You are on page 1of 34

Am J Physiol Lung Cell Mol Physiol 309: L1239–L1272, 2015.

First published September 11, 2015; doi:10.1152/ajplung.00268.2015. Perspectives

Recent advances in the mechanisms of lung alveolarization and the


pathogenesis of bronchopulmonary dysplasia
Diogo M. G. Silva,1,2 Claudio Nardiello,1,2 Agnieszka Pozarska,1,2 and Rory E. Morty1,2
1
Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the
German Center for Lung Research (DZL), Giessen, Germany; 2Department of Lung Development and Remodelling, Max
Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
Submitted 5 August 2015; accepted in final form 9 September 2015

Silva DM, Nardiello C, Pozarska A, Morty RE. Recent advances alveoli, which were first described by Marcello Malpighi in
in the mechanisms of lung alveolarization and the pathogenesis of 1661 (210, 211), are generated by a process of secondary
bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol septation that occurs during late lung development and are
309: L1239 –L1272, 2015. First published September 11, 2015; uniquely designed to facilitate gas exchange (364). A recent
doi:10.1152/ajplung.00268.2015.—Alveolarization is the process by report has indicated that these two phases of lung development
which the alveoli, the principal gas exchange units of the lung, are
formed. Along with the maturation of the pulmonary vasculature,
are intimately coordinated (61).
alveolarization is the objective of late lung development. The terminal Perturbations to lung development result in lungs that are
airspaces that were formed during early lung development are divided defective for gas exchange. This is exemplified in the clinical
by the process of secondary septation, progressively generating an setting by bronchopulmonary dysplasia (BPD) (169, 218), a
increasing number of alveoli that are of smaller size, which substan- common complication of preterm birth originally described by
tially increases the surface area over which gas exchange can take Northway and coworkers in 1967 (258). Preterm infants with
place. Disturbances to alveolarization occur in bronchopulmonary compromised respiratory function attributable to respiratory
dysplasia (BPD), which can be complicated by perturbations to the distress syndrome (RDS) require oxygen supplementation, of-
pulmonary vasculature that are associated with the development of ten by mechanical ventilation. In these infants, infection,
pulmonary hypertension. Disturbances to lung development may also inflammation, oxygen toxicity, and volu- and baro-trauma
occur in persistent pulmonary hypertension of the newborn in term from ventilation, together with other factors, stunt the post-
newborn infants, as well as in patients with congenital diaphragmatic
natal maturation of the lung (154). This maturational stunt-
hernia. These disturbances can lead to the formation of lungs with
fewer and larger alveoli and a dysmorphic pulmonary vasculature.
ing includes blunted alveolarization and the generation of a
Consequently, affected lungs exhibit a reduced capacity for gas dysmorphic pulmonary vasculature, accompanied by aber-
exchange, with important implications for morbidity and mortality in rant pulmonary vascular wall remodeling, causing pulmo-
the immediate postnatal period and respiratory health consequences nary hypertension (125). BPD is associated with significant
that may persist into adulthood. It is the objective of this Perspectives morbidity and mortality in the neonatal intensive care unit,
article to update the reader about recent developments in our under- and survivors exhibit long-term consequences that persist
standing of the molecular mechanisms of alveolarization and the into adulthood. Similar disturbances to alveolar structure
pathogenesis of BPD. and the pulmonary vasculature are noted in infants with
bronchopulmonary dysplasia; persistent pulmonary hypertension of congenital diaphragmatic hernia (CDH) (317, 344). The
the newborn; pulmonary hypertension; lung development; alveolar- improved medical management of preterm birth has inter-
ization; hyperoxia estingly led to an increase in the incidence of BPD (158,
159). This highlights the need to better understand normal
and aberrant late lung development. In particular, the mo-
THE TRANSPORT OF OXYGEN from the atmosphere into the blood- lecular mechanisms underlying the associated blunted al-
stream, and conversely carbon dioxide from the bloodstream veolarization and disturbed vascular development and ho-
into the atmosphere, is the primary function of the lung. To meostasis associated with BPD remain largely unclear.
optimize gas exchange, the barrier across which gas exchange To this end, there is much activity within the lung develop-
takes place, the alveolo-capillary barrier, must have a surface ment and pediatric communities to better understand the mo-
area as large as possible and must be as thin as possible. Thus lecular mechanisms of normal lung development (242) and the
the objective of lung development is to generate this optimized factors that influence the postnatal maturation of the mamma-
gas-exchange structure (131, 358). The development of the lian lung (366). It is the objective of this Perspectives article to
mammalian lung is broadly divided into two phases, namely, update the reader about advances in our understanding of lung
early lung development (357), when the core lung structure alveolarization reported in the literature since the beginning of
containing the branching conducting airways with their atten- 2013. It is intended that this update will pick up where the last
dant vasculature develops. This is followed by the structural update, published in 2013 (206), ended. This Perspectives
refinement of the gas-exchange surface, through the generation article will first address recent developments in methodologies,
of alveoli, the principal gas-exchange units of the lung. The including new state-of-the-art approaches to quantify the lung
architecture, as well as an overview and critique of the current
animal models employed to study alveolarization. These ele-
Address for reprint requests and other correspondence: R. Morty, Dept. of
Lung Development and Remodelling, Max Planck Institute for Heart and Lung
ments of this Perspectives article will be followed by a report
Research, Parkstrasse 1, D-61231 Bad Nauheim, Germany (e-mail: on novel players in normal and aberrant late lung development,
rory.morty@mpi-bn.mpg.de). either identified by transgenic mouse approaches or pharma-
http://www.ajplung.org 1040-0605/15 Copyright © 2015 the American Physiological Society L1239
Perspectives
L1240 ADVANCES IN LUNG ALVEOLARIZATION AND BPD

cological interventions in animal models of arrested lung investigators confirmed that Rhesus macaques represent a bet-
development. Recent advances in our understanding of the ter model for postnatal lung growth in humans than do rats.
long-term sequelae of arrested alveolarization and the process Furthermore, the studies revealed that the kinetics of postnatal
of neoalveolarization in developed lungs, as well as recent lung growth and alveolarization in rats were more comparable
advances in cell therapy for arrested alveolarization, will also to prenatal lung growth and alveolarization in humans. This
be addressed. idea that alveolarization persists into adolescence has been
supported by limited radiological evidence that alveolariza-
Quantitative Assessment of Lung Structure tion in school-age survivors of extreme prematurity and
term-born children is similar, suggesting that “catch-up”
Being able to robustly quantify structural changes to the lung alveolarization took place in the preterm group (252), al-
architecture is central to the study of normal and pathological though the suitability of 3He diffusion MRI to reach this
lung development (361). Historically, this has been performed conclusion has been questioned (271). As such, the idea of
by many investigators using paraffin-embedded lung tissue, catch-up alveolarization remains controversial. Together,
which is then assessed by light microscopy, usually with the these observations reinforce the utility of combining state-
determination of the mean linear intercept (MLI, or the related of-the-art lung structural analyses with alternative (radio-
mean chord length, Lm) or radial alveolar count (RAC) as a logical or physiological) supportive approaches.
surrogate for alveolar size and, hence, alveolar number. Addi- Presently, the use of stereological approaches to quantify
tionally, the septal thickness has been directly determined, structures in the developing lung is used by relatively few
usually at the “thinnest” point of the septal wall. While pro- groups. This methodology has been pioneered by Tschanz and
viding a wealth of meaningful and valuable data, this approach coworkers (341) to identify a biphasic formation of new alveoli
is facing increasing valid criticism. Among the critiques are the during postnatal rat lung development, in which a bulk forma-
lack of randomization in the sampling, the introduction of tion of new alveoli was observed between postnatal day 4 (P4)
artifacts associated with nonuniform tissue dehydration and and P21. This was followed by a second phase of continued
rehydration (“shrinkage”) during classical fixations protocols, alveolarization between P21 and P60. The stereology approach
and subsequent paraffin embedding. Another concern is the has also been applied to assess total alveoli number and mean
range in hydrostatic pressure used for pressure fixation via the septal wall thickness after pharmacological interventions in
trachea, with some investigators preferring 20 cmH2O, developing mouse pup lungs in a BPD model that relies on
whereas others employ 25 cmH2O. Presently, there is much exposure to hyperoxia (described in detail in the next section)
enthusiasm in the field for the implementation of improved (207, 238) and to assess lung vessel wall thickness during
fixation and embedding techniques and stereological ap- normal lung development in transgenic mice (205). Presently,
proaches to address these problems and to provide robust no stereological approach is available for the quantification of
quantitative data. capillaries in the lung. The development of such a methodol-
A very important study comparing three methods of lung ogy is highly desirable, given the drawbacks of the currently
tissue preparation was published by Schneider and Ochs (309), used surrogates. These surrogates include number of platelet/
in which lung tissue was embedded in paraffin, in glycol endothelial cell adhesion molecule (PECAM), or von Wille-
methacrylate alone (“simple”), or in glycol methacrylate with brand factor (vWF), stained vessels per microscopic high-
osmium tetroxide and uranyl acetate. The authors documented power field. In those approaches, differences in lung inflation,
a 40% shrinkage of cut face areas in the paraffin-embedded artifacts of fixation and embedding, and biased selection of
tissue, a 30% shrinkage with the simple glycol methacrylate, fields may impact the robustness of the data. The stereological
and ⬍3% shrinkage with glycol methacrylate with osmium analysis of lung structure is both technically complicated (in
tetroxide and uranyl acetate. The authors also documented the terms of lung sample preparation) and time consuming, which
superiority of glutaraldehyde-based fixative protocols. This has limited the enthusiasm of the lung community to embrace
study clearly demonstrated the need to move from a paraffin- the methodology despite the clear advantages to acquiring
based to a glycol methacrylate with osmium tetroxide and robust, unbiased data. Looking to the future, the time con-
uranyl acetate-based tissue-embedding system, with glutaral- straints may be circumvented by automation of the tissue
dehyde-based fixation. With this in mind, two review articles section analysis. Efforts in this direction are already underway
have been published in the pages of the American Journal of by the Tremblay group (307); however, there is most likely a
Physiology Lung Cellular and Molecular Physiology recently, long and difficult road ahead before this goal is attained to the
highlighting the challenges presently faced by investigators satisfaction of the stereology community.
interested in the quantitative determination of lung structure
and solutions to these challenges. These articles take a prob- Animal Models of Arrested Alveolarization
lem-based approach to illustrate basic principles of stereology
(267) and to highlight a stereological approach to the study of The study of lung development is entirely reliant on the use
lung structure (248). The stereological and morphological of animal models, and a broad range of animal models has been
analyses of lung tissue, including recent developments, have employed in the past (139). All the presently available BPD
also recently been reviewed (246, 247, 361). animal models have been recently reviewed in the American
Using a stereological approach, Herring and coworkers Journal of Physiology Lung Cellular and Molecular Physiol-
(134) studied postnatal alveolar growth in humans aged 2 mo ogy, including the use of mice (33), rats (264), rabbits (80),
to 15 yr and demonstrated that alveolar growth continues well lambs (15), and baboons (377). Although it is generally ac-
into adolescence. Furthermore, by comparing these data with knowledged that the large animal models, notably preterm
historical data derived from rats and Rhesus macaques, the lambs and preterm baboons, probably represent the best mod-

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00268.2015 • www.ajplung.org


Perspectives
ADVANCES IN LUNG ALVEOLARIZATION AND BPD L1241
els of human disease, there is considerable cost and ethical days of life led to a persistent disturbance to alveolarization
debate associated with the use of these models. For this reason, (assessed by MLI) that was evident 56 days after birth. It is
the mouse and rat models have found the most widespread use. clearly important to better understand how much oxygen is
The key advantages of the mouse and rat models are the required, and when to provoke a particular structural abnor-
relatively low costs, as well as access to transgenic strains, mality in the lung and what impact this has on the magnitude
particularly for mice. Additionally, the small size of newborn of that disturbance (346). To this end, Wang and coworkers
rodent pups translates to lower costs (because of the lower (355) have examined the impact of exposure to 40% O2 and
quantity required) of pharmacological agents delivered. How- 70% O2 over the first 7 days of life, followed by recovery in
ever, critical evaluation of the rodent models also reveals key room air for 14 days, in which no differences were observed in
concerns that are becoming more apparent. The mouse and rat alveolar structure or number (assessed by RAC) comparing the
BPD models are often promoted because mouse and rat pups 40% O2 and 70% O2 groups. However, the alveolar structure
are born in a stage of lung development comparable to the was not assessed at the conclusion of hyperoxia exposure (P7).
stages in which premature humans are born that go on to Another issue of standardization relates to nomenclature, in
develop BPD. As such, these are seen as “relevant” models. which some investigators consider P1 as the day of birth,
However, it is also true that these term mouse and rat pups are whereas others report P1 as the day after birth. Clearly, there is
born with lungs that are already fully competent for gas much room for improvement of the standardization and report-
exchange, which is not the case for premature human infants. ing of experimental protocols and nomenclature in rodent
Furthermore, another significant drawback of the term mouse models of BPD.
and rat models is the difficulty with prolonged mechanical Efforts are underway to further characterize and optimize the
ventilation, which can be undertaken for short periods (up to hyperoxia-exposure protocols in animal models of BPD.
about 24 h) (33, 38) but not beyond. In this sense, the lamb and Rieger-Fackeldy and coworkers (300) demonstrated the impact
baboon models are far superior although these models are also of abrupt vs. gradual weaning of FVB/N mouse pups from
complicated by high costs and ethical considerations. The hyperoxia (85% O2) to normoxia (21% O2). Interestingly, a
recent demonstration that infant baboons infected with respi- gradual weaning (10% reduction in O2 levels per day, over 6
ratory syncytial virus (RSV) develop clinical and pathological days) resulted in double the number of alveoli compared with
changes that parallel those of human infants (269) highlights age-matched controls subjected to abrupt return to room air.
the potential utility of the baboon model, given the relationship These data indicate that, not only the oxygen levels and the
between RSV and BPD in a clinical setting, in which infants duration of exposure, but also the steepness of the oxygen
with BPD are particularly susceptible to RSV infection (57). gradient posthyperoxia exposure can influence alveolarization.
Another key concern, largely with the rodent models, is a These ideas have also received attention in the pioneering work
lack of standardization. This is particularly evident with the of Michael O=Reilly’s group (52), which continues to explore
hyperoxia models. In the many studies reported in this Per- the role of different oxygen levels on lung structural develop-
spectives article, 1) investigators employed a wide variety of ment, particularly in relation to influenza A virus infection.
mouse strains, including BALB/c, C57BL/6, FVB/N, C3H/ There is also a need to further develop alternative animal
HeN, as well as mixed backgrounds. However, there is noto- models of BPD, not based (exclusively) on hyperoxia. The
rious variability in the susceptibility of mouse strains to hy- impact of cell stretch and lung distention is likely to be a key
peroxia-induced lung injury (147, 365), making comparisons cause of damage to the developing lung (151), highlighting the
of studies performed with different strains very difficult. 2) need for increased use of ventilation-based models to study the
This is exacerbated by the use of a broad range of oxygen impact of mechanical ventilation on the developing lung.
concentrations in the hyperoxia-based models, in which term Along these lines, Ratner and coworkers (294) demonstrated
mouse pups are exposed to a range of hyperoxia levels, that an 8-h period of aggressive mechanical ventilation (tidal
between 40% O2 and 100% O2 in the inspired air. 3) A further volume 15 ␮l/g) of 5-day-old mouse pups with room air caused
confounding factor impacting standardization is the duration pronounced mitochondrial dysfunction accompanied by
and window of hyperoxia exposure, in which term mouse pups blunted alveolarization (measured by RAC) that was evident
may be exposed to hyperoxia from the day of birth for up to 28 10 days later, which are important findings given the increasing
days. Alternatively, term mouse pups are exposed for a specific appreciation for mitochondria being more than just a “power-
window (for example, P4 to P8) of hyperoxia, followed by house” (311). However, using the same protocol employing
variable durations of normoxia. This variability in the hyper- gentle mechanical ventilation (tidal volume 8 ␮l/g), no effect
oxia models extracted from the studies reported in this Per- on alveolarization or mitochondrial dysfunction was noted.
spectives article, published between 2013 and 2015, is made Similarly, Young and coworkers (379) demonstrated that lung
clear in Fig. 1. There is a pressing need for a systematic elastase activity can be locally regulated by stretch during
comparison of the effects of different oxygen levels (assessed alveolar septation, which, together with the other studies re-
over the same time range) on lung development. Additionally, ported here, highlights the usefulness of mechanical ventilation
it remains important to assess the critical time window in models of BPD.
which lung development is exquisitely susceptible to oxygen The rabbit is often seen as a compromise between the
injury, should such a window exist. It may well be that oxygen advantages of small rodents (rats and mice) and larger verte-
exposure over the first few hours of postnatal life (in mice) is brates (sheep and nonhuman primates). The rabbit affords the
sufficient to irreversibly perturb lung development, but this possibility of preterm delivery of rabbit kittens, followed by
remains to be experimentally demonstrated. This idea is sup- hyperoxia exposure or other interventions. Additionally, the
ported by the observations of Firsova and coworkers (102) that rabbit lung is anatomically closer to human lungs than are rat
exposure of newborn mouse pups to 90% O2 over the first 4 or mouse lungs. The development of a preterm [at embryonic

