am J Respir Cell Mol Biol. 2007;36:32–42. doi: 10.1165/rcmb.2006-0116OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Buczynski BW, Yee M, Paige Lawrence B, et al. Lung development and the host response to influenza A virus are altered by different doses of neonatal oxygen in mice. Am J Physiol Lung Cell Mol Physiol. 2012;302:L1078–L1087. doi: 10.1152/ajplung.00026.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Cannizzaro V, Zosky GR, Hantos Z, et al. High tidal volume ventilation in infant mice. Respir Physiol Neurobiol. 2008;162:93–99. doi: 10.1016/j.resp.2008.04.010. [DOI] [PubMed] [Google Scholar]
  36. Cetinkaya M, Cansev M, Kafa IM, et al. Cytidine 5'-diphosphocholine ameliorates hyperoxic lung injury in a neonatal rat model. Pediatr Res. 2013;74:26–33. doi: 10.1038/pr.2013.68. [DOI] [PubMed] [Google Scholar]
  37. Chen S, Rong M, Platteau A, et al. CTGF disrupts alveolarization and induces pulmonary hypertension in neonatal mice: implication in the pathogenesis of severe bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol. 2011;300:L330–L340. doi: 10.1152/ajplung.00270.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Chetty A, Cao GJ, Severgnini M, et al. Role of matrix metalloprotease-9 in hyperoxic injury in developing lung. Am J Physiol Lung Cell Mol Physiol. 2008;295:L584–L592. doi: 10.1152/ajplung.00441.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Choi CW, Kim BI, Hong JS, et al. Bronchopulmonary dysplasia in a rat model induced by intra-amniotic inflammation and postnatal hyperoxia: morphometric aspects. Pediatr Res. 2009;65:323–327. doi: 10.1203/PDR.0b013e318193f165. [DOI] [PubMed] [Google Scholar]
  40. Choo-Wing R, Nedrelow JH, Homer RJ, et al. Developmental differences in the responses of IL-6 and IL-13 transgenic mice exposed to hyperoxia. Am J Physiol Lung Cell Mol Physiol. 2007;293:L142–L150. doi: 10.1152/ajplung.00434.2006. [DOI] [PubMed] [Google Scholar]
  41. Choo-Wing R, Syed MA, Harijith A, et al. 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. doi: 10.1165/rcmb.2012-0381OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Coalson JJ, Winter VT, Siler-Khodr T, et al. Neonatal chronic lung disease in extremely immature baboons. Am J Respir Crit Care Med. 1999;160:1333–1346. doi: 10.1164/ajrccm.160.4.9810071. [DOI] [PubMed] [Google Scholar]
  43. Cole FS, Alleyne C, Barks JD, et al. NIH Consensus Development Conference statement: inhaled nitric-oxide therapy for premature infants. Pediatrics. 2011;127:363–369. doi: 10.1542/peds.2010-3507. [DOI] [PubMed] [Google Scholar]
  44. Copland IB, Martinez F, Kavanagh BP, et al. High tidal volume ventilation causes different inflammatory responses in newborn versus adult lung. Am J Respir Crit Care Med. 2004;169:739–748. doi: 10.1164/rccm.200310-1417OC. [DOI] [PubMed] [Google Scholar]
  45. Cotton RB, Sundell HW, Zeldin DC, et al. Inhaled nitric oxide attenuates hyperoxic lung injury in lambs. Pediatr Res. 2006;59:142–146. doi: 10.1203/01.pdr.0000191815.60293.cc. [DOI] [PubMed] [Google Scholar]
  46. Couroucli XI, Liang YH, Jiang W, et al. Prenatal administration of the cytochrome P4501A inducer, Beta-naphthoflavone (BNF), attenuates hyperoxic lung injury in newborn mice: implications for bronchopulmonary dysplasia (BPD) in premature infants. Toxicol Appl Pharmacol. 2011;256:83–94. doi: 10.1016/j.taap.2011.06.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Dang H, Yang L, Wang S, et al. Calcitonin gene-related peptide ameliorates hyperoxia-induced lung injury in neonatal rats. Tohoku J Exp Med. 2012;227:129–138. doi: 10.1620/tjem.227.129. [DOI] [PubMed] [Google Scholar]
  48. Dasgupta C, Sakurai R, Wang Y, et al. Hyperoxia-induced neonatal rat lung injury involves activation of TGF-{beta} and Wnt signaling and is protected by rosiglitazone. Am J Physiol Lung Cell Mol Physiol. 2009;296:L1031–L1041. doi: 10.1152/ajplung.90392.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. De Paepe ME, Patel C, Tsai A, et al. Endoglin (CD105) up-regulation in pulmonary microvasculature of ventilated preterm infants. Am J Respir Crit Care Med. 2008;178:180–187. doi: 10.1164/rccm.200608-1240OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. de Visser YP, Walther FJ, Laghmani EH, et al. Phosphodiesterase-4 inhibition attenuates pulmonary inflammation in neonatal lung injury. Eur Respir J. 2008;31:633–644. doi: 10.1183/09031936.00071307. [DOI] [PubMed] [Google Scholar]