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00268.2015 • www.ajplung.org


Perspectives
L1242 ADVANCES IN LUNG ALVEOLARIZATION AND BPD

Prenatal Postnatal (days)


1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 28 56

[163]
[130]
[156,173, 369]
[299]
[323]
[326] 10%
[362]
21%
[270]
[207] 40%
[37, 217, 351, 352] 50%
[27] 60%
[292]
[9, 32, 220]
70%
[239] 75%
[120] 80%
[187]
85%
[9, 50, 156, 288]
[19]
90%
[11] 95%
[19] 100%
[19, 231, 342]
[83]
LPS Injection
[149]
[13]
[324]
[270]
[284]
[355]
[62]
[276]
[319]
[64]
[305]
[23]
[288, 300]
[300]
[236]
[324]
[335]
[8]
[51]

Fig. 1. Spectrum of oxygen levels and oxygen-exposure windows that are employed in the hyperoxia models of bronchopulmonary dysplasia in rodents. Only
hyperoxia-exposure protocols published between 1 January 2013 and 30 June 2015 are illustrated. The legends at right indicate the color scheme used to describe
the different oxygen concentrations applied, as a percentage (vol/vol) of the inspired air (for example, 21% indicates an FIO2 of 0.21).  indicate prenatal or
postnatal bacterial LPS administration. The numbers in parentheses after each bar indicated the corresponding citation in the references from which the
oxygen-exposure protocol was extracted.

day (E)28 vs. the E31 term date] rabbit model, followed by ventilation approach. To this end, the ventilated preterm lamb
hyperoxia exposure (95% O2 for 7 days), has been carefully model continues to be developed (49). Collins and coworkers
described by Richter and coworkers (299), highlighting the (73) have clarified the need for the optimization of this model,
potential utility of this model for studies on arrested alveolar- given the different transforming growth factor (TGF)-␤ re-
ization. Further refinement of this model has also been under- sponses of Merino ewes to intrauterine LPS vs. Ureaplasma
taken (215), demonstrating that caesarean delivery of preterm parvum exposure. Ureaplasma is commonly isolated from
rabbit kittens at E29 vs. E28 was accompanied by reduced pregnant mothers with chorioamnionitis and thus may pose an
mortality although the E29 group maintained a reduction in increased risk for preterm birth and the development of BPD in
alveoli number and thus remained suitable for modelling struc- the neonate (114, 161). The preterm lamb model has also been
tural changes to the lung associated with preterm birth. These used to study ventilation mechanics and most recently the
efforts to further develop the rabbit model are also being utility of recruitment maneuvers before mechanical ventilation
facilitated by transcriptional studies in rabbit lungs from pre- of preterm lambs to limit ventilator-induced lung injury (140).
term rabbit kittens exposed to hyperoxia (306). In that study, Hillman and coworkers (140) found that, irrespec-
So long as rodents cannot be ventilated for prolonged peri- tive of the functional residual capacity recruited, the recruitment
ods, the preterm lamb model remains the method of choice. maneuvers did not alter the acute phase and proinflammatory
This model has recently been used to demonstrate the useful- responses to mechanical ventilation at birth; however, alveolar
ness of high-frequency nasal ventilation to maintain adequate development was not quantified in that study.
gas exchange and to promote alveolarization in preterm lambs Antenatal inflammation can lead to pulmonary inflamma-
(260), which would not have been possible with the rodent tion, with important consequences for pre- and postnatal lung

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00268.2015 • www.ajplung.org


Perspectives
ADVANCES IN LUNG ALVEOLARIZATION AND BPD L1243
development (179), although chorioamnionitis as a risk factor by stereology, using airspace volume density and surface
for BPD remains controversial (180, 333). With antenatal density of septa as surrogates for alveolarization) (239), and,
inflammation in mind, several animal models of BPD have on the basis the expression of surfactant protein and other
been developed to include an intrauterine inflammation com- genes, this model is considered to closely mimic the clinical
ponent, which is translatable to the clinic, as described above situation (120). This concept of fetal priming has also been
for intrauterine Ureoplasma infection in sheep (73). Related observed in the rodent hyperoxia model using maternal diabe-
intrauterine inflammation models have been reported in rats to tes as a first hit, in which streptozotocin-induced diabetes in
study the utility of antenatal glucocorticoids to influence lung rats before pregnancy appeared to modulate the effects of
development in the background of antenatal inflammation hyperoxia on septal wall thickness in pup lungs, although the
(378). Studies have also examined NF-␬B function in the precise implications of this are not clear (173). In terms of
endothelium (329). Further work in preterm lambs examined other new models, the observation that maternal deprivation
inflammatory signaling and glucocorticoid-growth factor sig- (that is, separation of mother and pups) in rats also led to a
naling in fetal sheep lungs (73, 74). These are important and pronounced blunting of lung development (assessed by MLI),
clinically relevant models that demand more widespread use. ostensibly mediated by dipeptidyl peptidase IV, suggests that
To date, BPD has not been modeled in other large verte- maternal deprivation may represent a new (stress-induced) rat
brates apart from nonhuman primates and lambs; however, model of BPD (150). This idea possibly intersects with the
work has been initiated in piglets, with a recent report by recent observation that maternal stress during pregnancy alters
Bartlett and coworkers (31) on the proteomic analysis of the predisposition of pups to allergy susceptibility, in which a
bronchoalveolar lavage fluid and epithelial lining fluid in role for glucocorticoids was implicated (193). This idea has yet
newborn piglets. These authors suggest that the relative simi- to be tested with regard to lung parenchymal and vascular
larity of human and porcine lungs supports the further devel- development. Additionally, Ayala de la Peña and coworkers
opment of animal models in the porcine system. This idea has (26) have suggested that the expression of a KrasG12D variant
been reinforced by Caminita and coworkers (55), who have in the urothelium may represent a new BPD model, an idea
elegantly demonstrated that, between gestational days E98 and based on lung histopathology assessed at P1. The utility of this
E102, preterm piglets exhibited ventilation inadequacies as transgenic mouse strain as a BPD model awaits further evalu-
well as risks for the development of RDS that mimic those of ation, and further genetic models will be discussed later in this
preterm infants born during the saccular phase of lung devel- Perspectives article.
opment. The investigators concluded that these piglets were As an alternative to animal models, several recent reports
compatible with modern neonatal intensive care management have detailed the used of ex vivo lung culture (277), epithelial-
approaches and thus represented an alternative to nonhuman mesenchymal cocultures (122), in vitro-generated human plu-
primates and lambs. ripotent stem cell-derived lung organoids (90), 3D multicell
Although use of the primate models is steadily declining, microtissue culture models of airway smooth muscle (363),
largely attributable to cost and ethical issues, macaques have and a biomaterial-templated alveoli culture system (189) for
recently been used to evaluate the impact of ozone on postnatal the study of alveolarization and alveolar biology. Although not
lung maturation in nonhuman primates by Murphy and co- yet in widespread use, these systems potentially provide useful
workers (250), who noted that susceptibility to ozone damage alternatives to in vivo studies, in which high-throughput
is age dependent and that prior ozone exposure modulates the screening of pharmacological or genetic interventions could be
response to subsequent ozone exposure. These data have conducted to identify pathways relevant to alveolarization and
clearly translatable information to how children living in pol- to limit the use of experimental animals. Among these ap-
luted metropolitan areas during key stages of lung development proaches is the coculture of fetal (but not adult) mesenchymal
may respond to ozone exposure in later life. cells with A549 cells (a human lung epithelial cell line), in
Returning to rodents, intrauterine growth restriction (IUGR) which three-dimensional peaks formed that had a similar cel-
also impacts alveolarization, and thus IUGR is also occasion- lular orientation to alveolar structures in vivo (122). Addition-
ally employed as a BPD model. Zana-Taieb and coworkers ally, there is evidence that secondary septation continues in ex
(383) compared two IUGR models, namely protein deprivation vivo lung slices maintained in culture (277). This provides an
and nitric oxide synthase (NOS) inhibition. The investigators alternative model for the in vitro analysis of signaling path-
concluded that only the protein-deprivation approach yielded a ways and pharmacological or genetic interventions to study
sustained impairment of alveolarization (assessed by MLI and secondary septation. Clearly, this model lacks input from
RAC) in rat pups, whereas NOS inhibition blunted early lung physiological blood flow and cyclic stretch from breathing
development before alveolarization. The same investigators motions, suggesting areas where this potentially interesting in
followed up with a transcriptional profiling study in the pro- vitro approach could be further refined. Along these lines, the
tein-deprivation IUGR model (384) and identified the peroxi- reproducible uniform equibiaxial stretch of precision-cut lung
some proliferator-activated receptor (PPAR) pathway as a slices has recently been demonstrated (82).
likely key mediator of impaired alveolarization. Prenatal Last, concerning the analysis of gene expression in tissues
growth restriction also forms the basis of a new double-hit harvested from animal models, several investigators have sys-
mouse model of BPD, where pregnant mothers are exposed tematically addressed the selection of reference genes in real-
between E14 and E18 to hypoxia (10% O2), after which time PCR analysis. Mehta and coworkers (234) assessed the
offspring were exposed to 75% O2 for the first 14 days of life. utility of Tuba1a, Actb, Gapdh, Rn18S, and Hist4h4 and
Although not yet in widespread use, this model was used to identified Tuba1a as the least variable in terms of expression
demonstrate the therapeutic potential of umbilical cord mono- levels over the course of mouse lung development (E11.5 to
nuclear cells to promote proper lung alveolarization (assessed P28). Unfortunately, other popularly used references genes,

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00268.2015 • www.ajplung.org


Perspectives
L1244 ADVANCES IN LUNG ALVEOLARIZATION AND BPD

including Hmbs and Polr2a, were not evaluated. In a related New Mediators and Modulators of Impaired Alveolarization
study, Bouhaddioui and coworkers (43) screened reference
genes for the normalization of microRNA gene expression and The past 2.5 yr have seen the addition of several new
mediators of alveolarization that have been identified in animal
concluded from the panel of Sno135, Sno142, Sno202, Sno234,
models of BPD. Among these is soluble guanylate cyclase
and Sno251 that the expression of Sno234 was the most stable
(sGC), in which lung development was particularly impaired
over the course of mouse lung development studied (E15.5 to
after hyperoxic injury in mouse pups carrying a targeted
P30) although other popularly used reference genes, such as deletion of the gene encoding the ␣1 subunit of sGC (27).
the U6 small nuclear 1 RNA (Rnu6-1), were not assessed. These data support the idea of activating sGC as a management
strategy in hyperoxia-induced lung injury. Along similar lines,
Imaging Lung Structure and Function McKenna and coworkers (231) demonstrated that sustained
NF-␬B activation, through overexpression of the NF-␬B inhib-
The imaging of the lung in real time is an emerging area of
itor-␤ (I␬-B␤), protected mice against the deleterious effects of
interest and technological development, leading to a special
hyperoxia (95% O2) on alveolarization (assessed by MLI and
call by the American Journal of Physiology Lung Cellular and
RAC), suggesting that the potentiation of NF-␬B activity may
Molecular Physiology for papers, “Real-time visualization of also represent a management strategy in hyperoxia-induced
lung function: from micro to macro” (244). In response to this lung injury. Although not yet studied in a BPD model, over-
call, a review article that broadly described the imaging of the expression of the receptor for glycation end products (RAGE)
lung using a spectrum of tomographic and X-ray approaches blunted alveolarization (assessed by MLI) in mouse pups,
was recently published (116). Recent advances in lung imaging adding RAGE to the catalog of negative regulators of alveo-
include the development of noninvasive MRI approaches to larization (101). Although the receptor for RAGE is expressed
quantify lung volume changes in mice and the application of in alveolar type I cells (304), other studies have indicated that
this approach to monitor interventional studies, in this case, the RAGE expression in the lung is suppressed by exposure of rat
impact of the somatostatin analog pasireotide (SOM230) on the pups to hyperoxia (198); as such, it remains unclear whether
progress of bleomycin-induced lung injury (91). This method- RAGE plays a functional role in limiting alveolarization in
ological approach has not yet been extended to the study of experimental BPD. Further studies on RAGE in lung develop-
postnatal lung development in mice. Very recently, a study has ment are clearly indicated, particularly in light of the recent
emerged (354) that demonstrated that pulmonary MRI can report that RAGE also acts as a mediator of endothelial
reveal quantifiable and significant differences between patients activation (214).
with BPD, premature patients without BPD, and full-term Isoforms of hypoxia-inducible factor (HIF) continue to re-
controls. These new data suggest that pulmonary MRI might be ceive attention as mediators of lung development, including
implemented to individually phenotype disease, which would HIF-1␣ (335), HIF-2␣ (270), and HIF-3␣ (146). The expres-
be most useful for clinical care and to predict outcome. sion of HIF-1 is known to be downregulated in patients with
Other types of imaging are also emerging for the study of BPD, leading Tibboel and coworkers (335) to overexpress a
lung development and BPD. For example, optical cryoimaging constitutively active HIF-1␣ subunit (HIF-1␣⌬ODD) in the
has been employed to address the mitochondrial redox state of distal lung epithelium. This increased lung vessel density
mouse lungs during lung development in Bcl-2-deficient mice, upregulated the expression of proangiogenic factors and drove
in which the authors determined that Bcl-2 deficiency was increased alveolarization in mice maintained in room air.
associated with oxidative stress that correlated with perturbed However, contrary to expectations, when these HIF-1␣⌬ODD-
alveolarization (221). Thus these authors have added fluores- expressing C57BL/6 mice were exposed to hyperoxia (80% O2
cent imaging to our lung development methodology toolbox. for 28 days), lung function worsened, and the surfactant and
Additional recent reports reveal the exciting potential of con- lung architecture abnormalities (assessed by MLI and RAC)
seen in hyperoxia-exposed wild-type mice persisted. Opposite
trast-enhanced micro-CT in rodents (77) and automated intra-
results were obtained by Vadivel and coworkers (342), who
vital microscopy to study vascular function (129); however,
delivered wild-type HIF-1␣ via adenovirus in the rat hyperoxia
these approaches have not yet been applied to developing
model (95% O2 for 14 days) and, in doing so, conferred
lungs. Other tried and tested imaging approaches continue to striking protection against the deleterious impact of hyperoxia
support studies on lung vascular development, for example, on alveolarization (assessed by MLI), which was comparable
barium angiography, which allows the extraordinary visualiza- to that of room air-exposed rat pups. The reasons for the
tion of vascular pruning in the rat hyperoxia model of BPD discordant observations reported by Tibboel and coworkers
(342). (335) vs. Vadivel and coworkers (342) might be due to the
Although not imaging per se, the three-dimensional recon- epithelial-restricted overexpression of constitutively active
struction of serial sections of lungs from patients with BPD has HIF-1␣ in the Tibboel study. This idea is supported by the
been reported by Galambos and coworkers (111) and facili- dramatically improved vascularization seen in the Vadivel
tated the identification of intrapulmonary anastomoses in af- study, which might suggest that the HIF-1␣ must be delivered
fected lungs. These investigators employed the same method- to the endothelium to confer protection and promote lung
ology to describe curious pathological perturbations to lung maturation in the background of hyperoxia.
vascular structure in patients with alveolar capillary dysplasia MicroRNA are also emerging as candidate mediators of
with misalignment of pulmonary veins (ACDMPV) (112, 113) fibroblast (41, 148) and pulmonary artery smooth muscle cell
and CDH (2). This approach has not yet been employed in behavior (157), alveolarization, and BPD (165); however, to
animal models of arrested lung development. date, this idea is supported only by the observed changes in

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00268.2015 • www.ajplung.org


Perspectives
ADVANCES IN LUNG ALVEOLARIZATION AND BPD L1245
expression of some microRNA species that correlate with mation in the background of hyperoxia, it remains unclear
arrested alveolarization. These candidate microRNA species whether an impact on alveolarization accompanies this effect.
have recently been reviewed (165, 249, 375), and integration of Sex differences are emerging as possible confounders in
microRNA pathways into disease processes by modeling (310) studies on clinical and experimental BPD (121), as recently
and systems biology approaches (36, 127, 235) continues to be discussed in the context of pulmonary hypertension (182), and
developed; however, the field awaits the first demonstration of it is important that the sex of experimental animals is consid-
a causal role for any microRNA species in arrested late lung ered in the design of experimental studies and interpretation of
development. Narasaraju and coworkers (251) have suggested data. There is a growing body of evidence that both androgens
a role for miR-150 in blunted alveolarization associated with and steroidogenic enzymes may influence postnatal lung de-
hyperoxia, using a miR-150⫺/⫺ mouse strain, which, after velopment (339). The importance of considering sex bias in
hyperoxia exposure (95% O2 from P1 to P10), exhibited studies in animal models of BPD is underscored by the obser-
transient rescue of alveolarization (assessed by MLI) seen at vations that adult (C57BL/6J) mice exhibit sex-specific dimor-
P6, but this rescue did not persist to P10. Capillary density was phic effects on inflammation and antioxidant enzyme expres-
apparently improved in this model; however, the strain back- sion in response to hyperoxia (196), with male animals exhib-
ground of the miR-150⫺/⫺ mouse strain was not declared, and, iting greater lung injury, neutrophilic inflammation, and
as such, the appropriateness of the C57BL/6J background as a apoptosis, all of which are also characteristic of clinical BPD.
control in these studies cannot be assessed. Some studies in In support of this idea, Martin and coworkers (219) reported a
clinical BPD material have indicated that the expression of pronounced impact of hyperoxia on estradiol metabolism in
miR-206 is downregulated in the lungs of patients with BPD postnatal airway smooth muscle cells. Additionally, the effects
(388); however, miR-206 has yet to be causally implicated in of glucocorticoids on alveolar type II cell function in vitro were
arrested alveolarization. A role in alveolarization for long sex dependent (167). To date, no study has addressed sex
noncoding RNA, which are involved in early lung develop- differences in animal models of BPD, and this represents a
ment (132), is yet to be addressed. priority area for research for future work in the field, particu-
The mechanisms leading to cell death in the lungs in patients larly given the differences in the kinetics of structural changes
with BPD and experimental (hyperoxia-based) animal models in the ageing lung for male vs. female Rhesus macaques (133).
of BPD have also been addressed. One notable recent finding Gasotransmitters including NO, carbon monoxide (CO), and
is that exposure to hyperoxia leads to elevated interferon-␥ hydrogen sulfide (H2S) are also emerging as mediators of late
lung development and, possibly, arrested lung development
levels, which in turn drive endoplasmic reticulum stress-de-
associated with BPD. NO has a long history of use in prema-
pendent cell death in a manner that relies on cyclooxygenase-2
ture infants with or at risk for BPD (168, 340). One recent
(COX-2) (72). This represents a novel cell-death pathway that
report has indicated that treatment of newborn rats with high-
is relevant to clinical and experimental BPD. These findings
dose (20 ppm) inhaled NO improved lung growth, most likely
support the exploration of COX-2- and DNA-damage-induc-
through increased expression of hemeoxygenase-1 and VEGF/
ible transcript 3 (Ddit3; also called Chop)-based approaches in VEGF receptor 2 (VEGFR2), as well as matrix metalloprotei-
the management of BPD, and, indeed, efforts along these lines nases (MMPs). Interestingly, increased gene expression was
are already underway, with the report of Masood and cowork- seen over the course of NO administration but was lost after
ers (220), who employed a highly selective COX-2 inhibitor discontinuation of NO (89). The immediate implication of this
5,5-dimethyl-3-(3-fluorophenyl)4-(4-methylsulfonyl)phenyl- study is not clear because NO administration was made in
2(5H)-furanone in the rat hyperoxia model (60% O2 from P1 to room air to healthy, properly developing rat lungs. Interest-
P14). In that study, COX-2 inhibition blunted inflammatory ingly, 20 ppm NO inhalation yielded an ⬃20% increase in
infiltration provoked by hyperoxia and improved lung vascu- RAC, suggesting that healthy lungs are indeed capable of
larization and alveolarization (assessed by MLI). Conflicting hyperalveolarization. In a different line, continued work on the
data have been obtained in an alternative study by Britt and inhibition of phosphodiesterase (PDE)5 by Park and coworkers
coworkers (50), who could also demonstrate the ability of both (270) demonstrated that sildenafil modulation of the NO/cGMP
aspirin and the COX-2-specific inhibitor celecoxib to limit pathway promoted alveolarization (assessed by Lm) in the
pulmonary inflammation provoked by hyperoxia (85% O2 from background of hyperoxia. These data highlight PDE5 as a
P1 to P14) in C3H/HeN mice; however, pharmacological candidate druggable target to drive alveolarization, and this has
inhibition of COX-2 did not improve lung structure (assessed stimulated additional studies on the interaction between differ-
by number of alveoli per high-power field) in the background ent cGMP- or cAMP-dependent agonists in the pulmonary
of hyperoxia. These data were supported using Cox2⫹/⫺ and vasculature in newborn rats (96). Interestingly, working in
Cox2⫺/⫺ C57BL/6 mice, which were also protected against adult mice, Czövek and coworkers (79) have demonstrated the
pulmonary inflammation provoked by hyperoxia; however, utility of VIP to stimulate the NO pathway and preserve lung
genetic ablation of COX-2 expression did not improve lung function and structure in the background of hyperoxia in adult
structure in the background of hyperoxia. The reasons for these rats. The modulation of NO signaling with VIP in neonates
divergent observations are not clear; however, it is apparent awaits investigation. Future work on inhaled NO approaches in
that no impact of COX-2 inhibition (or gene ablation) was seen hyperoxia-based animal models will have to take into account
at the higher oxygen level, highlighting a possible influence of the impact of hyperoxia on S-nitrosothiol accumulation and
the model (different O2 levels) on the outcome. Additionally, L-type amino acid transporter 1 expression and function, given
one study was performed in rats, whereas the other was the recent important report from Richard Auten’s group that
performed in mice. As such, although the data clearly implicate the oxidative stress generated by hyperoxia blocks a major
COX-2 as a candidate druggable target to limit lung inflam- route of inhaled NO action (45, 46).

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00268.2015 • www.ajplung.org


Perspectives
L1246 ADVANCES IN LUNG ALVEOLARIZATION AND BPD

The alternative gasotransmitter CO has also received atten- both BALB/c and C57BL/6 mice. This could be attenuated by
tion by Anyanwu and coworkers (23), who reported the pro- administration of mecamylamine, a nicotinic acetylcholine
tective effects of CO administration concomitant with hyper- receptor antagonist, to pregnant mothers during gestation,
oxia (75% O2, from P3, for up to 21 days) exposure in which concomitantly with cigarette smoke exposure attenuated
C57BL/6J mice on alveolar development. These studies were the impact of cigarette smoke on alveolarization (assessed by
supported with hemeoxygenase-1 knockout mice, which were MLI) in mouse pups. Some components of cigarette smoke
demonstrably more susceptible to the injurious effects of hy- have also been studied in isolation; Thankur and coworkers
peroxia. This phenomenon has also been reported by Yang and (331) administered benzo[a]pyrene to pregnant rats at E17,
coworkers (374), who suggested that the ␤-catenin/heteroge- E18, and E19. After delivery, newborn pups were exposed to
neous nuclear ribonucleoprotein K axis mediated the protective hyperoxia (85% O2 from P1 to P14). Maternal benzo[a]pyrene
effects of hemeoxygenase-1 during hyperoxia exposure. The exposure appeared to impact alveolarization; however, lung
protective effects of CO were attributed to the modulation of a structure was not quantified in that study. Additionally, the
broad spectrum of inflammatory cytokines, in which the ex- translatability of the very high benzo[a]pyrene dose (25 mg/kg
pression of proinflammatory cytokines was suppressed, and the per day, for 3 days) is unclear, as the comparability of this dose
expression of anti-inflammatory cytokines (IL-10) was in- to benzo[a]pyrene levels in smoking mothers is not reported.
creased by CO treatment. These studies highlight the CO/ Still much remains to be investigated about how exactly
hemeoxygenase-1 axis as a potentially exciting lung-protective cigarette smoke impacts lung development; however, some
axis and also, together with work by Luo and coworkers (201), lead studies, such as the observation that cigarette smoke
emphasize a role for regulators of the Wnt/␤-catenin system in drives hyperplasia and extracellular matrix (ECM) production
regulating lung development. Indeed, given the recent reports by fetal airway smooth muscle cells (347), are laying important
of the ␤-catenin system regulating epithelial repair (185, 385) groundwork in this area. Studies have not been confined to
and smooth muscle responses to hypoxia (381), ␤-catenin is tobacco-containing cigarettes, with a recent report (229) that
emerging as an exciting mediator of the postnatal lung re- exposure of newborn C57BL/6J mouse pups to E-cigarette
sponse to injury, which might impact lung maturation. vapor (containing nicotine in propylene glycol vehicle) once
The third gasotransmitter H2S has also received attention. In per day on days 2 and 3 of life, followed by twice daily on days
pioneering work by Vadivel and coworkers (343), exogenous 4-10 of life, led to an attenuation of alveolarization (assessed
administration of H2S to rats in the hyperoxia model (95% O2 by MLI), compared with mice exposed to propylene glycol
from P1 to P14) improved lung alveolarization (assessed by vehicle alone. The alveolarization defect was accompanied by
MLI), limited hyperoxia-provoked increases in pulmonary ar- detectable systemic levels of cotinine. These data add to the
teriole medial wall thickness, restored the pulmonary arterial argument that exposure to both tobacco and E-cigarettes is an
acceleration time/right ventricular ejection time ratio, and par- important consideration in postnatal lung maturation and as-
tially restored the Fulton index. These data support a protective sume further importance in light of recent reports that perinatal
role for H2S administration, primarily related to restoration of nicotine can induce transgenerational asthma (297) and that
vascular remodeling and pulmonary hemodynamics in the nicotine drives epithelial-mesenchymal transition through
hyperoxia model. In a subsequent study by Madurga and Wnt/␤-catenin signaling in human airway epithelial cells
coworkers (207), daily administration of the H2S donor (390). Apart from cigarette smoke, nanoparticles are another
GYY4137 in the mouse hyperoxia model (85% O2 from P1 to environmental factor that can impact lung development. Nano-
P14) protected C57BL/6 mice against the damaging impact of particles are present in cosmetics, paint, sunscreens, and food
hyperoxia (85% O2 from P1 to P10) on alveolarization (as- (287). Using titanium oxide nanoparticles (average diameter
sessed by stereology). This appeared to be correlated with 8 –10 nm), Ambalavanan and coworkers (22) reported that
blunted inflammatory responses provoked by hyperoxia, per- exposure of mouse pups at P4 to single or multiple doses of
haps mediated by increased IL-10 levels. Taken together, these nanoparticles provoked an increase in the expression of proin-
reports underscore H2S as a mediator of lung development that flammatory mediators and blunted lung development (assessed
warrants further study. This idea was recently reinforced by at P14; by MLI and RAC). In a related study, Chan and
another report by Madurga and coworkers (205), who dem- coworkers (60) described the muted expression of Cyp1A1, a
onstrated that two of the three endogenous H2S-generating cytochrome P-450 superfamily member, in neonatal, but not
enzyme systems, cystathionine ␤-synthase (Cbs) and cysta- adult rats, after exposure to ultrafine particular matter, which is
thionine ␥-lyase (Cth), are mediators of normal lung alveo- a key component of vehicle exhaust. These data underscore the
larization and vascular development. These studies are not need for further investigation into the public health impact of
conclusive, however, because, although the necessity of Cbs exposure of children to nanoparticles and particulate matter
and Cth for alveolarization was documented using knockout during periods of postnatal lung growth.
mice, it remains unclear whether the defective alveolariza- A genome-wide transcriptional analysis approach has been
tion and vascular development seen in knockout mouse pups used by Bhattacharya and coworkers (37) in the mouse hyper-
was due to a lack of H2S production or a toxic buildup of oxia model to identify the aryl hydrocarbon receptor (Ahr) as
enzyme substrates. Furthermore, the contribution of the a candidate key regulator of responses to hyperoxia in
third H2S-generating enzyme, mercaptopyruvate sul- C57BL/6 mouse pups. This observation is interesting because
furtransferase, awaits study. Shang and coworkers (387) have reported that Ahr is necessary
Singh and coworkers (316) have recently added cigarette to protect fetal human pulmonary microvascular endothelial
smoke to the list of potential inducers of arrested alveolariza- cells against hyperoxic injury. Genome-wide association stud-
tion. Interestingly, gestational (maternal) but not postnatal ies (GWAS) have been used by Nichols and coworkers (254)
exposure to cigarette smoke blunted alveolar development in to evaluate the role played by genetic background in the

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00268.2015 • www.ajplung.org


Perspectives
ADVANCES IN LUNG ALVEOLARIZATION AND BPD L1247
response of mouse pups from 36 inbred mouse strains to In addition to these newly identified mediators of alveolar-
hyperoxia (95% O2 from P1 to P3). These investigators iden- ization, several recent studies have identified changes in the
tified the muscarinic acetylcholine receptor M2 gene (Chrm2) expression of further candidate mediators that accompany
as a susceptibility gene, and mouse strains carrying a Chrm2 disturbances to lung alveolarization, which have yet to be
single-nucleotide polymorphism (SNP; causing a P265L sub- validated in interventional or transgenic mouse studies. These
stitution in Chrm2) exhibited reduced hyperoxia-induced in- candidate mediators include neuronal Per Arnt Sim domain
flammation. Importantly, that study employed hyperoxia-in- protein 3 (274) and SOD-3 (283). SOD-3 is an antioxidant that
duced lung inflammation as an endpoint readout, and similar is curiously targeted to the ECM and epithelial lining fluid of
GWAS studies using perturbations to lung architecture have the lung and is not regulated by nuclear factor, erythroid-
not yet been performed. GWAS have been less successful in derived 2-like 2 (Nfe2l2, also called Nrf2) (283) and adds
patients with BPD, in whom, in one recent report, no new SOD-3 and thioredoxin-1 (66) to the list of antioxidants that
SNPs were identified that could be associated with BPD; might play a role in perturbed lung alveolarization in response
furthermore, this study could not validate any previously iden- to hyperoxia (103, 337). This idea is all the more interesting
tified SNPs (356). In another GWAS involving material from given the recent report by Gupta and coworkers (124) that loss
patients with BPD, no new SNPs were identified, but Amba- of SOD-2, studied in Sod2⫹/⫺ C57BL/6 mice, did not impact
lavanan and coworkers (20) highlighted the miR-219 pathway arrested alveolarization (assessed by changes in alveolar area),
[which includes platelet-derived growth factor (PDGF)-␣] and changes in Fulton index, or pulmonary arteriole medial wall
the IGF family pathway as areas that warrant further study in thickness that were caused by hyperoxia (75% O2, from P1 to
BPD. Studies on DNA methylation patterns are also emerging P14) exposure, leading these authors to conclude that other
(78), which have implicated epigenetic regulation of growth antioxidant systems may compensate the loss of SOD-2 or that
factor, ECM, and antioxidant enzyme expression during nor- a single sod allele generated sufficient SOD to elicit a wild-
mal lung development and epigenetic (dys)regulation of de- type response.
toxifying enzymes and growth factors in clinical BPD (126).
Interestingly, whole-genome expression studies in plasma sam- Epithelial Cells and Epithelial Cell Transdifferentiation
ples in patients with BPD revealed alteration of the expression
Epithelial growth and differentiation and epithelial interac-
of nearly 10% of the genome in affected patients (279).
tions with the ECM are key events in alveolarization. Using a
Recent studies have also shed new light on classical medi- lung epithelium-specific [surfactant-associated protein C
ators and pathways that remain controversial players in lung (Sftpc)-driven] deletion of integrin-␤1, Plosa and coworkers
development and candidate druggable targets and pathways in (281) demonstrated a pivotal role for integrin-␤1 in branching
BPD. Lykkedegn and coworkers (203) thoughtfully appraised morphogenesis and alveolarization. The alveolarization defect
the impact of vitamin D on lung maturation in a systematic was accompanied by abnormal epithelial differentiation, as
review and highlighted the discord between observations made well as blunted secondary septation (assessed by MLI), exces-
in animal models, in which vitamin D dramatically stimulated sive ECM production, hypercellularity of the mesenchyme, and
lung maturation, whereas clinical data currently do not support persistent alveolar inflammation. Using a clodronate approach,
a role for vitamin D in the prevention or treatment of infant the investigators further demonstrated that disturbed alveolar-
RDS or BPD. This report highlights a need for further studies ization after loss of integrin-␤1 in the epithelium was second-
on vitamin D deficiency or insufficiency as a risk factor for ary to persistent inflammation. These investigators proposed
RDS and BPD and supported further investigations into the that loss of integrin-␤1 in the epithelium increased epithelial
optimal dosing (in terms of dose and timeframe) of vitamin D production of chemokine (C-C motif) ligand 2 (Ccl2), chemo-
and its analogs for the managements of infant RDS and BPD. kine (C-X-C motif) ligand 1 (Cxcl1), and reactive oxygen
To this end, Mandell and coworkers (213) reported that ante- species (ROS), which drove increased recruitment of macro-
natal vitamin D administration improved survival and lung phages, blunting alveolar development. These observations add
structure (assessed by RAC) after intra-amniotic endotoxin to a growing body of data, discussed in detail below, implicat-
exposure in rats [which is also known to modulate airway ing inflammation as a key mediator of both normal and aber-
reactivity in mice following hyperoxia exposure (71)], suggest- rant lung alveolarization. Lafemina and coworkers (181) ad-
ing a potential role for vitamin D in the prevention of arrested dressed a role for a related cell-communication and adhesion
alveolarization in this rat model. Along these lines, Yurt and molecule, claudin 18, in postnatal lung growth. Claudin 18 is
coworkers (382) demonstrated a biphasic response to cholecal- reported to be a lung-specific tight junction protein that is
ciferol nutritional supplementation, in which both alveolar abundant on alveolar type I cells. Genetic ablation of claudin
development (assessed by RAC and MLI) as well as PPAR-␥ 18 expression had no impact on newborn mouse lung structure
and CCAAT/enhancer binding protein-␣ expression in the lung but did stunt postnatal lung maturation (assessed by MLI). The
peaked at a supplementation of 500 U/kg cholecalciferol, precise role played by claudin 18 in postnatal lung growth is
which were all reduced at both 250 U/kg and 1,000 U/kg. not yet clear; however, loss of Cldn18 did upregulate the
These data highlight the concerns about dose in vitamin D expression of amphiregulin, sonic hedgehog (Shh), and elastin
supplementation studies. In the context of the impact of nutrition and downregulated the expression of fibroblast growth factor
on lung development, Mayor and coworkers (222) identified (FGF) receptor (FGFR)4, adrenomedullin, and VEGF-A, hint-
maternal high-fat diet as a risk factor for alveolar simplification ing at a possible mechanism. Along these lines, p66Shc (188),
(assessed by MLI), possibly related to placental inflammation. IL-1␤ (142), and homeobox proteins Hox3a and Hox5a (42)
These studies provide a firm basis for further work on the role of have also recently been added to the list of new mediators of
maternal nutrition on postnatal lung maturation. lung epithelial cell differentiation that impact the structural