  51. de Visser YP, Walther FJ, Laghmani el H, et al. Phosphodiesterase 4 inhibition attenuates persistent heart and lung injury by neonatal hyperoxia in rats. Am J Physiol Lung Cell Mol Physiol. 2012;302:L56–L67. doi: 10.1152/ajplung.00041.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Franco-Montoya ML, Bourbon JR, Durrmeyer X, et al. Pulmonary effects of keratinocyte growth factor in newborn rats exposed to hyperoxia. Am J Physiol Lung Cell Mol Physiol. 2009 doi: 10.1152/ajplung.00136.2009. [DOI] [PubMed] [Google Scholar]
  53. Franco ML, Waszak P, Banalec G, et al. LPS-induced lung injury in neonatal rats: changes in gelatinase activities and consequences on lung growth. Am J Physiol Lung Cell Mol Physiol. 2002;282:L491–L500. doi: 10.1152/ajplung.00140.2001. [DOI] [PubMed] [Google Scholar]
  54. Frank L. Protective effect of keratinocyte growth factor against lung abnormalities associated with hyperoxia in prematurely born rats. Biol Neonate. 2003;83:263–272. doi: 10.1159/000069480. [DOI] [PubMed] [Google Scholar]
  55. Funk AJ, Mandrell TD, Lokey SJ, et al. Effects of leukotriene inhibition on pulmonary morphology in rat pup lungs exposed to hyperoxia. Comp Med. 2007;57:186–192. [PubMed] [Google Scholar]
  56. Ganter MT, Pittet JF. Bombesin-like peptides: modulators of inflammation in acute lung injury? Am J Respir Crit Care Med. 2006;173:1–2. doi: 10.1164/rccm.2510002. [DOI] [PubMed] [Google Scholar]
  57. Gauldie J, Galt T, Bonniaud P, et al. Transfer of the active form of transforming growth factor-beta 1 gene to newborn rat lung induces changes consistent with bronchopulmonary dysplasia. Am J Pathol. 2003;163:2575–2584. doi: 10.1016/s0002-9440(10)63612-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Gien J, Kinsella JP. Pathogenesis and treatment of bronchopulmonary dysplasia. Curr Opin Pediatr. 2011;23:305–313. doi: 10.1097/MOP.0b013e328346577f. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Gortner L, Monz D, Mildau C, et al. Bronchopulmonary dysplasia in a double-hit mouse model induced by intrauterine hypoxia and postnatal hyperoxia: closer to clinical features? Ann Anat. 2013;195:351–358. doi: 10.1016/j.aanat.2013.02.010. [DOI] [PubMed] [Google Scholar]
  60. Harijith A, Choo-Wing R, Cataltepe S, et al. A role for matrix metalloproteinase 9 in IFNgamma-mediated injury in developing lungs: relevance to bronchopulmonary dysplasia. Am J Respir Cell Mol Biol. 2011;44:621–630. doi: 10.1165/rcmb.2010-0058OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Harijith A, Pendyala S, Reddy NM, et al. Sphingosine kinase 1 deficiency confers protection against hyperoxia-induced bronchopulmonary dysplasia in a murine model: role of S1P signaling and Nox proteins. Am J Pathol. 2013;183:1169–1182. doi: 10.1016/j.ajpath.2013.06.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Hazinski TA, France M, Kennedy KA, et al. Cimetidine reduces hyperoxic lung injury in lambs. J Appl Physiol. 1989;67:2586–2592. doi: 10.1152/jappl.1989.67.6.2586. (1985) [DOI] [PubMed] [Google Scholar]
  63. Hilgendorff A, Parai K, Ertsey R, et al. Inhibiting lung elastase activity enables lung growth in mechanically ventilated newborn mice. Am J Respir Crit Care Med. 2011;184:537–546. doi: 10.1164/rccm.201012-2010OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Hilgendorff A, Parai K, Ertsey R, et al. Neonatal mice genetically modified to express the elastase inhibitor elafin are protected against the adverse effects of mechanical ventilation on lung growth. Am J Physiol Lung Cell Mol Physiol. 2012;303:L215–L227. doi: 10.1152/ajplung.00405.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Hillman NH, Polglase GR, Pillow JJ, et al. Inflammation and lung maturation from stretch injury in preterm fetal sheep. Am J Physiol Lung Cell Mol Physiol. 2011;300:L232–L241. doi: 10.1152/ajplung.00294.