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00268.2015 • www.ajplung.org


Perspectives
L1248 ADVANCES IN LUNG ALVEOLARIZATION AND BPD

development of the lung although the underlying mechanisms protein 6 accounted, at least in part, for the effects of Sox2 on
remain to be fully elucidated. Additionally, lung epithelial stem-cell phenotype (266). This finding most likely has rele-
TNF-␣-converting enzyme (TACE) was implicated by Xu and vance for the regeneration lung tissue, perhaps by modulating
coworkers (373) as a regulator of lung saccular formation, in Sox2 levels. Along the lines of this thinking, Jain and cowork-
which genetic ablation of epithelial TACE retarded fetal lung ers (155) demonstrated a surprising plasticity of type I cells,
development but not postnatal alveolarization. As yet, the which up until now have been considered terminally differen-
molecular mechanisms at play in this process have not been tiated, as well as a bidirectional relationship between differen-
defined. tiated distal alveolar epithelial cells in a process regulated by
The dynamics of type II to type I cell differentiation are TGF-␤. With these ideas in mind, Desai and colleagues (86)
currently a hot topic in lung developmental biology, and Hou have challenged the view that type I cells arise from type II
and coworkers (143) have provided data to suggest that hyper- cells, providing data to suggest that type I and type II cells arise
oxia drives type II cell transdifferentiation in neonatal rat pups directly from a bipotent progenitor in the prenatal lung but can
although the relevance to disease and the molecular mecha- arise from type II cells in the postnatal lung, a stem cell
nisms at play have not been clarified. Zhao and coworkers function that is activated by type I cell injury. The next few
(389) made the striking observation that the balance between years will no doubt see much progress in our understanding of
TGF-␤ and bone morphogenetic protein signaling directs the the interrelated plasticity of type I and type II cells, and this
transdifferentiation of type II cells into type I cells, perhaps work has received further clinical impetus by the recent report
hinting at a mechanism to explain the observations of Hou and that alveolar stem cell failure may be related to telomere
coworkers (143). Further to this idea, Ghosh and coworkers dysfunction (17).
(117) identified antagonism between Wnt3a and Wnt5a as a Work in primary type I and type II cells continues to be
mediator of alveolar epithelial cell phenotype and proposed hampered by the lack of 1) availability of type I cells for in
that IGF-1 stimulated type II to type I cell transdifferentiation vitro studies and 2) efficient delivery of genes to alveolar type
through activation of Wnt5a. This process also drove cell II cells, other than by viral-mediated transduction. Some prog-
wound repair in vitro. These ideas are integrated into a scheme ress has been made on both fronts over the past two years. Kim
in Fig. 3, are emphasized by the demonstration by Keith and coworkers (166) have reported the enrichment of alveolar
Tanswell’s group that the IGF/IGF receptor (IGFR)1 system is type I cell preparations for in vitro studies by panning with
a key regulator of alveolarization (192), and tie into earlier commercially available anti-rat podoplanin (T1␣) IgG. Grzesik
work by Tom Mariani’s group (318), in which epithelial/ and coworkers (123) have also recently reported the ability to
mesenchymal cross talk and IGF-1 contributed to FGFR- deliver genes on plasmids to primary alveolar epithelial cells
dependent alveolar elastogenesis and proper airspace forma- by nucleofection technology, and Ramachandran and cowork-
tion. The ability to drive repair is likely to be relevant to the ers (291) have described the efficient delivery of RNA inter-
lung response to ventilation because ventilation and cyclic ference oligonucleotides to polarized airway epithelia in vitro.
stretch have recently been described by Kroon and coworkers Although none of these methodologies has yet found wide-
(176) to drive alveolar type II cell (but not fibroblast) death by spread use, these important advances will no doubt facilitate
the extrinsic apoptotic pathway. These and other data reinforce further studies with alveolar type I and alveolar type II cells in
the idea that promoting epithelial repair is a candidate strategy the future. Work is also currently ongoing to develop unbiased
to manage aberrant alveolarization following hyperoxia and protein screening approaches to detect quantitative changes in
ventilator injury to the lung. Consistent with this idea, Kim and protein expression in type II cells in response to hyperoxia
coworkers (166) identified tripartite motif-containing 72 using isobaric labeling (isobaric tags for relative and absolute
(Trim72) as a mediator of alveolar repair in ventilated adult quantitation) with tandem mass spectrometry (35). To date, this
mouse lungs, in which overexpression of Trim72 protected approach remains at the proof-of-principle stage.
against, and genetic ablation of Trim72 increased, susceptibil-
ity to ventilator-induced lung injury. The molecular mecha- Fibroblasts and Fibroblast Differentiation
nisms of the protection conferred by Trim72 are not known;
however, these observations made in adult mice highlight McGowan and McCoy (227) have examined the molecular
Trim72 as a molecule that warrants investigation in neonatal pathways that direct fibroblast migration during alveologen-
models of hyperoxic and ventilator-induced lung injury. esis. These investigators reported data that suggest that Shh,
Several investigators have recently built on the key finding which is also known to play a role in lung fibrosis (209), directs
of Barkauskas and coworkers (30), who demonstrated that type the uniform orientation of lung fibroblasts and organizes the
II cells are stem cells in the adult lung. Among key develop- migration of lung fibroblasts toward the distal secondary sep-
ments in this fast-moving field are observations recently made tum. These investigators also made the observation that a
by Ochieng and coworkers (162), who documented that induc- larger proportion of PDGF receptor (PDGFR)␣-expressing
tion of sex determining region Y-box 2 (Sox2) in type II cells lung fibroblasts bore more primary cilia than other cells present
in vivo could “deprogram” type II cells, which reverted to a in the alveolus did, and this may impart increased migratory
less differentiated cell type that expressed the stem cell mark- capacity to these cells and all these cells to respond to PDGF-A
ers lymphocyte antigen 6 complex, locus A (also called Sca1), chemotactic gradients. Lu and coworkers (200) further ex-
and fucosyltransferase 4 (also called Ssea1), and coexpressed plored upstream mediators of Shh signaling and, using
Sftpc and secretoglobin, family 1A, member 1 [Scgb1a1; also Eya1⫺/⫺ and Six1⫺/⫺ knockout mice and Eya1⫺/⫺/Six1⫺/⫺
called club (formerly Clara) cell protein 10], which are char- double-knockout mice, identified that the transcription factors
acteristics of bronchoalveolar stem cells. The ability of Sox2 to Eya1 and Six1 act together to regulate saccular development by
regulate transformation-related protein 63 and GATA-binding modulating Shh levels.

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00268.2015 • www.ajplung.org


Perspectives
ADVANCES IN LUNG ALVEOLARIZATION AND BPD L1249
The characterization of mesenchymal progenitors in the the tumor-associated calcium signal transducer 2 (224, 225)
embryonic lung as well as the postnatal lung continues to be an and cysteine-rich secretory protein LCCL domain containing 2
area of intensive efforts. Recent advances include the demon- (also Lgl1) (386). How these mediators function to impact lung
stration by El Agha and coworkers (92, 93) that Fgf10-express- development awaits clarification. Other elements of fibroblast
ing cells represent a pool of mesenchymal progenitors in both behavior have also received attention; for example, MMP-9
the embryonic and the postnatal lung, possibly suggesting that was identified as a mediator of fibroblast contractility in a
Fgf10-positive cells might be useful for the development of TGF-␤-dependent mechanism (171). The lung mesenchyme is
stem cell-based therapies for parenchymal lung diseases (65). regarded as a regulator of lung development (223), and coming
Furthermore, using tracing based on the expression of the work in this area will no doubt address roles for fibroblast
Wilm’s tumor suppressor gene, a reliable marker for cells subsets in directing alveolarization, not only through the pro-
derived from the mesothelium, Cano and coworkers (56) de- duction of the lung ECM “skeleton”, but also through cross
scribed the extraordinary heterogeneity of mesenchymal-de- talk between the mesenchyme, in particular, as a regulator of
rived cells during lung morphogenesis. These studies continue epithelial cell behavior (152, 348). Looking ahead, Li and
to set the stage for our understanding of the original of “the coworkers (190) have recently reported that progenitors of
cellular players” in late lung development. secondary crest myofibroblasts are developmentally committed
Fibroblast differentiation and the existence and function of in the early lung mesoderm and highlighted the utility of the
fibroblast subsets are an exciting area of intensive work in the Gli1-CreERT2 line for the study of formation of the alveoli.
lung development field. Lipofibroblasts in particular have re-
ceived much attention. This fibroblast subset is characterized ECM Production and Remodeling
by the presence of lipid bodies although the presence of
lipofibroblasts in the human lung remains controversial. Using Alveolarization occurs through a complex process of cellular
light and electron microscopy, Tahedl and colleagues (327) and ECM interactions (226) assisted by mesenchymal and
reported that, although lipofibroblasts could be detected in other progenitors and physical forces such as breathing mo-
adult rodent lungs, lipofibroblasts were not noted in any other tions. Recent work addressing roles for the ECM in lung
mammalian lungs examined, including human, dog, goat, and development has examined both the production of ECM com-
horse lungs, among others. Furthermore, lipofibroblasts were ponents (Fig. 3), as well as subsequent assembly and remod-
not seen in postnatal human lungs although this cell type was eling of the ECM, building on the pioneering work of Richard
abundant during postnatal mouse lung development. Ahlbrecht Bland and Kurt Albertine and colleagues (16, 38 – 40). One
and McGowan (7) subsequently pointed out that these data recent study by Hilgendorff and coworkers (138) reported that
conflicted with reports of lipofibroblasts in human lungs (298) elastin haploinsufficient C57BL/6 mice exhibited perturbed
and suggested that alternative methodologies to identify lipo- lung capillary growth and attenuated elastin deposition in the
fibroblasts in autopsy tissue may play a role in whether lipo- developing parenchyma although no alveolarization defect was
fibroblasts are detected or not. Clearly, additional work re- noted (assessed by Lm). This study provides further impetus for
mains to be done to clarify the existence of lipofibroblasts at the study of collagen dynamics as well as elastin and collagen
particular time points in the development of the lung across cross linking in alveolarization.
several species and also to clarify a functional role for lipofi- In this line, there is currently much interest in the balance
broblasts in alveolarization. between increased ECM production and impaired ECM deg-
The presence or role of the lipofibroblast and a role for FGF radation, in the context of fibrotic lung disease (97). This topic
and PDGF signaling in arrested lung development associated has been reviewed in detail recently, while asking whether
with BPD (228) and CDH (104 –108) have also received recent idiopathic pulmonary fibrosis is a disease of impaired ECM
attention, with one recent report highlighting the temporal degradation, rather than the classical view of increased ECM
commitment of the PDGFR-␣⫹ cell lineage to lipofibroblasts production (232). This idea may be extended to lung develop-
and myofibroblasts during late lung development (259). Con- ment. Several recent studies have addressed the posttransla-
tinuing in this line, McGowan and McCoy (228) also suggested tional processing of collagen and elastin fibers in experimental
that a transition from lipofibroblasts to myofibroblasts in BPD and clinical BPD. Studies have included the examination of
drives thickened alveolar septa attributable to collagen depo- enzyme systems that direct the cross linking of collagen and
sition. These investigators further suggested that PDGFR-␣- elastin fibers, which include the lysyl oxidase, lysyl hydroxy-
driven Sox9 expression maintained the lipofibroblast state, and lase, and transglutaminase enzyme families. The expression of
that Sox9 expression was lost with the acquisition of the lysyl oxidases (namely Lox and LoxL1) (178), lysyl hydrox-
myofibroblast state. As such, targeting PDGFR-␣ downstream lyases (namely lysyl hydroxylase 2, also called Plod2) (369),
signaling pathways (involving Sox9) may represent a strategy and transglutaminase 2 (368) has been associated with BPD in
to drive fibroblasts from the myofibroblast back to the fibro- clinical subjects and with arrested alveolarization in the hyper-
blast phenotype, which would limit profibrotic activity and oxia-based (85% O2 for 14 days) C57BL/6 mouse model of
possibly drive lung (re)generation. These ideas are presented BPD. Studies employing pharmacological interventions or
schematically in Fig. 2. These data have a clinical implication knockout mouse strains are required to causally implicate these
as well, given the report of Popova and colleagues (284), who enzyme families in aberrant lung development. To this end,
documented that mesenchymal stem cells from patients with Mižíková and coworkers (238) set out to test a pathogenic role
BPD hold stable alterations to PDGFR-␣ gene expression that for lysyl oxidases in arrested alveolarization associated with
drive hypoalveolarization. hyperoxia exposure. Inhibition of lysyl oxidases with ␤-amino-
Several new mediators of fibroblast motility, proliferation, propionitrile (BAPN) in developing C57BL/6 mouse pups in
and ECM production have recently been identified, including the hyperoxia BPD model was without effect on alveolariza-

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00268.2015 • www.ajplung.org


Perspectives
L1250 ADVANCES IN LUNG ALVEOLARIZATION AND BPD

Mechanical
ventilation

O2

Activation of
MMP-2/9

Increase of
Periostin Glucorticoids
TGF-β signaling

FIBROBLAST

Loss of Sox9
Hif-2α Decreased sCG activity
activation Gain of Sox9 PDGF-A

(MYO)FIBROBLAST
Increased
miR-210 Trop2
expression Crispld2

Increased expression Increased


Blocked Mnt Increased Increased
of ECM remodeling expression of
expression proliferation migration
enzymes ECM components

Blocked
miR-206
expression
Fibronectin Collagen Elastin

BAPN
Lysyl oxidase
Transglutaminases
Lysyl hydroxylase

Arrested Increased ECM


alveolarization stability

Fig. 2. Integration of selected, recently identified signaling pathways that can be induced by either hyperoxia or mechanical ventilation, which might impact
fibroblast function during alveolarization. The scheme only details new developments that were published between 1 January 2013 and 30 June 2015. Crispld2,
cysteine-rich secretory protein LCCL domain containing 2; MMP-2/9, matrix metalloproteinase 2 and 9; TGF-␤, transforming growth factor-␤; Sox9, sex
determining region Y-box 9; sCG, soluble guanylate cyclase; Trop2, trophoblast antigen 2; HIF-2␣, hypoxia-inducible factor 2␣; ECM, extracellular matrix; miR,
microRNA; BAPN, ␤-aminopropionitrile; MNT, MAX network transcriptional repressor.

tion (assessed by stereology) at P10 and worsened lung devel- animals (238), although BAPN is well tolerated in adult mice
opment at P20, perhaps indicating a need for a careful balance when organogenesis is complete (253). Contrasting data were
of lysyl oxidase activity during organogenesis (238). Indeed, obtained by Mammoto and coworkers (212), who reported that
BAPN proved to be very toxic in mouse pups, limiting the lysyl oxidase inhibition with BAPN improved alveolarization
suitability of BAPN as an interventional tool in developing (assessed by MLI) in the mouse hyperoxia model. Both groups

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00268.2015 • www.ajplung.org


Perspectives
ADVANCES IN LUNG ALVEOLARIZATION AND BPD L1251

BMP signaling

O2
Trim72
Fig. 3. Some recently described pathways by
which hyperoxia might modulate type II cell-
IGF-I IL-1β
type I cell transdifferentiation. The proposed
roles for bone morphogenetic protein (BMP)
Wnt5a Wound healing signaling, IGF, IL-1␤, TGF-␤ signaling, tripar-
tite motif-containing 72 (Trim72), Wnt3a and
Wnt5a, are indicated. The scheme only details
Wnt3a new developments that were published between
Alveolar epithelial Alveolar epithelial 1 January 2013 and 30 June 2015.
Transdifferentiation
type II cell type I cell