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Hirakawa H, Pierce RA, Bingol-Karakoc G, et al. Cathepsin S deficiency confers protection from neonatal hyperoxia-induced lung injury. Am J Respir Crit Care Med. 2007;176:778–785. doi: 10.1164/rccm.200704-519OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Hislop AA. Airway and blood vessel interaction during lung development. J Anat. 2002;201:325–334. doi: 10.1046/j.1469-7580.2002.00097.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Hogmalm A, Sheppard D, Lappalainen U, et al. beta6 Integrin subunit deficiency alleviates lung injury in a mouse model of bronchopulmonary dysplasia. Am J Respir Cell Mol Biol. 2010;43:88–98. doi: 10.1165/rcmb.2008-0480OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Hummler SC, Rong M, Chen S, et al. Targeting glycogen synthase kinase-3beta to prevent hyperoxia-induced lung injury in neonatal rats. Am J Respir Cell Mol Biol. 2013;48:578–588. doi: 10.1165/rcmb.2012-0383OC. [DOI] [PubMed] [Google Scholar]
  70. Ikegami M, Moss TJ, Kallapur SG, et al. Minimal lung and systemic responses to TNF-alpha in preterm sheep. Am J Physiol Lung Cell Mol Physiol. 2003;285:L121–L129. doi: 10.1152/ajplung.00393.2002. [DOI] [PubMed] [Google Scholar]
  71. Iosef C, Alastalo TP, Hou Y, et al. Inhibiting NF-kappaB in the developing lung disrupts angiogenesis and alveolarization. Am J Physiol Lung Cell Mol Physiol. 2012;302:L1023–L1036. doi: 10.1152/ajplung.00230.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. James ML, Ross AC, Nicola T, et al. VARA attenuates hyperoxia-induced impaired alveolar development and lung function in newborn mice. Am J Physiol Lung Cell Mol Physiol. 2013;304:L803–L812. doi: 10.1152/ajplung.00257.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Joshi S, Kotecha S. Lung growth and development. Early Hum Dev. 2007;83:789–794. doi: 10.1016/j.earlhumdev.2007.09.007. [DOI] [PubMed] [Google Scholar]
  74. Kevill KA, Bhandari V, Kettunen M, et al. A role for macrophage migration inhibitory factor in the neonatal respiratory distress syndrome. J Immunol. 2008;180:601–608. doi: 10.4049/jimmunol.180.1.601. [DOI] [PubMed] [Google Scholar]
  75. Kim DH, Choi CW, Kim EK, et al. Association of increased pulmonary interleukin-6 with the priming effect of intra-amniotic lipopolysaccharide on hyperoxic lung injury in a rat model of bronchopulmonary dysplasia. Neonatology. 2010;98:23–32. doi: 10.1159/000263056. [DOI] [PubMed] [Google Scholar]
  76. Kompass KS, Deslee G, Moore C, et al. Highly conserved transcriptional responses to mechanical ventilation of the lung. Physiol Genomics. 2010;42:384–396. doi: 10.1152/physiolgenomics.00117.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Kornecki A, Tsuchida S, Ondiveeran HK, et al. Lung development and susceptibility to ventilator-induced lung injury. Am J Respir Crit Care Med. 2005;171:743–752. doi: 10.1164/rccm.200408-1053OC. [DOI] [PubMed] [Google Scholar]
  78. Kotecha S. Lung growth: implications for the newborn infant. Arch Dis Child Fetal Neonatal Ed. 2000;82:F69–F74. doi: 10.1136/fn.82.1.F69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Kramer EL, Deutsch GH, Sartor MA, et al. Perinatal increases in TGF-{alpha} disrupt the saccular phase of lung morphogenesis and cause remodeling: microarray analysis. Am J Physiol Lung Cell Mol Physiol. 2007;293:L314–L327. doi: 10.1152/ajplung.00354.2006. [DOI] [PubMed] [Google Scholar]
  80. Kroon AA, Wang J, Huang Z, et al. Inflammatory response to oxygen and endotoxin in newborn rat lung ventilated with low tidal volume. Pediatr Res. 2010;68:63–69. doi: 10.1203/PDR.0b013e3181e17caa. [DOI] [PubMed] [Google Scholar]
  81. Kroon AA, Wang J, Kavanagh BP, et al. Prolonged mechanical ventilation induces cell cycle arrest in newborn rat lung. PLoS One. 2011;6:e16910. doi: 10.1371/journal.pone.0016910. [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Kunig AM, Balasubramaniam V, Markham NE, et al. Recombinant human VEGF treatment enhances alveolarization after hyperoxic lung injury in neonatal rats. Am J Physiol Lung Cell Mol Physiol. 2005;289:L529–L535. doi: 10.1152/ajplung.00336.2004. [DOI] [PubMed] [Google Scholar]