TGF-β signaling

employed C57BL/6 mice and a 10-day hyperoxia-exposure the two studies, it remains unclear whether periostin partici-
period; however, Mižíková (238) employed 85% O2 and a pates in aberrant alveolarization. There are several possible
stereological analysis, whereas Mammoto (212) employed explanations for the opposing trends observed by the two
75% O2 and a morphometric (MLI) analysis. Whether these groups of investigators. It is clear that different oxygen levels
differences explain the discordant observations is unclear. A and different exposure protocols were employed. Additionally,
weakness of the Mammoto study (212) is that the investigators two different mouse strains and backgrounds have been em-
supplemented maternal nutrition with BAPN and assumed ployed, in which Ahlfield and coworkers (9) employed a
maternal transmission to pups through the breastmilk; how- mouse strain (301) in which exon 1 of the periostin gene was
ever, no data on BAPN levels or lysyl oxidase activity levels in insertionally inactivated with a LacZ cassette. Additionally,
the lungs of pups were provided. As such, it is not known this strain was backcrossed over eight generations onto a
whether lysyl oxidase activity levels were impacted at all in C57BL/6J background. In contrast, Boyzk and coworkers (44)
hyperoxia-exposed neonates. The investigators claimed to have employed the B6;129-Postntm1Jmol/J strain, in which the exons
repeated the intervention with intraperitoneal application of 4 –10 of the periostin gene were replaced with a neomycin
BAPN every other day to mouse pups and apparently obtained resistance cassette (268), and the strain was maintained on a
comparable effects on protecting the developing lung from mixed C57BL/6 and 129 background. As a further potential
hyperoxia-induced arrest of alveolarization. However, these confounding variable, periodontal disease was present in
data are neither provided in the manuscript proper nor in the Postn⫺/⫺ mice, wild-type mice, and knockout animals fed with
online supplement. These studies highlight the need 1) to powdered chow by Boyzk and coworkers (44). These studies
demonstrate the delivery of pharmacological agents to the exemplify intermodel comparisons that arrive at opposite con-
target organs or at least to demonstrate that pharmacological clusions, possibly attributable to the use of different oxygen
agents that were applied did have the desired impact in the injury protocols or different background strains, as highlighted
target organs; and 2) to provide supporting data, in this case, in Animal Models of Arrested Alveolarization, or attributable to
the intraperitoneal application of BAPN in the Mammoto study different approaches to targeting genes in model animals.
(212), which could have been compared with the analogous Changes in ECM abundance in the airways have recently
study by Mižíková (238). been correlated with lung function in equine heaves (314), and
Other ECM components such as periostin, a ligand for ␣v␤3 the influence of mechanical ventilation on ECM production in
and ␣v␤5 integrins to support the adhesion and migration of neonatal lungs has also recently been addressed. Kroon and
epithelial cells, have also received attention (9, 44). Both coworkers (177) demonstrated that mechanical ventilation of
studies localized periostin to myofibroblasts and clearly indi- 8-day-old mouse pups with low (3.5 ml/kg), moderate (8.5
cated that periostin (Postn) expression is driven by hyperoxia. ml/kg), or high (25 ml/kg) tidal volumes impacted ECM gene
Ahlfield and coworkers (9) demonstrated that hyperoxia-in- expression. Increased expression of the tropoelastin, fibulin 5,
duced arrest of alveolarization has not been impacted (assessed lysyl oxidase-like 1, and tenascin C genes was noted after an
by MLI) by global loss of periostin in C57BL/6J Postn⫺/⫺ 8-h high tidal volume ventilation with room air. This was
mice, when mice were exposed to 60% O2 or 85% O2 between correlated with an altered appearance of parenchymal elastin
P1 and P7 and assessed at P7. Opposing data were obtained by fibers and reduced vessel density; however, lung structure was
Boyzk and coworkers (44), who reported that exposure of not quantified. These data demonstrate the power of mechan-
Postn⫺/⫺ mice to 75% O2 from P2 or P3 for 14 days fully ical ventilation to drive ECM gene expression, but the impact
protected against hyperoxia-induced arrest of alveolarization of altered gene expression reported in this study on alveolar-
(assessed by Lm). These investigators also described a circle of ization remains unclear.
periostin driving TGF-␤ and TGF-␤ driving periostin expres- The ability to therapeutically modulate ECM production by
sion (highlighted in Fig. 2). Given the opposing trends noted in fibroblasts continues to be an area of interest in terms of

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00268.2015 • www.ajplung.org


Perspectives
L1252 ADVANCES IN LUNG ALVEOLARIZATION AND BPD

influencing aberrant late lung development in premature neo- identified as the mediator of epithelial-to-mesenchymal transi-
nates. Among the key reports that have emerged in the past 2.5 tion in the lungs of rat pups exposed to hyperoxia (85% O2
yr are the exciting finding that glucocorticoids such as flutica- from P1 to P7) (350). Using transgenic mice that overexpress
sone can have divergent effects on ECM production by fibro- TGF-␤ or mice with genetic ablation of the type II TGF-␤
blasts, in which fluticasone drove decorin production in airway receptor (Tgfbr2) in the lung epithelium, Sureshbabu and
fibroblasts but inhibited biglycan and procollagen production coworkers (323) described a pivotal role for the TGF-␤/Tgfbr2
by parenchymal fibroblasts (47). These data also highlight the axis in mediating arrested alveolarization in the background of
idea of compartmentalized responses to therapy in different hyperoxia. To this end, the ability of several pharmacological
cell types. More generally, it has also recently emerged that agents to limit hyperoxia-induced arrest of alveolarization has
administration of dexamethasone to newborn mice over the been attributed to the ability of these agents to blunt hyperoxia-
first 4 days of postnatal life impaired alveolarization, ostensi- induced TGF-␤ signaling, as was the case with curcumin (305)
bly by the suppression of tenascin C and elastin expression and nutritional supplementation with docosahexaenoic acid
(303). Glucocorticoids have also recently been demonstrated to (345) and an activin A receptor type IIB-Fc antagonist (194).
modulate TGF-␤ signaling by redirecting TGF-␤ activity from Other studies have addressed drug-cell interactions in the
the TGF-␤ receptor 1 (Tgfbr1)/smooth muscle actin/mothers context of growth factors; for example, Schwartze and cowork-
against decapentaplegic (Smad)2/3 axis to the activin A recep- ers (312) demonstrated that glucocorticoids modulated TGF-␤
tor type II-like 1 (Acvrl1)/Smad1 axis in lung fibroblasts. This signaling in lung fibroblasts and could shift TGF-␤ signaling
may represent a relevant drug-pathway interaction in lung from the Tgfbr1/Smad2/Smad3 axis to the Acvrl1/Smad1 axis
disease and development (312); however, ECM component and, in doing so, promote lung myofibroblast differentiation.
expression was not addressed in that study. Preliminary evi- Given the widespread use of steroids in the management of
dence has also been provided for some new players in the patients with BPD, these studies reveal a drug-cell interaction
regulation of ECM production and reorganization during of potential clinical significance. The interaction between
lung development, including the serine peptidase chymot- TGF-␤ signaling and glucocorticoids has also been studied by
rypsin-like elastase 1 (197) and cathepsin K (170) and the Collins and coworkers (74) in the preterm lamb model in the
possible interaction between integrins and the ECM (128); context of intrauterine inflammation, in which betamethasone
however, the relevance of these candidates in alveolariza- was documented to blunt TGF-␤ signaling in fetal lungs. The
tion remains to be demonstrated. potential importance of these observations is underscored by
the report of Alanis and coworkers (12), who described that the
Growth Factors boundary separating the two distinct lung compartments, the
proximal conducting airways and the peripheral gas exchange
Along with physical forces (stretch and breathing motions), regions, the bronchoalveolar duct junction, may undergo a
the ECM, and coordinated transcription factor expression, proximal or distal positional shift after premature activation or
growth factors are credited with regulatory roles in alveolar- loss of glucocorticoid signaling. Other modulators of TGF-␤
ization (243). Belcastro and coworkers (32) have added to a signaling in developing lungs have been identified, including
growing wealth of evidence implicating disturbances to growth all-trans-retinoic acid, which dampened aberrantly high
factor dynamics in arrested alveolarization. Using a hyperoxia- TGF-␤ signaling in a caloric restriction model of BPD in rats
based mouse model of BPD (60% O2 for 14 days), the (1, 199). Some recent observations made in early lung devel-
investigators confirmed the increased parenchymal expression opment, such as the control of epithelial differentiation by the
of TGF-␤ in hyperoxia-exposed mouse lungs and further dem- hippo pathway, in which yes-associated protein 1 regulates the
onstrated that in vivo inhibition of TGF-␤ signaling with ability of epithelial progenitor cells to respond to TGF-␤-
SB431542 concomitant with hyperoxia exposure limited prox- induced cues (208), await exploration in a postnatal context.
imal TGF-␤ signaling (as assessed by Smad3 phosphoryla-
tion), and this was accompanied by an increase in secondary Inflammation
crest formation. These investigators further documented that
increased peroxynitrite, thought to be generated by macro- Inflammation is a common complication of BPD, both in a
phages, drove increased TGF-␤ production. Furthermore, the clinical setting and in experimental animal models that use
peroxynitrite caused nitration of IGFR1, thereby preventing inflammation as an injurious stimulus to mimic chorioamnio-
binding of IGF-1 to its cognate receptor and hence limiting nitis (34). The role played by inflammation in limiting lung
downstream IGF-1 signaling, with deleterious consequences development in other BPD models, notably those based on
for alveolarization. In support of a role for IGF-1 in alveolar- hyperoxia and mechanical ventilation, remains relatively un-
ization, Galvis and coworkers (115) described that the repres- explored. The direct (parenteral) attenuation of inflammation
sion of IGF-1 expression by the histone methyl transferase has been undertaken with daily, subcutaneous administration
Ezh2 prevented basal cell differentiation in the developing of an IL-1 receptor antagonist (IL-1Ra) in two slightly different
lung. However, this idea has not yet been assessed in postnatal BPD models (257). Interestingly, in a perinatal inflammation
lung maturation. This study, along with the work of Brechbuhl plus hyperoxia model, in which pregnant dams were first
and coworkers (48), adds the IGF system and the EGF system treated with LPS and then newborn pups were exposed to 85%
to the cocktail of modulators of basal cell differentiation and O2 for 28 days, no impact of IL-1Ra administration on lung
proliferation. development (assessed by calculation of the surface area-to-
The TGF-␤ system, which is activated by exposure to a volume ratio using ImageJ software) was noted. However, in a
hyperoxic environment (18), continues to receive attention as a “gentler” alternative model, in which, instead of 85% O2,
key mediator of alveolarization (359) and has recently been newborn mouse pups were exposed to 65% O2, a dramatic

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00268.2015 • www.ajplung.org


Perspectives
ADVANCES IN LUNG ALVEOLARIZATION AND BPD L1253
improvement in lung structure was noted. Similarly, adminis- Efforts have been made to further delineate the specific
tration of resolvin D1 and lipoxin A4 every third day to inflammatory players that impact lung maturation. Among
C57BL/6 pups exposed to 95% O2 from P1 to P10 improved recent studies, Stouch and coworkers (320) identified I␬B as a
alveolarization (assessed by MLI and RAC), which was asso- mediator of fetal macrophage maturation and suggested that
ciated with reduced proinflammatory cytokine gene expression the maturation of fetal lung macrophages into mature alveolar
(217). These data indicate 1) the potential benefit of attenuating macrophages might include proinflammatory features, hitherto
inflammation on alveolarization in BPD models and 2) that the unrecognized aspects of macrophage activation. Syed and
selection of oxygen levels may dramatically influence the Bhandari (326) have revealed that macrophage polarization
outcome of interventional studies. It is clear that broadly was skewed in favor of an M1 phenotype after hyperoxia
blunting inflammation in BPD models improves alveolariza- exposure (100% O2 for 7 days). These studies support a role for
tion. Future work must address how inflammation modulates macrophage polarization in promoting aberrant alveolarization
alveolarization. (assessed by Lm) in response to hyperoxia and add to a growing
Jagarapu and coworkers (153) evaluated leukadherin-1 body of data defining very diverse roles for macrophages in
(LA1; applied by daily intraperitoneal injection), a novel ago- lung disease (6). This idea was recently elaborated upon by
nist of leucocyte surface integrins, and CD11b/CD18, which Eldredge and coworkers (94), who report existence of an
blunts transendothelial leukocyte migration to sites of injury in anti-inflammatory population of CD11b⫹ mononuclear cells
the hyperoxia-based (85% O2 from P1 to P14) rat model of that are protective against hyperoxia-induced injury to neonatal
BPD. LA1 administration decreased neutrophil and macro- lungs; however, only survival and inflammation were assessed
phage infiltration into the lungs, provoked hyperoxia, improved in that study, without consideration of changes to the lung
alveolarization (assessed by MLI and RAC) and angiogenesis, architecture. A follow-up study is eagerly awaited, particularly
and limited aberrant pulmonary vascular remodeling. These considering the emerging roles of inflammatory cell types and
data clearly demonstrate that inflammation plays a key role in subpopulations in lung development and disease (6, 367), to
aberrant alveolarization in response to hyperoxia. Identical identify causal roles for specific inflammatory cell subsets in
observations were made by Drummond and coworkers (88), lung development. By way of examples, Podolin and cowork-
who applied AMD3100, an antagonist of Cxcr4 in a similar rat ers (282) recently demonstrated that T-cell depletion protects
hyperoxia model (90% O2 from P1 to P16, AMD3100 applied against alveolar destruction attributable to chronic cigarette
between P5 to P15, in a therapeutic regimen). Cxcr4 antago- smoke exposure in mice, but the depletion of T cells has yet to
nism also decreased neutrophil and macrophage infiltration be evaluated in the context of lung development.
into the lungs, provoked hyperoxia, and improved alveolariza- Efforts to modulate regulators of inflammation in experi-
tion (assessed by MLI) and angiogenesis, and limited aberrant mental BPD have also received attention, with McGrath-
pulmonary vascular remodeling. A role for epithelial integrins Morrow and coworkers (230) focusing on Nfe2l2 (also called
in macrophage recruitment has already been described earlier Nrf2), which was formerly demonstrated to be protective in
in this Perspectives article, relating the work of Plosa and hyperoxia-induced arrest of alveolarization (assessed by Lm).
coworkers (281) using an epithelial-specific (Sftpc-driven) Interestingly, using sulforaphane, an inducer of Nfe2l2, the
deletion of integrin-␤1, which promoted macrophage recruit- investigators demonstrated that, although inflammation could
ment and blunted alveolarization (assessed by MLI) in mice, be attenuated by Nfe2l2 induction, alveolar growth was not
ostensibly through increased proinflammatory cytokine (Ccl2 improved, possibly attributable to proliferation-suppressive ef-
and Cxcl1) and ROS production by lung epithelial cells. These fects of Nfe2l2. This study makes the important point that
data highlight the regulatory cross talk between the epithelium caution should be exercised when anti-inflammatory strategies
and inflammatory cells during alveolarization and add to a may have the side effect of growth inhibition during a critical
growing body of data highlighting the connection between phase of rapid postnatal growth and highlights the need for
inflammatory signaling and ROS production (130). Further additional studies both on Nfe2l2 and other molecules that
studies by Carver and coworkers (58) have linked innate protect against oxidative stress, such as Sirt1 (376). An inter-
immune signaling and regulatory developmental programs in esting angle on the Nfe2l2 line has been developed in adult
the lung, in which a link between NF-␬B signaling and the mice by Kawamura and coworkers (163), who reported that the
expression of FGF-10, a regulator of lung development, was anti-inflammatory effects of hydrogen gas [2% (vol/vol)] in the
demonstrated. The authors suggested that NF-␬B-driven in- background of hyperoxia (98% for 60 h) were Nfe2l2 depen-
flammatory pathways may suppress the expression of growth dent. The use of hydrogen gas to limit hyperoxia-induced
factors and other mediators of alveolarization, which would damage in neonates has not yet been reported. Another regu-
negatively impact lung structure. These data take on increased lator of inflammation that has received recent attention is
importance in light of the documented regulation of lung macrophage migration inhibitory factor (MIF) (321, 322), in
epithelial stem cell growth by TGF-␤ and FGF-10 (93, 233). which it was demonstrated that both too much (overexpression
Interestingly, in contrast to the ideas of Carver and coworkers in transgenic mice) and too little (MIF knockout mice) MIF
(58), Hou and coworkers (144), using BAY 11–7082, which is arrested alveolarization (assessed by Lm) in both room air-
an inhibitor of NF-␬B signaling, have indicated that it may be exposed and hyperoxia-exposed mice. Thus the correct balance
desirable to promote some elements of NF-␬B inflammatory of MIF levels in the developing lung was essential for proper
signaling to suppress damaging chemokine (C-X-C motif) lung development. Administration of a small molecular MIF
ligand 2 (Cxcl2) [also called macrophage inflammatory protein agonist to wild-type mice in the hyperoxia BPD model partially
(MIP)-2] production. Clearly much remains to be learned about restored normal lung alveolarization, further supporting the
the contribution of NF-␬B signaling to postnatal lung matura- idea that modulating inflammation represents a viable approach
tion. for the restoration of lung development in BPD.

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00268.2015 • www.ajplung.org


Perspectives
L1254 ADVANCES IN LUNG ALVEOLARIZATION AND BPD

In addition to a role for inflammation in modulating alveo- pulmonary vascular development in BPD, CDH, and ACD,
larization, innate immunity has also been addressed in the including ACDMPV.
context of BPD, in which where Raffay and coworkers (288) Further advances have also been made in our understanding
demonstrated that hyperoxia exposure (85% O2 for 14 days, of the molecular mechanisms at play that drive PPHN. Among
with a posthyperoxia recovery phase) influenced club (for- these, Delaney and coworkers (85) reported that serotonin
merly Clara) cell function, evident by decreased secretoglobin, (5-hydroxytryptamine) contributed to elevated pulmonary vas-
family 3A, member 1 (Scgb3a1) and Scgb1a1 protein expres- cular resistance in an experimental sheep model of PPHN via
sion, which would have implications for the innate immune the 5-HT2A receptor. The angiotensin system has also received
response. These studies may indicate a mechanism by which attention in the context of postnatal lung development, in
infants with BPD are more susceptible to lung infections. which Castro and coworkers (59) describe the downregulation
Along the lines of this study, Cai and coworkers (54) demon- of angiotensin-converting enzyme (ACE) expression in the
strated that administration of a recombinant-related secretoglo- lungs of patients with BPD, suggesting a pathophysiological
bin, secretoglobin, family 3A, member 2 (Scgb3a1), to preg- contribution of diminished ACE levels to the development of
nant mice promoted the development of terminal air sacs in clinical BPD. This idea is supported by Wagenaar and cowork-
preterm (E17.5) pups (as assessed by RAC). Together, these ers (352), who reported that stimulation of the MAS oncogene
data might suggest that the decreased expression of secreto- with cyclic (Ang)1–7 or stimulation of the type II angiotensin
globin in the hyperoxia model observed by Raffay and col- receptor with [D-lysine]cyclic(Ang)1–7 in the rat hyperoxia
leagues may have also directly negatively impacted terminal model (100% O2 for 10 days, from P1) partially protected
air-sac development or alveolarization, in addition to limiting against hyperoxia-induced damage to alveolarization, with an
innate immune responses. increased number of alveolar crests and decreased septal
wall thickness observed. Additionally, the arteriolar medial
wall thickness and Fulton index were partially normalized.
Pulmonary Vascular Development and PPHN Collectively, these observations support a role for dysregu-
Pulmonary vascular development and vascular endothelial lation of the angiotensin system in clinical and experimental
and smooth muscle cell function are perturbed in a spectrum of BPD. In another study, Nijmeh and coworkers (256) re-
lung developmental abnormalities, including BPD (21, 245, vealed the existence of high proliferative potential colony-
319), CDH (3), ACD (172, 302), and persistent pulmonary forming cells (HPP-CFC) in the adventitial vaso vasorum of
hypertension of the newborn (PPHN) (84, 183, 237). Under- the pulmonary arteries in a hypoxia-induced PPHN model in
standing the molecular regulation of pulmonary vascular de- calves. These authors proposed that, given the high prolif-
velopment remains a major challenge (273). The past 2 yr have erative potential and vasculogenic capacity of HPP-CFC,
seen the identification of several new mediators of pulmonary these cells may be pathogenic mediators and perhaps targets
vascular development, including Wntless (75) and Sox17 for the management of pulmonary vascular remodeling
(184), which play a role in embryonic vessel development, but associated with hypoxia exposure in neonates.
Several lines of evidence point to PPAR-␥ as a key contrib-
a function for either mediator in normal or aberrant late lung
utor to perturbed vascular development and homeostasis in
development is not yet clear. In contrast, the semaphorin/
PPHN. Wolf and colleagues (370) demonstrated that increased
neuropilin 1 axis was demonstrated to play a role in fetal, but
levels of endothelin (ET)-1, which are indeed observed in
not postnatal, pulmonary vessel development (160), and pig-
clinical PPHN, decreased PPAR-␥ signaling and blocked pul-
ment epithelium-derived factor appeared to be involved in
monary artery endothelial cell tube formation. PPAR-␥ ago-
perturbed vascular development associated with hyperoxia nists promoted endothelial tube formation in pulmonary artery
(69). Additionally, antagonism of PDE5 with sildenafil pro- endothelial cells in an NO-dependent manner. These observa-
moted pulmonary vascular development and alveolarization, tions suggest that targeting ET-1 production may restore an-
ostensibly via activation of the phosphatidylinositol-4,5-bis- giogenesis in PPHN. In another study, Wagenaar and cowork-
phosphate 3-kinase/Akt/mammalian target of rapamycin path- ers (351) also addressed the ET-1 system using ambrisentan
way and enhanced HIF-1␣ and HIF-2␣ activity (270). Other (an endothelin receptor type A antagonist) in the rat hyperoxia
candidate regulators of lung vessel cell behavior have also been model (100% O2 from P1 to P10), which improved survival,
identified in vitro, including heat-shock protein 70 (5), the promoted alveolar development by normalizing septal wall
oxygen-sensitive voltage-gated potassium channel Kv1.5 thickness (without impacting the number of alveolar crests per
(202), and AMPK (330); however, a role for these molecules in field), limited excessive ECM deposition, and dramatically
pulmonary vascular development awaits clarification. improved aberrant remodeling of vessel walls and the heart that
Along with reports that the transcription factor forkhead box were provoked by hyperoxia exposure. Interestingly, these
(Fox)M1 regulates pulmonary inflammatory responses to hy- positive effects were not seen in a late treatment protocol (pups
peroxia (371), FoxF1 has also been recently implicated as a were exposed to 100% O2 for 9 days, followed by 9 days of
candidate regulator of aberrant pulmonary vascular development recovery in room air, with daily ambrisentan application initi-
in a spectrum of congenital diseases. FoxF1 haploinsufficiency is ated on day 6). A potential connection between the HIF-1␣ and
characteristic of ACDMPV. Therefore, a transcriptional profiling ET-1, as demonstrated by Larissa Shimoda’s group in smooth
study of lung from Foxf1⫹/⫺ mice was undertaken by Sen and muscle cells in adult rats (280), has not yet been explored in the
coworkers (313), who reported that FoxF1 regulated the expres- context of PPHN.
sion of a spectrum of genes, including multiple collagens and Returning to PPAR-␥, Gien and coworkers (118) revealed
members of the IGF pathway. These data contribute to our an interaction between PPAR-␥ and Rho kinase (ROCK) that
further understanding of the mechanisms underlying disturbed increased pulmonary artery smooth muscle cell proliferation

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00268.2015 • www.ajplung.org


Perspectives
ADVANCES IN LUNG ALVEOLARIZATION AND BPD L1255
and proposed that this interaction was relevant to the aberrant cued both vascularization and vascular growth. Taking this
vascular remodeling seen in PPHN. These data are very inter- idea further, Morales and coworkers (240) nebulized the
esting considering the observations of Lee and coworkers PPAR-␥ agonists rosiglitazone and pioglitazone into the lungs
(186), who reported that inhibition of ROCK with Y-27632, of 1-day-old rat pups, which were subsequently exposed to
not only attenuated the deleterious impact of bleomycin in hyperoxia (95% O2 for 3 days, intervention once daily). Neb-
alveolarization in rat pups, but also normalized pulmonary ulized PPAR-␥ agonists protected against hyperoxia-induced
vascular resistance, decreased right-ventricular hypertrophy, retardation of alveolarization (assessed by MLI and RAC) and
and attenuated vascular remodeling. Thus PPAR-␥ and ROCK blunted hyperoxia-provoked lung inflammation. Although not
are exciting candidate targets in the management of pulmonary assessed in pulmonary vascular development, in the context of
hypertension associated with BPD, as well as PPHN; however, sickle cell disease-associated pulmonary hypertension, Li and
the demonstration that some elements of pulmonary artery coworkers (191) described the downregulation of ET-1 by
contraction are likely Rho independent (98) suggests that other fenofibrate-stimulated PPAR-␥, which upregulated miR-
mediators are also involved. It is worth noting at this junction 199a2, which in turn targeted ET-1 expression. This pathway
that stimulation of PPAR-␥ signaling can rescue alveolariza- may also be relevant to the ET-1/PPAR-␥ axis in the context of
tion and vessel development in the hyperoxia model, in which vascular development as well. Thus the PPAR-␥ system rep-
Lee and coworkers (187) applied Escherichia coli LPS to the resents an exciting druggable target to drive lung alveolar and
amniotic sac of pregnant mice. After spontaneous delivery, vascular development. These ideas are integrated into a scheme
pups were exposed to hyperoxia (80% O2 from P1 to P7) in Fig. 4. Apart from PPAR-␥/ET-1, AMPK has also recently
followed by recovery in room air (P8 to P14), which blunted been introduced as a candidate target to drive angiogenesis in
both alveolarization (assessed by Lm) and vessel density (as- utero (330).
sessed by visual inspection and PECAM immunohistochemis- In an effort to delineate the impact of congenital heart
try). Administration of the PPAR-␥ agonist rosiglitazone res- defects on lung development, Razavi and coworkers (296)

Normalization of lung development

Proper cascular development and alveolarization

Normalized proliferation Enhanced tube formation

Decreased Increased Fig. 4. Recently identified roles for peroxi-


some proliferator-activated receptor-␥
ROCK eNOS
(PPAR-␥) in pulmonary vascular develop-
activity activity
ment and homeostasis associated with persis-
Rosiglitazone tent pulmonary hypertension of the newborn
Increased
Pioglitazone ACTIVATION (PPHN). Modulation of PPAR-␥ is poten-
Fenofibrate miR-199a2 tially relevant to pulmonary vascular devel-
expression opment and homeostasis in bronchopulmo-
SMC EC
nary dysplasia (BPD) and PPHN. PPAR-␥
can be modulated by activators (green) or by
PPARγγ the suppression of inhibitory pathways (red)
by attenuating the effects of endothelin
SMC EC (ET)-1 mediated by the endothelin 1A
(AT1A) and 1B (ET1B) receptors. The effects
ET1A of PPAR-␥ agonism may impact smooth
INHIBITION ET1B
ET-1 muscle cells (SMC) and endothelial cells
(EC) by modulating endothelial nitric oxide
Increased Decreased synthase (eNOS) or Rho kinase (ROCK) ac-
ROCK eNOS tivity. The scheme only details new develop-
ments that were published between 1 January
activity activity
Ambrisentan 2013 and 30 June 2015.
Bosentan

Hyperproliferation Impaired tube formation

Increased vascular remodeling and decreased angiogenesis

PPHN/BPD

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00268.2015 • www.ajplung.org


Perspectives
L1256 ADVANCES IN LUNG ALVEOLARIZATION AND BPD

employed a model of pulmonary artery stenosis in rats. After called Nrf2) in the airways as a mediator of oxidant damage
the left pulmonary artery was banded at 21 days of life, lung responses in the airways (70). The recently described role of
structure was assessed at 160 days. Left pulmonary artery the transcription factors Spdef and FoxA3 in goblet cell meta-
banding resulted in an apparent decrease in alveolar size plasia and Th2-mediated inflammatory responses in the post-
compared with sham-operated animals, suggesting that dis- natal lung (290) provides impetus for such investigations. A
turbed blood flow to the lung can limit alveolarization. It must role for the pulmonary neuroendocrine cells in aberrant lung
be kept in mind that arterial banding study was performed after alveolarization also awaits attention.
the phase of bulk alveolarization but before the continued Some studies have started to address the impact of hyperoxia
alveolarization described by Tschanz and coworkers (341) that on small airway structure. Wang and coworkers (355) de-
occurs between P21 and P60. As such, it remains unclear scribed the differential impact of elevated oxygen levels on
whether the apparently reduced alveolar size results from a smooth muscle mass and collagen deposition in the developing
failure of late alveolar development or from alveolar destruc- airways in mice exposed to 40% O2 or 70% O2 from P1 to P7,
tion in response to blood flow limitation. followed by recovery in room air between P8 and P21. Other
The role of perturbed ECM deposition and remodeling, reports, for example those documenting the impact of cigarette
which result in vessel stiffness, is an exciting and new area of smoke on mitochondrial function (24, 25) and the proliferation
research in the pulmonary vasculature. Recent work by Nave and ECM production (347) in airway smooth muscle cells, as
and coworkers (253) described aberrant collagen and elastin well as the impact of hyperoxia on estradiol metabolism in
cross linking in pulmonary vessels in pulmonary arterial hy- postnatal airway smooth muscle (219), highlight the potential
pertension in adult mice. Additionally, vascular stiffening is relevance of perturbations to airway development to lung
presently being debated as a cause or effect of pulmonary maturation. Furthermore, Chen and colleagues (67) provide
hypertension in human adults (328). Along these lines, Dodson evidence that endogenous retinoic acid, a key player in lung
and coworkers (87) reported that the main pulmonary arteries development, restricts airway smooth muscle cell differentia-
in lambs with PPHN exhibited increased stiffness and exces- tion during formation of the airways. Similarly, Xie and co-
sive ECM remodeling. These exciting data point to hitherto workers (372) connected Wnt/␤-catenin and Sox2, two other
unrecognized roles for perturbations to vessel stiffness in key players in lung development already discussed in this
proximal (as opposed to distal) pulmonary vessels as a patho- Perspectives article, in airway submucosal gland morphogen-
genic factor in PPHN and highlight the examination of ECM esis. Clearly, much interesting work in this direction lies ahead.
deposition and the activity of ECM cross-linking enzymes as a
worthwhile avenue to pursue in understanding the mechanistic Long-Term Sequelae of Hyperoxia Exposure
basis of PPHN.
There is currently much interest in the long-term follow-up
A Role for the Small Airways? of patients with BPD to assess the implications of having had
BPD on morbidity in later life (137, 263). One recent report
The development of the alveoli and the pulmonary vascular suggested that early classification of BPD severity in preterm
tree has received much attention in the context of late lung infants did not provide useful information about future lung
development. However, despite the pioneering reports in the function, when patient lung function was assessed at 6 and 18
early 1990s by Marc Hershenson and Julian Solway demon- mo after preterm birth (334); however, additional studies are
strating that exposure to hyperoxia in the immediate postnatal clearly indicated. Similar studies are also now being conducted
period causes airway hyperresponsiveness and aberrant airway in animal models of BPD, in which the time course to adult-
remodeling (135, 136) and studies by Shenberger and col- hood is relatively shorter. Some notable studies have revealed
leagues (315) that demonstrated pulmonary neuroendocrine that perturbed late lung development, for example after hyper-
cell hyperplasia in developing rats in response to hyperoxia, oxia exposure in the postnatal period, does indeed have con-
the small airway compartment has been very neglected. Given sequences for the lung architecture in adult animals. O’Reilly
the intimate relationship that must exist between the develop- and coworkers (262) demonstrated that exposure of mouse
ing airways and the associated parenchyma and vessels (53), pups to 65% O2 over the first 7 days of life resulted in
together with many recent advances in airway smooth muscle remodeling of the bronchiolar walls and loss of bronchiolar-
cell biology (286), further investigations into airway biology alveolar attachments that was evident at 10 mo of age, leading
during normal and aberrant late lung development are war- the investigators to propose that immediate postnatal hyperoxia
ranted. This idea is supported by the predisposition of preterm exposure could lead to impaired lung function and airway
neonates to clinical problems related to the airways, such as hyperreactivity in adulthood. In a related study, these investi-
wheeze and asthma (50), and the recent exciting observation gators demonstrated that the exposure to hyperoxia caused
from Wolfgang Kübler’s group that upper airway distension persistent alteration to the bronchiolar wall that contributed to
triggers proinflammatory responses in mechanically ventilated perturbed lung function that had a stronger impact on the male
adult C57BL/6 mice (255). sex (261). These data thus also contribute to current thinking
Some recent reports have addressed the role of postnatal that sex plays a key role in lung responses to hyperoxic injury.
hyperoxia exposure on bronchiolar remodeling that is evident These observations are supported by a report from Ramani and
in adult experimental animals (261–263, 293); however, a coworkers (293), in which exposure of C57BL/6 mouse pups
comprehensive assessment of small airway architecture in to either hyperoxia (85% O2) or normoxia (12% O2) over the
neonates has not been undertaken. This appears all the more period P2-P14, followed by a 10-wk recovery period under
important given the recent summary by Hye-Youn Cho and normoxic (21% O2) conditions, caused stunted alveolar devel-
Steven Kleeberger highlighting the perspective of Nfe2l2 (also opment (assessed by MLI and RAC), loss of bronchiolar-

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00268.2015 • www.ajplung.org