  83. Kunig AM, Balasubramaniam V, Markham NE, et al. Recombinant human VEGF treatment transiently increases lung edema but enhances lung structure after neonatal hyperoxia. Am J Physiol Lung Cell Mol Physiol. 2006;291:L1068–L1078. doi: 10.1152/ajplung.00093.2006. [DOI] [PubMed] [Google Scholar]
  84. Ladha F, Bonnet S, Eaton F, et al. Sildenafil Improves Alveolar Growth and Pulmonary Hypertension in Hyperoxia-Induced Lung Injury. Am J Respir Crit Care Med. 2005 doi: 10.1164/rccm.200503-510OC. [DOI] [PubMed] [Google Scholar]
  85. Lassus P, Turanlahti M, Heikkila P, et al. Pulmonary vascular endothelial growth factor and Flt-1 in fetuses, in acute and chronic lung disease, and in persistent pulmonary hypertension of the newborn. Am J Respir Crit Care Med. 2001;164:1981–1987. doi: 10.1164/ajrccm.164.10.2012036. [DOI] [PubMed] [Google Scholar]
  86. Lee HJ, Choi CW, Kim BI, et al. Serial changes of lung morphology and biochemical profiles in a rat model of bronchopulmonary dysplasia induced by intra-amniotic lipopolysaccharide and postnatal hyperoxia. J Perinat Med. 2010;38:675–681. doi: 10.1515/jpm.2010.091. [DOI] [PubMed] [Google Scholar]
  87. Lee PT, Holt PG, McWilliam AS. Role of alveolar macrophages in innate immunity in neonates: evidence for selective lipopolysaccharide binding protein production by rat neonatal alveolar macrophages. Am J Respir Cell Mol Biol. 2000;23:652–661. doi: 10.1165/ajrcmb.23.5.4016. [DOI] [PubMed] [Google Scholar]
  88. Lee PT, Holt PG, McWilliam AS. Ontogeny of rat pulmonary alveolar macrophage function: evidence for a selective deficiency in il-10 and nitric oxide production by newborn alveolar macrophages. Cytokine. 2001;15:53–57. doi: 10.1006/cyto.2001.0894. [DOI] [PubMed] [Google Scholar]
  89. Li Z, Choo-Wing R, Sun H, et al. A potential role of the JNK pathway in hyperoxia-induced cell death, myofibroblast transdifferentiation and TGF-beta1-mediated injury in the developing murine lung. BMC Cell Biol. 2011;12:54. doi: 10.1186/1471-2121-12-54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Li Z, Choo-Wing R, Sun H, et al. A potential role of the JNK pathway in hyperoxia-induced cell death, myofibroblast transdifferentiation and TGF-beta1-mediated injury in the developing murine lung. BMC Cell Biol. 2011 Dec 15;12:54. doi: 10.1186/1471-2121-12-54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Lindsay L, Oliver SJ, Freeman SL, et al. Modulation of hyperoxia-induced TNF-alpha expression in the newborn rat lung by thalidomide and dexamethasone. Inflammation. 2000;24:347–356. doi: 10.1023/a:1007096931078. [DOI] [PubMed] [Google Scholar]
  92. Londhe VA, Belperio JA, Keane MP, et al. CXCR2/CXCR2 ligand biological axis impairs alveologenesis during dsRNA-induced lung inflammation in mice. Pediatr Res. 2005;58:919–926. doi: 10.1203/01.PDR.0000181377.78061.3E. [DOI] [PubMed] [Google Scholar]
  93. Londhe VA, Maisonet TM, Lopez B, et al. Conditional deletion of epithelial IKKbeta impairs alveolar formation through apoptosis and decreased VEGF expression during early mouse lung morphogenesis. Respir Res. 2011;12:134. doi: 10.1186/1465-9921-12-134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Londhe VA, Sundar IK, Lopez B, et al. Hyperoxia impairs alveolar formation and induces senescence through decreased histone deacetylase activity and up-regulation of p21 in neonatal mouse lung. Pediatr Res. 2011;69:371–377. doi: 10.1203/PDR.0b013e318211c917. [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Ma L, Li N, Liu X, et al. Arginyl-glutamine dipeptide or docosahexaenoic acid attenuate hyperoxia-induced lung injury in neonatal mice. Nutrition. 2012;28:1186–1191. doi: 10.1016/j.nut.2012.04.001. [DOI] [PubMed] [Google Scholar]
  96. Maeda Y, Dave V, Whitsett JA. Transcriptional control of lung morphogenesis. Physiol Rev. 2007;87:219–244. doi: 10.1152/physrev.00028.2006. [DOI] [PubMed] [Google Scholar]
  97. Manisalco WM, Bhandari V. Disruption of lung microvascular development. In: Abman SH, editor. Bronchopulmonary Dysplasia. New York: Informa Healthcare; 2010. pp. 146–166. [Google Scholar]