Perspectives
ADVANCES IN LUNG ALVEOLARIZATION AND BPD L1257
alveolar attachments, increased lung compliance, and, in the diaphragmatic contraction as a controlling factor in compen-
case of the hyperoxia group, increased arteriole medial wall satory lung growth. On the vascular side, Ackermann and
thickening that persisted into adulthood. An important struc- coworkers (4) have used remarkable imaging approaches to
ture-function correlate has also been reported by Ahlfeld and demonstrate sprouting and intussusceptive angiogenesis as
coworkers (8), in which mouse pups were exposed to hyper- important mechanisms of lung alveolarization during post-
oxia (90% O2 from P1 to P7) followed by recovery in room air pneumonectomy lung growth. A key mechanistic finding
from P8 to P56. At P56, an alveolarization (assessed by MLI) was provided by Rafii and coworkers (289), who docu-
and vessel density defect persisted. Furthermore, the alveolar- mented that platelets drive postpneumonectomy lung regen-
ization defect corresponded with a reduced functional gas eration by supplying stromal cell-derived factor (SDF)-1,
exchange (measured by the diffusing factor for carbon mon- which activates the SDF-1 receptors Cxcr4 and Cxcr7 on
oxide, DFCO). This report also highlights the usefulness of capillary endothelial cells, which in turn stimulate alveolar
combining structural analyses with physiological measures of epithelial cell expansion, via the angiocrine membrane-type
lung function, which together provide complementary views metalloproteinase. This idea also hints at worthwhile ave-
on the same idea, contributing to the robustness of the argu- nues to pursue for the stimulation of lung maturation in
ment. To this end, the measurement of pressure-volume curves neonates.
in mouse lungs has recently been considered in detail (195).
Experimental studies on susceptibility to viral infection in Pharmacological Interventions to Identify New Pathogenic
later life, after exposure to hyperoxia in the neonatal period Pathways
have also been undertaken by Maduekwe and coworkers (204),
who demonstrated that exposure to a particular threshold of Several interventional pharmacological studies have identi-
hyperoxia in the neonatal period altered host susceptibility to fied candidate pathways or molecules that might be targeted in
influenza virus infection in the long term. Taking this idea a translational sense to promote better lung growth or to
further, Buczynski and coworkers (51) explored a protective identify new pathways relevant to normal or aberrant late lung
role for SOD during hyperoxia exposure of mouse pups in the development. These targets are summarized in Table 1 and
immediate postnatal period (100% O2 between P1 and P4) on have identified possible lead compounds for further develop-
alveolar development quantified in late life (8 –10 wk). Over- ment, with the aim to drive lung maturation by pharmacolog-
expression of SOD in alveolar type II cells during hyperoxia ical interventions. Additionally, these studies have highlighted
exposure conferred protection against hyperoxia-induced dam- new pathways relevant to normal and aberrant alveolarization,
age to lung structure, with a partial normalization of alveolar which warrant further study.
development reported, although this was assessed by visual One area of current intensive investigation is the impact of
inspection of lung sections, in which lung structure was not caffeine and other xanthine derivatives on lung maturation
quantified. Exposure of these mice, as adults, to influenza A (218), highlighted in Fig. 5. Weichelt and colleagues (362)
virus indicated that wild-type mice with postnatal hyperoxia administered caffeine [10 mg/kg per day, ip; from P6, concom-
exposure were more sensitive to influenza A virus infection, itant with the onset of hyperoxia (80% O2 for up to 48 h)
exhibiting increased mortality and increased lung collagen exposure] to rat pups and observed an apparent improvement in
production, which was prevented in transgenic mice overex- alveolarization by visual inspection of sections (lung structure
pressing SOD in alveolar type II cells. In sum, these observa- was not quantified). Additionally, blunted neutrophil and mac-
tions support the idea that immediate postnatal exposure pre- rophage infiltration and a normalization of Cxcl1, Cxcl2 (also
disposes adults to respiratory viral infection. called MIP-2), Ccl2, TNF-␣, and IL-6 levels were observed in
the caffeine-treated group in the background of hyperoxia. In
Neoalveolarization another study, Dayanim and colleagues (83) administered caf-
feine (20 mg/kg on P1, followed by 10 mg/kg per day there-
Postpneumonectomy lung growth is an increasingly used after) to FVB/N mouse pups in a dosing regimen directly
model to study lung development in which mice regenerate translated from clinical studies (272, 308). In the latter study,
lung tissue (neoalveolarization) after surgical resection (pneu- caffeine administration has only deleterious effects, with in-
monectomy) (332). There is currently controversy about the creased pulmonary inflammation and increased epithelial apo-
molecular pathways that drive neoalveolarization. To date, ptosis, as well as worsening of the lung architecture (as
lung regeneration after pneumonectomy has been attributed to assessed by RAC) in the background of hyperoxia. Clearly,
reinitiation or acceleration of cellular growth, with the proposal these two studies yield conflicting results, which may be
that, analogous to limb regeneration, neoalveolarization would attributable to a lack of dose range finding, as acknowledged
result from reactivation of lung organogenesis genes (145, by Dayanim and colleagues in their report (83). Interestingly,
360). In a transcriptional profiling study, Kho and colleagues both studies reported that caffeine applied under room-air
(164) make the observation that, rather than the engagement of conditions dysregulated Cxcl1 expression, perhaps suggesting
early lung organogenesis pathways, postpneumonectomy lung a proinflammatory activity of caffeine in the lungs under room
growth was characterized by the engagement of pathways air conditions. Comparing these studies is difficult because of
driving later stages of lung development (alveolarization) the different animal models and oxygen exposure protocols,
rather than pathways that direct branching and primary airway and the impact of caffeine on alveolar architecture in the
enlargement. In addition to transcriptional control of postpneu- Weichelt study (362) remains to be validated because these
monectomy lung growth, Ysasi and coworkers (380), using a investigators did not quantify changes in lung structure, which
unilateral diaphragmatic paralysis approach, have also demon- were observed by visual inspection only. Clearly, the potential
strated the critical importance of cyclic stretch associated with utility of caffeine to manage lung inflammation and promote

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00268.2015 • www.ajplung.org


Perspectives
L1258 ADVANCES IN LUNG ALVEOLARIZATION AND BPD

Table 1. Pharmacological interventions in animal models of bronchopulmonary dysplasia and associated persistent
pulmonary hypertension of the newborn undertaken since 1 January 2013
Animal Model

Drug (dose and frequency) Species and Strain Injury Stimulus Molecular/Pathway Targets Lung Pathological Findings Reference
ActRIIB-Fc (5 mg/kg tiw, ip) Mouse C57BL/6 85% O2 P1-P14 Activin A receptor antagonist Improved alveolarization (194)
Blunted PASMC proliferation
Blunted TGF-␤ signaling
Ambrisentan (1-20 mg/kg daily, sc) Rat S/ND 100% O2 P1-P10 ET receptor type A antagonist Improved septal thickness (351)
Suppressed inflammation Improved vascular
structure
AMD3100 (240 ␮g/kg daily, sc) Rat S-D 90% O2 P2-P16 Cxcr4 antagonist Suppressed Improved alveolarization (88)
inflammation Improved vascular
structure
Ascorbylperoxide (infused) Guinea pig Drug application Increased oxidative stress Destruction of alveoli (95)
Hartley
Aspirin (40 mg/kg daily, R/ND) Mouse C3H/HeN 85% O2 P1-P14 Cyclooxygenase-2 inhibitor No improvement in (50)
Suppressed inflammation alveolarization
ATRA (5 mg/kg daily, ip) Rat S-D Nitrofen E9.5 Retinoic acid (primary) Increased Improved alveolarization (109)
leptin signaling
BAPN (15 mg/kg daily, ip) Mouse C57BL/6 85% O2 P1-P10/P20 Lysyl oxidase inhibitor Altered Worsened alveolarization (238)
ECM morphology
BAPN (dose indeterminate) Mouse C57BL/6 85% O2 P1-P10 Lysyl oxidase inhibitor Altered Improved alveolarization (212)
ECM morphology
BAY 11-7082 (10 mg/kg once, ip) Mouse C57BL/6 LPS NF-␬B pathway inhibitor Worsened alveolarization (144)
Increased MIP-2 levels Worsened vascular
structure
Benzo[a]pyrene (25 mg/kg, ip, Rat Fisher 344 90% O2 P1-P14 Increased oxidative stress Worsened alveolarization (331)
E18/19/20) Increased levels of isoprostanes Increased inflammation
Caffeine (10 mg/kg daily, ip) Mouse FVB/N 80% O2 P1-P8 Increased inflammation No improvement in (83)
alveolarization
Caffeine (10 mg/kg, once at P6) Rat Wistar 80% O2 P6-P8 Suppressed inflammation Improved alveolarization (362)
(apparent, not
quantified)
Carbon monoxide (250 ppm, Mouse C57BL/6 75% O2 P3-P21 Suppressed inflammation Improved alveolarization (23)
continuous)
Celecoxib (5 mg/kg daily, R/ND) Mouse C3H/HeN 85% O2 P1-P14 Cyclooxygenase-2 inhibitor No improvement in (50)
Suppressed inflammation alveolarization
Curcumin (5 mg/kg daily, ip) Rat S-D 95% O2 P1-P5 Attenuated TGF-␤ signaling Improved alveolarization (305)
Blunted ECM deposition Improved septal
thickness
cyclic (Ang)1–7 (10–50 ␮g/kg daily, sc) Rat Wistar 100% O2 P1-P10 MAS oncogene receptor agonist Improved septal thickness (352)
Suppressed inflammation Improved vascular
structure
Cyclosporine (15 mg/kg daily, sc) Rat S-D 60% O2 P1-P14 Not reported No improvement in (285)
alveolarization
DFU (10 ␮g/g daily, ip) Rat S-D 60% O2 P1-P14 Cox2 inhibitor Suppressed Improved alveolarization (220)
inflammation
DHA (diet supplement) Mouse C3H/HeN LPS ⫹ 85% O2 P1-P14 Suppressed inflammation Improved alveolarization (345)
GYY4137 (50 mg/kg daily, ip) Mouse C57Bl/6 85% O2 P1-P10 H2S donor Suppressed Improved alveolarization (207)
inflammation Improved septal
thickness
GYY4137 (37 mg/kg daily, ip) Rat S-D 90% O2 P1-P14 H2S donor Blunted PASMC Improved alveolarization (343)
proliferation Improved vascular
structure
5-HT (12–20 ␮g infused) Sheep C-R Fetal d.a.constriction 5-HT2A receptor agonist Increased PVR (85)
ICG001 (10 mg/kg daily, ip) Rat S-D 90% O2 P2-P14 ␤-catenin inhibitor Blunted Improved alveolarization (13)
PASMC proliferation Blunted Improved vascular
ECM production structure
IL-1Ra (10 mg/kg daily, sc) Mouse C57BL/6 LPS ⫹ 65% or 85% O2 (to P28) IL-1 receptor antagonist Improved alveolarization (257)
Suppressed inflammation
iNOS (5 ppm E21-P7) Rat S-D 80% O2 P1-P7 Increased neurotrophin expression Improved alveolarization (276)
Improved vascular
structure
Ketanserin (20 mg infused) Sheep C-R Fetal d.a.constriction 5-HT2A receptor antagonist Decreased PVR (85)
␣-Klotho (60 pmol daily, ip) Rat S-D 90% O2 ⱕ3 days Not reported Reduced edema (295)
LA-1 (1 mg/kg b.i.d., ip) Rat S-D 85% O2 P1-P14 CD11b/CD18 agonist Suppressed Improved alveolarization (153)
inflammation Improved vascular
structure
Lipoxin A4 (2 ng/g, ip, on P1/3/6/9) Mouse C57BL/6 100% O2 P1-P10 Suppressed inflammation Improved alveolarization (217)
Normalized septal
thickness
Liver growth factor (1.7 ␮g biw, ip) Mouse AKR/J Chronic smoke exposure Suppressed inflammation Blunted No improvement in (275)
MMP activity alveolarization
[D-lysine]cyclic(Ang)1–7 (5–20 ␮g/kg Rat Wistar 100% O2 P1-P10 ATII type 2 receptor agonist Improved septal thickness (352)
daily, sc) Suppressed inflammation Improved vascular
structure
Mesd (10 mg/kg, ip, q.a.d.) Rat S-D 90% O2 P1-P14 Lrp5/6 inhibitor Attenuated Wnt/ Improved vascular structure (14)
␤-catenin signaling
Metformin (25-100 mg/kg daily, sc) Rat Wistar 100% O2 P1-P10 Suppressed inflammation Improved septal thickness (68)
Suppressed fibrin deposition Improved vascular
structure
MIF020 (0.4 mg/g daily, P1-P4, id, Mouse C57BL/6 100% O2 P1-P4 MIF agonist Inflammation Improved alveolarization (321)
P5-P14, ip) unaffected
MIF098 (0.4 mg/g daily, P1-P4, id, Mouse C57BL/6 100% O2 P1-P4 MIF antagonist Suppressed No improvement in (321)
P5-P14, ip) inflammation alveolarization
Continued

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00268.2015 • www.ajplung.org


Perspectives
ADVANCES IN LUNG ALVEOLARIZATION AND BPD L1259
Table 1.—Continued
Animal Model

Drug (dose and frequency) Species and Strain Injury Stimulus Molecular/Pathway Targets Lung Pathological Findings Reference
mitoTEMPO (0.7 ␮g/g daily, sc) Mouse C57BL/6 75% O2 72 h at P0 or P4 Not reported Improved alveolarization (81)
Improved vascular
structure
MnTBAP (1-10 mg/kg, ip, P1/2/3) Rat S-D 50% O2 Inter. 12% O2 P1-P14 Superoxide dismutase mimetic Not assessed (62)
Modulated VEGF levels
Blunted MMP-9 levels
PD123319 (0.5-2.0 mg/kg daily, ip) Rat Wistar 100% O2 P1-P10 ATII type 2 receptor antagonist Improved septal thickness (353)
Suppressed inflammation Improved vascular
Blunted ECM deposition structure
Pioglitazone (3 mg/kg, nebulized) Rat S-D 95% O2 P1-P3 PPAR-␥ agonist Improved alveolarization (240)
Resolvin D1 (2 ng/g, ip, P1/3/6/9) Mouse C57BL/6 100% O2 P1-P10 Suppressed inflammation Normalized septal (217)
thickness
Rosiglitazone (3 mg/kg daily, ip) Rat S-D LPS ⫹ 80% O2 P1-P7 PPAR-␥ agonist Improved alveolarization (187)
Improved vascular
structure
Rosiglitazone (3 mg/kg, nebulized) Rat S-D 95% O2 P1-P3 PPAR-␥ agonist Improved alveolarization (240)
SB216763 (10 mg/kg daily, ip) Rat S-D 90% O2 P2-P15 Inhibited GSK3␤ signaling Improved alveolarization (149)
Suppressed inflammation Blunted PH
SB265610 (4 mg/kg daily, ip) Rat S-D Bleomycin Cxcr2 antagonist Suppressed No improvement in (186)
inflammation alveolarization
Sertraline (10 mg infused) Sheep C-R Fetal d.a. constriction Serotonin reuptake inhibitor Increased PVR (85)
Sildenafil (100 ␮g/g daily, ip) Ra S/ND LPS ⫾ 85% O2 P2-P14 Phosphodiesterase 5 (primary) Improved alveolarization (270)
Increased PI3K/Akt/mTOR
axis
D-Sphingosine (1.25 ␮g/g daily, ip) Mouse C57BL/6 80% O2 P1-P28 Inhibited ceramide production Improved alveolarization (336)
Stem cell factor (100 ␮g/kg daily, sc) Rat S-D 90% O2 P2-P15 c-kit agonist Improved alveolarization (236)
Improved vascular
structure
VARA (oral on alternate days) Mouse C57BL/6 85% O2 P1-P14 Retinoic acid pathway (primary) Improved alveolarization (156)
Attenuated oxidative stress
Vitamin D Rat S-D Endotoxin at E20 (intra amniotic) Vitamin D pathway (primary) Improved alveolarization (213)
Increased VEGF/VEGFR axis Improved vascular
structure
Y-27632 (10 mg/kg daily, ip) Rat S-D Bleomycin ROCK inhibitor Suppressed Improved alveolarization (186)
inflammation

ActRIIB-Fc, activin A receptor type IIB-Fc antagonist; Akt, thymoma viral proto-oncogene 1; ATII, angiotensin II; BAPN, ␤-aminopropionitrile; b.i.d., bis
in die (twice daily); biw. biweekly (twice weekly); C-R, Colombia-Rambouillet; DHA, docosahexaenoic acid; Cox2, cycloxygenase-2; d.a., ductus arteriosis;
DFU, 5,5-dimethyl-3-(3-fluorophenyl)4-(4-methylsulfonyl)phenyl-2(5H)-furanone; E, embryonic day; ECM, extracellular matrix; ET, endothelin; GYY4137,
[morpholin-4-ium-4-methoxyphenyl(morpholino) phosphinodithioate]; IL-Ra, interleukin-1 receptor antagonist; iNOS, inducible nitric oxide synthase; Inter.,
intermittent; LA-1, leukadherin-1; LRP5/6, low-density lipoprotein receptor–related proteins 5 and 6; MIF, macrophage migration inhibitory factor; MIF020,
4-[4-(pyridin-3-yl)-1H-1,2,3-triazol-1-yl]phenol; MIF098, 3-(3-hydroxybenzyl)-5-methylbenzooxazol-2-one; MIP-2, macrophage inflammatory protein 2; MMP,
matrix metalloproteinase; mTOR, mammalian target of rapamycin; P, postnatal day; PASMC, pulmonary artery smooth muscle cell; PI3K, phosphatidylinositol-
4,5-bisphosphate 3-kinase; PPAR, peroxisome proliferator-activated receptor; ppm, parts per million; PVR, pulmonary vascular resistance; q.a.d., quaque alternis
die (every other day); R/ND, route not declared; ROCK, Rho kinase; S-D, Sprague-Dawley; S/ND, strain not declared; TGF, transforming growth factor; tiw.
thrice weekly; VARA, vitamin A and retinoic acid.

lung maturation is exciting, but reports on caffeine toxicity compliance and airway resistance. These studies continue to
raised concerns about dose (338), and the observed impact of add ideas to the ongoing debate about the use of and role of
early vs. late administration of caffeine to infants (272) high- retinoids in lung development and lung structural homeostasis
lights the need for optimization of treatment protocols. Some that started in 1927 with Harry Goldblatt and Maria Benis-
recent evidence suggests that caffeine may also exert an effect chek’s original observations that vitamin A deficiency causes
on TGF-␤-driven epithelial-to-mesenchymal transition of lung lung metaplasia (119). It appears that we have some way to go
epithelial cells (100) and may influence steroid-mediated sur- yet.
factant protein B expression (99); however, the relevance of
this observation to lung alveolarization has not yet been estab- Cell Therapy to Promote Alveolarization
lished.
The potential application of vitamin A and vitamin A ana- The applicability of stem cells to drive the repair of damaged
logs to promote lung maturation continues to receive attention lungs and the regeneration of lung tissue to promote that repair
because retinoid signaling is essential for proper diaphragm are currently a hot topic in pulmonary research (131, 141, 174,
and lung development. Costa and coworkers (76) have added 241), including BPD (265). Some strides forward have been
several retinoid-metabolizing genes to the list of retinoid path- made in the validation of cell therapy as a viable route to
way genes that are deregulated in human CDH, providing restore and promote lung maturation in experimental BPD.
further impetus for animal model studies. Along these lines, Key recent observations include the report of Alphonse and
James and coworkers (156) reported that a combination of coworkers (19), who demonstrated that, after exposure to
vitamin A and all-trans retinoic acid (in a 10:1 ratio) admin- hyperoxia in vitro, endothelial colony-forming cells (ECFCs)
istered every second day to newborn mice in the hyperoxia from human fetal lungs exhibited blunted proliferative and
BPD model (85% O2 for 14 days) improved lung architecture vessel-formation activity, as did ECFCs from hyperoxia-
(assessed by MLI and RAC) and partially normalized lung treated rat lungs (95% O2, from P1 to P14). Intrajugular