  98. Maniscalco WM, Watkins RH, Pryhuber GS, et al. Angiogenic factors and alveolar vasculature: development and alterations by injury in very premature baboons. Am J Physiol Lung Cell Mol Physiol. 2002;282:L811–L823. doi: 10.1152/ajplung.00325.2001. [DOI] [PubMed] [Google Scholar]
  99. Martin TR, Ruzinski JT, Wilson CB, et al. Effects of endotoxin in the lungs of neonatal rats: age-dependent impairment of the inflammatory response. J Infect Dis. 1995;171:134–144. doi: 10.1093/infdis/171.1.134. [DOI] [PubMed] [Google Scholar]
  100. Mascaretti RS, Mataloun MM, Dolhnikoff M, et al. Lung morphometry, collagen and elastin content: changes after hyperoxic exposure in preterm rabbits. Clinics (Sao Paulo) 2009;64:1099–1104. doi: 10.1590/S1807-59322009001100010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. McGrath-Morrow S, Lauer T, Yee M, et al. Nrf2 increases survival and attenuates alveolar growth inhibition in neonatal mice exposed to hyperoxia. Am J Physiol Lung Cell Mol Physiol. 2009;296:L565–L573. doi: 10.1152/ajplung.90487.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. McGrath-Morrow SA, Cho C, Soutiere S, et al. The effect of neonatal hyperoxia on the lung of p21Waf1/Cip1/Sdi1-deficient mice. Am J Respir Cell Mol Biol. 2004;30:635–640. doi: 10.1165/rcmb.2003-0049OC. [DOI] [PubMed] [Google Scholar]
  103. McGrath-Morrow SA and Stahl J. Apoptosis in neonatal murine lung exposed to hyperoxia. Am J Respir Cell Mol Biol. 2001;25:150–155. doi: 10.1165/ajrcmb.25.2.4362. [DOI] [PubMed] [Google Scholar]
  104. Meller S, Bhandari V. VEGF levels in humans and animal models with RDS and BPD: temporal relationships. Exp Lung Res. 2012;38:192–203. doi: 10.3109/01902148.2012.663454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Min JH, Codipilly CN, Nasim S, et al. Synergistic protection against hyperoxia-induced lung injury by neutrophils blockade and EC-SOD overexpression. Respir Res. 2012;13:58. doi: 10.1186/1465-9921-13-58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Mokres LM, Parai K, Hilgendorff A, et al. Prolonged mechanical ventilation with air induces apoptosis and causes failure of alveolar septation and angiogenesis in lungs of newborn mice. Am J Physiol Lung Cell Mol Physiol. 2010;298:L23–L35. doi: 10.1152/ajplung.00251.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Naik AS, Kallapur SG, Bachurski CJ, et al. Effects of ventilation with different positive end-expiratory pressures on cytokine expression in the preterm lamb lung. Am J Respir Crit Care Med. 2001;164:494–498. doi: 10.1164/ajrccm.164.3.2010127. [DOI] [PubMed] [Google Scholar]
  108. Nakanishi H, Sugiura T, Streisand JB, et al. TGF-beta-neutralizing antibodies improve pulmonary alveologenesis and vasculogenesis in the injured newborn lung. Am J Physiol Lung Cell Mol Physiol. 2007;293:L151–L161. doi: 10.1152/ajplung.00389.2006. [DOI] [PubMed] [Google Scholar]
  109. O'Reilly MA, Marr SH, Yee M, et al. Neonatal hyperoxia enhances the inflammatory response in adult mice infected with influenza A virus. Am J Respir Crit Care Med. 2008;177:1103–1110. doi: 10.1164/rccm.200712-1839OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Ohki Y, Mayuzumi H, Tokuyama K, et al. Hepatocyte growth factor treatment improves alveolarization in a newborn murine model of bronchopulmonary dysplasia. Neonatology. 2009;95:332–338. doi: 10.1159/000187651. [DOI] [PubMed] [Google Scholar]
  111. Ozdemir R, Yurttutan S, Talim B, et al. Colchicine protects against hyperoxic lung injury in neonatal rats. Neonatology. 2012;102:265–269. doi: 10.1159/000341424. [DOI] [PubMed] [Google Scholar]
  112. Park HS, Park JW, Kim HJ, et al. Sildenafil alleviates bronchopulmonary dysplasia in neonatal rats by activating the hypoxia-inducible factor signaling pathway. Am J Respir Cell Mol Biol. 2013;48:105–113. doi: 10.1165/rcmb.2012-0043OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Pierro M, Ionescu L, Montemurro T, et al. Short-term, long-term and paracrine effect of human umbilical cord-derived stem cells in lung injury prevention and repair in experimental bronchopulmonary dysplasia. Thorax. 2013;68:475–484. doi: 10.1136/thoraxjnl-2012-202323. [DOI] [PubMed] [Google Scholar]
  114. Polglase GR, Hillman NH, Ball MK, et al. Lung and systemic inflammation in preterm lambs on continuous positive airway pressure or conventional ventilation. Pediatr Res. 2009;65:67–71. doi: 10.1203/PDR.0b013e318189487e. [DOI] [PubMed] [Google Scholar]
  115. Rehan VK, Fong J, Lee R, et al. Mechanism of reduced lung injury by high-frequency nasal ventilation in a preterm lamb model of neonatal chronic lung disease. Pediatr Res. 2011;70:462–466. doi: 10.1203/PDR.0b013e31822f58a1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Rehan VK, Torday JS. Lower parathyroid hormone-related protein content of tracheal aspirates in very low birth weight infants who develop bronchopulmonary dysplasia. Pediatr Res. 2006;60:216–220. doi: 10.1203/01.pdr.0000228328.93773.27. [DOI] [PubMed] [Google Scholar]
  117. Reyburn B, Li M, Metcalfe DB, et al. Nasal ventilation alters mesenchymal cell turnover and improves alveolarization in preterm lambs. Am J Respir Crit Care Med. 2008;178:407–418. doi: 10.1164/rccm.200802-359OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Sakurai R, Li Y, Torday JS, et al. Curcumin augments lung maturation, preventing neonatal lung injury by inhibiting TGF-beta signaling. Am J Physiol Lung Cell Mol Physiol. 2011;301:L721–L730. doi: 10.1152/ajplung.00076.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. Schittny JC, Burri PH. Development and growth of the lung. In: Fishman AP, editor. Fishman's Pulmonary Diseases and Disorders. Vol. 1. McGraw-Hill; 2008. pp. 91–114. [Google Scholar]
  120. Shashikant BN, Miller TL, Welch RW, et al. Dose response to rhCC10-augmented surfactant therapy in a lamb model of infant respiratory distress syndrome: physiological, inflammatory, and kinetic profiles. J Appl Physiol. 2005;99:2204–2211. doi: 10.1152/japplphysiol.00246.2005. (1985) [DOI] [PubMed] [Google Scholar]
  121. Shivanna B, Zhang W, Jiang W, et al. Functional deficiency of aryl hydrocarbon receptor augments oxygen toxicity-induced alveolar simplification in newborn mice. Toxicol Appl Pharmacol. 2013;267:209–217. doi: 10.1016/j.taap.2013.01.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Smith VC, Zupancic JA, McCormick MC, et al. Trends in severe bronchopulmonary dysplasia rates between 1994 and 2002. J Pediatr. 2005;146:469–473. doi: 10.1016/j.jpeds.2004.12.023. [DOI] [PubMed] [Google Scholar]
  123. Sohn MH, Kang MJ, Matsuura H, et al. The chitinase-like proteins breast regression protein-39 and YKL-40 regulate hyperoxia-induced acute lung injury. Am J Respir Crit Care Med. 2010;182:918–928. doi: 10.1164/rccm.200912-1793OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Subramaniam M, Bausch C, Twomey A, et al. Bombesin-like peptides modulate alveolarization and angiogenesis in bronchopulmonary dysplasia. Am J Respir Crit Care Med. 2007;176:902–912. doi: 10.1164/rccm.200611-1734OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Subramaniam M, Sugiyama K, Coy DH, et al. Bombesin-like peptides and mast cell responses: relevance to bronchopulmonary dysplasia? Am J Respir Crit Care Med. 2003;168:601–611. doi: 10.1164/rccm.200212-1434OC. [DOI] [PubMed] [Google Scholar]
  126. Sun H, Choo-Wing R, Fan J, et al. Small molecular modulation of macrophage migration inhibitory factor in the hyperoxia-induced mouse model of bronchopulmonary dysplasia. Respir Res. 2013;14:27. doi: 10.1186/1465-9921-14-27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  127. Sun H, Choo-Wing R, Sureshbabu A, et al. A critical regulatory role for macrophage migration inhibitory factor in hyperoxia-induced injury in the developing murine lung. PLoS One. 2013;8:e60560. doi: 10.1371/journal.pone.0060560. [DOI] [PMC free article] [PubMed] [Google Scholar]
  128. Sunday ME, Yoder BA, Cuttitta F, et al. Bombesin-like peptide mediates lung injury in a baboon model of bronchopulmonary dysplasia. J Clin Invest. 1998;102:584–594. doi: 10.1172/JCI2329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  129. Syed MA, Bhandari V. Hyperoxia exacerbates postnatal inflammation-induced lung injury in neonatal BRP-39 mull mutant mice promoting the M1 macrophage phenotype. Mediators Inflamm. 2013 doi: 10.1155/2013/457189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  130. Takeda K, Okamoto M, de Langhe S, et al. Peroxisome proliferator-activated receptor-g agonist treatment increases septation and angiogenesis and decreases airway hyperresponsiveness in a model of experimental neonatal chronic lung disease. Anat Rec (Hoboken) 2009;292:1045–1061. doi: 10.1002/ar.20921. [DOI] [PMC free article] [PubMed] [Google Scholar]
  131. Tambunting F, Beharry KD, Hartleroad J, et al. Increased lung matrix metalloproteinase-9 levels in extremely premature baboons with bronchopulmonary dysplasia. Pediatr Pulmonol. 2005;39:5–14. doi: 10.1002/ppul.20135. [DOI] [PubMed] [Google Scholar]
  132. Tambunting F, Beharry KD, Waltzman J, et al. Impaired lung vascular endothelial growth factor in extremely premature baboons developing bronchopulmonary dysplasia/chronic lung disease. J Investig Med. 2005;53:253–262. doi: 10.2310/6650.2005.53508. [DOI] [PubMed] [Google Scholar]
  133. Tayman C, Cekmez F, Kafa IM, et al. Protective Effects of Nigella sativa Oil in Hyperoxia-Induced Lung Injury. Arch Bronconeumol. 2013;49:15–21. doi: 10.1016/j.arbres.2012.03.013. [DOI] [PubMed] [Google Scholar]
  134. ter Horst SA, Walther FJ, Poorthuis BJ, et al. Inhaled nitric oxide attenuates pulmonary inflammation and fibrin deposition and prolongs survival in neonatal hyperoxic lung injury. Am J Physiol Lung Cell Mol Physiol. 2007;293:L35–L44. doi: 10.1152/ajplung.00381.2006. [DOI] [PubMed] [Google Scholar]
  135. Thebaud B, Ladha F, Michelakis ED, et al. Vascular endothelial growth factor gene therapy increases survival, promotes lung angiogenesis, and prevents alveolar damage in hyperoxia-induced lung injury: evidence that angiogenesis participates in alveolarization. Circulation. 2005;112:2477–2486. doi: 10.1161/CIRCULATIONAHA.105.541524. [DOI] [PubMed] [Google Scholar]
  136. Thomas W, Speer CP. Chorioamnionitis is essential in the evolution of bronchopulmonary dysplasia - The case in favour. Paediatr Respir Rev. 2013 doi: 10.1016/j.prrv.2013.09.004. [DOI] [PubMed] [Google Scholar]
  137. Thomson MA, Yoder BA, Winter VT, et al. Delayed extubation to nasal continuous positive airway pressure in the immature baboon model of bronchopulmonary dysplasia: lung clinical and pathological findings. Pediatrics. 2006;118:2038–2050. doi: 10.1542/peds.2006-0622. [DOI] [PubMed] [Google Scholar]
  138. Thomson MA, Yoder BA, Winter VT, et al. Treatment of immature baboons for 28 days with early nasal continuous positive airway pressure. Am J Respir Crit Care Med. 2004;169:1054–1062. doi: 10.1164/rccm.200309-1276OC. [DOI] [PubMed] [Google Scholar]
  139. Thurlbeck WM. Lung growth and alveolar multiplication. Pathobiol Annu. 1975;5:1–34. [PubMed] [Google Scholar]
  140. Tibboel J, Joza S, Reiss I, et al. Amelioration of hyperoxia-induced lung injury using a sphingolipid-based intervention. Eur Respir J. 2013;42:776–784. doi: 10.1183/09031936.00092212. [DOI] [PubMed] [Google Scholar]
  141. Tillema MS, Lorenz KL, Weiss MG, et al. Sublethal endotoxemia promotes pulmonary cytokine-induced neutrophil chemoattractant expression and neutrophil recruitment but not overt lung injury in neonatal rats. Biol Neonate. 2000;78:308–314. doi: 10.1159/000014285. [DOI] [PubMed] [Google Scholar]
  142. Trembath A, Laughon MM. Predictors of bronchopulmonary dysplasia. Clin Perinatol. 2012;39:585–601. doi: 10.1016/j.clp.2012.06.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  143. Trummer-Menzi E, Gremlich S, Schittny JC, et al. Evolution of gene expression changes in newborn rats after mechanical ventilation with reversible intubation. Pediatr Pulmonol. 2012;47:1204–1214. doi: 10.1002/ppul.22613. [DOI] [PubMed] [Google Scholar]
  144. Vadivel A, Aschner JL, Rey-Parra GJ, et al. L-citrulline attenuates arrested alveolar growth and pulmonary hypertension in oxygen-induced lung injury in newborn rats. Pediatr Res. 2010;68:519–525. doi: 10.1203/PDR.0b013e3181f90278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  145. van Haaften T, Byrne R, Bonnet S, et al. Airway delivery of mesenchymal stem cells prevents arrested alveolar growth in neonatal lung injury in rats. Am J Respir Crit Care Med. 2009;180:1131–1142. doi: 10.1164/rccm.200902-0179OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  146. Velten M, Heyob KM, Rogers LK, et al. Deficits in lung alveolarization and function after systemic maternal inflammation and neonatal hyperoxia exposure. J Appl Physiol. 2010;108:1347–1356. doi: 10.1152/japplphysiol.01392.2009. (1985) [DOI] [PMC free article] [PubMed] [Google Scholar]
  147. Vicencio AG, Lee CG, Cho SJ, et al. Conditional overexpression of bioactive transforming growth factor-beta1 in neonatal mouse lung: a new model for bronchopulmonary dysplasia? Am J Respir Cell Mol Biol. 2004;31:650–656. doi: 10.1165/rcmb.2004-0092OC. [DOI] [PubMed] [Google Scholar]
  148. Viscardi RM. Perinatal inflammation and lung injury. Semin Fetal Neonatal Med. 2012;17:30–35. doi: 10.1016/j.siny.2011.08.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  149. Visser YP, Walther FJ, Laghmani el H, et al. Apelin attenuates hyperoxic lung and heart injury in neonatal rats. Am J Respir Crit Care Med. 2010;182:1239–1250. doi: 10.1164/rccm.200909-1361OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  150. Vosdoganes P, Lim R, Koulaeva E, et al. Human amnion epithelial cells modulate hyperoxia-induced neonatal lung injury in mice. Cytotherapy. 2013;15:1021–1029. doi: 10.1016/j.jcyt.2013.03.004. [DOI] [PubMed] [Google Scholar]
  151. Warner BB, Stuart LA, Papes RA, et al. Functional and pathological effects of prolonged hyperoxia in neonatal mice. Am J Physiol. 1998;275:L110–L117. doi: 10.1152/ajplung.1998.275.1.L110. [DOI] [PubMed] [Google Scholar]
  152. Weichelt U, Cay R, Schmitz T, et al. Prevention of hyperoxia-mediated pulmonary inflammation in neonatal rats by caffeine. Eur Respir J. 2013;41:966–973. doi: 10.1183/09031936.00012412. [DOI] [PubMed] [Google Scholar]
  153. Wilson TC, Bachurski CJ, Ikegami M, et al. Pulmonary and systemic induction of SAA3 after ventilation and endotoxin in preterm lambs. Pediatr Res. 2005;58:1204–1209. doi: 10.1203/01.pdr.0000185269.93228.29. [DOI] [PubMed] [Google Scholar]
  154. Woyda K, Koebrich S, Reiss I, et al. Inhibition of phosphodiesterase 4 enhances lung alveolarisation in neonatal mice exposed to hyperoxia. Eur Respir J. 2009;33:861–870. doi: 10.1183/09031936.00109008. [DOI] [PubMed] [Google Scholar]
  155. Wu S, Capasso L, Lessa A, et al. High tidal volume ventilation activates Smad2 and upregulates expression of connective tissue growth factor in newborn rat lung. Pediatr Res. 2008;63:245–250. doi: 10.1203/PDR.0b013e318163a8cc. [DOI] [PubMed] [Google Scholar]
  156. Wu S, Platteau A, Chen S, et al. Conditional overexpression of connective tissue growth factor disrupts postnatal lung development. Am J Respir Cell Mol Biol. 2010;42:552–563. doi: 10.1165/rcmb.2009-0068OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  157. Yee M, Chess PR, McGrath-Morrow SA, et al. Neonatal oxygen adversely affects lung function in adult mice without altering surfactant composition or activity. Am J Physiol Lung Cell Mol Physiol. 2009;297:L641–L649. doi: 10.1152/ajplung.00023.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  158. Yoo HS, Chang YS, Kim JK, et al. Antenatal betamethasone attenuates intrauterine infection-aggravated hyperoxia-induced lung injury in neonatal rats. Pediatr Res. 2013;73:726–733. doi: 10.1038/pr.2013.51. [DOI] [PubMed] [Google Scholar]
  159. Zhang H, Fang J, Su H, et al. Bone marrow mesenchymal stem cells attenuate lung inflammation of hyperoxic newborn rats. Pediatr Transplant. 2012;16:589–598. doi: 10.1111/j.1399-3046.2012.01709.x. [DOI] [PubMed] [Google Scholar]
  160. Zhang X, Peng W, Zhang S, et al. MicroRNA expression profile in hyperoxia-exposed newborn mice during the development of bronchopulmonary dysplasia. Respir Care. 2011;56:1009–1015. doi: 10.4187/respcare.01032. [DOI] [PubMed] [Google Scholar]
  161. Zhang X, Wang H, Shi Y, et al. Role of bone marrow-derived mesenchymal stem cells in the prevention of hyperoxia-induced lung injury in newborn mice. Cell Biol Int. 2012;36:589–594. doi: 10.1042/CBI20110447. [DOI] [PubMed] [Google Scholar]

RESOURCES