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00268.2015 • www.ajplung.org


Perspectives
L1260 ADVANCES IN LUNG ALVEOLARIZATION AND BPD

O2

Modulation of
reactive oxygen
species (ROS) Lung
generation
Adenosine receptor
Acute
Bronchodilatation activation/inhibition

Respiratory cAMP
phosphodiesterase Chronic
drive
modulation
Modulation Inflammation
cell death

Modulation of CAFFEINE
inflammatory
response
Cxcl1 Ccl2

Cxcl2 TNF-α IL-6


Late Early
treatment treatment
Low dose
?
AEC Macrophage Neutrophil
High dose
apoptosis influx influx

Reduced Increased
inflammation inflammation
Reduced Increased
apoptosis apoptosis

Ameliorated
Impaired
histological
alveolarization
damage

Improved Worsened
BPD status BPD status

Fig. 5. Possible mechanisms by which caffeine might attenuate BPD and arrested alveolarization in the developing lung. Caffeine administration has been
examined in early and late applications in clinical settings and experimentally in an acute and chronic administration protocol. Upward arrow indicates an
increased effect. The scheme only details new developments that were published between 1 January 2013 and 30 June 2015. AEC, alveolar epithelial cell; Ccl2,
chemokine (C-C motif) ligand 2; Cxcl1, chemokine (C-X-C motif) ligand 1; MIP-2, macrophage inflammatory protein 2; ROS, reactive oxygen species.

administration of human umbilical cord blood-derived ECFCs (MSCs) delivered by the intratracheal route in the rat hyperoxia
after hyperoxic lung injury in Rag1⫺/⫺ mouse pups (85% O2, model, in which alveolarization (assessed by MLI) was rescued
from P4 to P14) restored lung function and also restored both in the short and long term (6 mo) (278). In a similar study,
alveolarization (assessed by MLI) and lung vascular growth Sutsko and coworkers (324) compared whether intratracheal
and blunted pulmonary hypertension. The low level of engraft- administration of either MSCs or conditioned medium from
ment of ECFCs, together with protective effects of ECFC- MSCs influenced lung development in the hyperoxia rat model
conditioned medium, prompted the investigators to suggest (90% O2, between P2 and P16). Both MSC and MSC-condi-
that a paracrine effect conferred the restorative effects of tioned medium blunted the impact of hyperoxia on alveolar
ECFCs (110). Notably, 10 mo after ECFC administration, the development (assessed by MLI) and lung vessel density. Along
improvement in lung structure persisted. The same group of these lines, Baker and coworkers (29) applied conditioned
investigators reported the beneficial impact of human umbilical medium from late-outgrowth colony-forming cells, a type of
cord-derived perivascular cells and mesenchymal stem cells endothelial progenitor cell, in a bleomycin-based rat model of

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00268.2015 • www.ajplung.org


Perspectives
ADVANCES IN LUNG ALVEOLARIZATION AND BPD L1261
BPD. Although application of conditioned medium blunted identifying the correct stem cell type and route of administra-
heart remodeling (assessed by Fulton index) provoked in the tion, keeping in mind that a cocktail of multiple stem cell types
model, no impact on alveolarization (assessed by RAC) of rat may be the best approach, understanding how these cells
pup lungs was noted, leading these authors to criticize the promote lung maturation or correct blunted alveolarization,
potential benefit of conditioned medium administration in this and clarifying whether stem cell application may have harmful
model. Why the study of Baker and coworkers (29) could not long-term consequences, such as tumorigenesis. The consider-
demonstrate a beneficial impact of the intervention on alveo- able progress made over the past few years in applying stem
larization is not clear but may be related to the use of late- cells in animal models that recapitulate the arrested alveolar-
outgrowth colony-forming cells or a different model (bleomy- ization associated with clinical BPD has provided impetus for
cin), compared with the other three studies discussed above. translation of this idea to the clinical setting. The safety and
Along similar lines, Ahn and coworkers (11) evaluated the feasibility of transplanting allogeneic human umbilical cord
relative efficiency of human umbilical cord blood-derived blood-derived MSCs have recently been successfully demon-
MSCs, human adipose tissue-derived MSCs, and human um- strated in preterm infants in a phase I clinical trial
bilical cord blood mononuclear cells to protect against blunted (NCT01297205) (63). The results of a follow-up phase II
alveolar development induced by hyperoxia (90% O2 for 14 clinical trial to assess therapeutic efficacy (NCT01828957), as
days) in rat pups. The investigators assessed that the human well as a long-term follow-up safety assessment study of the
umbilical cord blood-derived MSCs were most efficient at MSC transplant recipients (NCT01897987) (10), are eagerly
promoting improved lung maturation in the background of awaited.
hyperoxia, which was correlated with increased VEGF and
hepatocyte growth factor production. The investigators fol- Synthesis and Concluding Remarks
lowed up with a timing-of-administration study (64) and re-
vealed that the application of umbilical cord blood-derived It is clear that many developments in our understanding of
MSCs was only beneficial when applied at P3 but not P10 alveolarization and the pathogenesis of BPD have been made
(90% O2, from P1 to P14, followed by 60% O2 from P14 to since the beginning of 2013. It is also clear that many open
P21). In another cord blood study, Mao and coworkers (216) questions remain, and new questions have been asked. Notable
applied cord blood-derived CD34⫹ cells that had been ex- advances in methodology used to study lung development have
panded in vitro, via the intranasal route, to mice in an FVB/N been made, including the development of new tissue fixation
transgenic mouse model of BPD with club (formerly Clara) and embedding protocols and stereology approaches for the
cell-driven expression of Fas ligand. When cord blood-derived unbiased quantification of lung structure. Further implementa-
CD34⫹ cells were expanded in vitro in the presence of dexa- tion of these methodologies, moving from RAC or MLI to
methasone, a moderate improvement in alveolarization (as- stereology measurements, would represent an important ad-
sessed by Lm) was noted. Similarly, the application of human vance in how we study lung structure although concerns about
amnion epithelial cells at P5, P6, and P7 to mouse pups time and cost are likely to remain barriers to the widespread
exposed to 85% O2 from P1 to P14 improved alveolarization implementation of these approaches in the foreseeable future.
(as assessed by MLI) (349). This Perspectives article has also highlighted the tremendous
Studies have also been undertaken by Ramachandran and heterogeneity of animal models to study arrested alveolariza-
coworkers (292) with single application of bone marrow- tion. The hyperoxia-based approaches in particular are prob-
derived c-Kit⫹ cells, which, when applied via the intratracheal lematic, given the current range of oxygen levels employed,
route, led to a significant improvement in both alveolarization together with variable periods of oxygen exposure, the combi-
(assessed by MLI) and vessel density in the background of nation of a second hit and a period of recovery under room air
hyperoxia (90% O2 from P2 to P15, intervention on P8). conditions, and posthyperoxia exposure. It appears that there
Directly in line with this study, Miranda and coworkers (236) remains scope for a systematic study, using state-of-the-art
applied stem cell factor (SCF; the ligand of the c-kit receptor) stereological approaches, to clarify whether an optimal injuri-
by daily injection (from P15 to P21) to rat pups exposed to ous oxygen concentration and window of exposure exist.
hyperoxia (90% O2 from P1 to P15, followed by recovery in Along these lines, it would be important to strike a balance
room air from P16 to P28). In that study, which represents a between the amount of oxygen employed as an injurious
therapeutic (not preventive) protocol, the treatment of oxygen- stimulus and the degree of damage to the developing lung that
injured lungs with SCF increased lung septal density and can be quantified. This is particularly important in pharmaco-
alveolarization (assessed by MLI), normalized lung collagen logical or transgenic animal studies in which a potentially
deposition, and also had a pronounced impact on the vascula- important subtle effect of a pharmacological or genetic inter-
ture, with partially normalized right ventricular systolic pres- vention on the structural development of an injured lung may
sure and a fully normalized Fulton index and lung vessel be missed because the injurious stimulus was unnecessarily
medial wall thickness. These data identify the c-Kit/SCF axis severe. In this way, the potential subtle modulation of alveo-
as an extremely exciting avenue for the management of stunted larization by interventions at superoxia levels (ⱖ85% O2) may
lung maturation and potentially tie in with the idea of c-Kit not be noted in lung structural analyses. Concerns also persist
delineating a putative endothelial progenitor cell population in about the sex of the experimental animals employed and the
developing human lungs (325). range of strains with variable responses to oxygen injury,
As highlighted by Stella Kourembanas (175) and Christo- which has resulted in opposite observations being made in
pher Baker and Steven Abman (28) in two recent editorials, ostensibly the same studies. Also related to mouse strains, there
many questions remain regarding stem cell therapy for the is increasing concern about the appropriateness of controls
management of arrested lung development. These include employed in transgenic mouse studies, particularly where

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00268.2015 • www.ajplung.org


Perspectives
L1262 ADVANCES IN LUNG ALVEOLARIZATION AND BPD

strains are maintained on mixed mouse strain backgrounds. It Sprouting and intussusceptive angiogenesis in postpneumonectomy lung
is clearly important that, in all experimental studies, the mouse growth: mechanisms of alveolar neovascularization. Angiogenesis 17:
541–551, 2014.
strain, the backcrossing and background of the transgenic mice, 5. Afolayan AJ, Teng RJ, Eis A, Rana U, Broniowska KA, Corbett JA,
and the sex of the mice should be clearly reported. Pritchard K, Konduri GG. Inducible HSP70 regulates superoxide
A large number of correlative studies have been reported dismutase-2 and mitochondrial oxidative stress in the endothelial cells
since 2013, revealing expression changes in molecules of from developing lungs. Am J Physiol Lung Cell Mol Physiol 306:
L351–L360, 2014.
interest that correlate with arrested alveolarization. Demonstra-
6. Aggarwal NR, King LS, D’Alessio FR. Diverse macrophage popula-
tion of a causal role for these pathogenic candidates remains to tions mediate acute lung inflammation and resolution. Am J Physiol Lung
be reported. Similarly, several transgenic mouse studies have Cell Mol Physiol 306: L709 –L725, 2014.
causally implicated particular proteins or pathways in normal 7. Ahlbrecht K, McGowan SE. In search of the elusive lipofibroblast in
or arrested alveolarization, and the further delineation of these human lungs. Am J Physiol Lung Cell Mol Physiol 307: L605–L608,
2014.
pathways to reveal new developmental mechanisms is eagerly 8. Ahlfeld SK, Gao Y, Conway SJ, Tepper RS. Relationship of structural
awaited. New technologies (such as the clustered regularly to functional impairment during alveolar-capillary membrane develop-
interspaced short palindromic repeats/Cas9), which will mas- ment. Am J Pathol 185: 913–919, 2015.
sively ease genetic manipulation in vitro and in vivo, together 9. Ahlfeld SK, Gao Y, Wang J, Horgusluoglu E, Bolanis E, Clapp DW,
with the continued production of new floxed mouse strains and Conway SJ. Periostin downregulation is an early marker of inhibited
neonatal murine lung alveolar septation. Birth Defects Res A Clin Mol
driver lines, will open many doors for the further dissection of Teratol 97: 373–385, 2013.
molecular pathways that drive normal and aberrant late lung 10. Ahn SY, Chang YS, Park WS. Stem cell therapy for bronchopulmonary
development. Among these, the role of ECM remodeling, dysplasia: bench to bedside translation. J Korean Med Sci 30: 509 –513,
ECM cell cross talk, and cell-cell cross talk will no doubt 2015.
feature in work that will emerge over the coming years, along 11. Ahn SY, Chang YS, Sung DK, Yoo HS, Sung SI, Choi SJ, Park WS.
Cell type-dependent variation in paracrine potency determines therapeu-
with attention to the role of progenitor cells and distinct cell tic efficacy against neonatal hyperoxic lung injury. Cytotherapy 17:
subpopulations and the regulation of alveolar epithelial cell 1025–1035, 2015.
plasticity, together with the mechanisms of disturbed vascular 12. Alanis DM, Chang DR, Akiyama H, Krasnow MA, Chen J. Two
growth and homeostasis. It is clear that much challenging but nested developmental waves demarcate a compartment boundary in the
mouse lung. Nat Commun 5: 3923, 2014.
exciting work lies ahead. 13. Alapati D, Rong M, Chen S, Hehre D, Hummler SC, Wu S. Inhibition
of beta-catenin signaling improves alveolarization and reduces pulmo-
ACKNOWLEDGMENTS
nary hypertension in experimental bronchopulmonary dysplasia. Am J
The authors thank Dr. José Alberto Rodríguez Castillo (Morty Laboratory, Respir Cell Mol Biol 51: 104 –113, 2014.
Department of Lung Development and Remodelling, Max Planck Institute for 14. Alapati D, Rong M, Chen S, Lin C, Li Y, Wu S. Inhibition of
Heart and Lung Research, Bad Nauheim, Germany) for outstanding assistance LRP5/6-mediated Wnt/beta-catenin signaling by Mesd attenuates hyper-
with the preparation of this Perspectives article. oxia-induced pulmonary hypertension in neonatal rats. Pediatr Res 73:
719 –725, 2013.
GRANTS 15. Albertine KH. Utility of large-animal models of BPD: chronically
ventilated preterm lambs. Am J Physiol Lung Cell Mol Physiol 308:
This study was supported by the Max Planck Society (to D. Silva, C.
L983–L1001, 2015.
Nardiello, A. Pozarska, and R. Morty), the German Research Foundation
16. Albertine KH, Jones GP, Starcher BC, Bohnsack JF, Davis PL, Cho
through grant Mo 1789/1 (to R. Morty), Excellence Cluster 147 “Cardio-
SC, Carlton DP, Bland RD. Chronic lung injury in preterm lambs.
Pulmonary System” (to R. Morty), the Federal Ministry of Higher Education,
Disordered respiratory tract development. Am J Respir Crit Care Med
Research, and the Arts of the State of Hessen LOEWE Program (to R. Morty),
159: 945–958, 1999.
and the German Center for Lung Research (Deutsches Zentrum für Lungen-
17. Alder JK, Barkauskas CE, Limjunyawong N, Stanley SE, Kembou
forschung; to D. Silva, C. Nardiello, A. Pozarska, and R. Morty).
F, Tuder RM, Hogan BL, Mitzner W, Armanios M. Telomere dys-
DISCLOSURES function causes alveolar stem cell failure. Proc Natl Acad Sci USA 112:
5099 –5104, 2015.
No conflicts of interest, financial or otherwise, are declared by the authors. 18. Alejandre-Alcázar MA, Kwapiszewska G, Reiss I, Amarie OV,
Marsh LM, Sevilla-Pérez J, Wygrecka M, Eul B, Köbrich S, Hesse
AUTHOR CONTRIBUTIONS M, Schermuly RT, Seeger W, Eickelberg O, Morty RE. Hyperoxia
D.M.S., C.N., and A.P. prepared figures; D.M.S., C.N., A.P., and R.E.M. modulates TGF-␤/BMP signaling in a mouse model of bronchopulmo-
drafted manuscript; D.M.S., C.N., A.P., and R.E.M. edited and revised man- nary dysplasia. Am J Physiol Lung Cell Mol Physiol 292: L537–L549,
uscript; D.M.S., C.N., A.P., and R.E.M. approved final version of manuscript. 2007.
19. Alphonse RS, Vadivel A, Fung M, Shelley WC, Critser PJ, Ionescu L,
REFERENCES O’Reilly M, Ohls RK, McConaghy S, Eaton F, Zhong S, Yoder M,
Thebaud B. Existence, functional impairment, and lung repair potential
1. Abdala-Valencia H, Berdnikovs S, Soveg FW, Cook-Mills JM. ␣-To- of endothelial colony-forming cells in oxygen-induced arrested alveolar
copherol supplementation of allergic female mice inhibits development growth. Circulation 129: 2144 –2157, 2014.
of CD11c⫹CD11b⫹ dendritic cells in utero and allergic inflammation in 20. Ambalavanan N, Cotten CM, Page GP, Carlo WA, Murray JC,
neonates. Am J Physiol Lung Cell Mol Physiol 307: L482–L496, 2014. Bhattacharya S, Mariani TJ, Cuna AC, Faye-Petersen OM, Kelly D,
2. Acker SN, Mandell EW, Sims-Lucas S, Gien J, Abman SH, Galam- Higgins RD; Genomics and Cytokine Subcommittees of the Eunice
bos C. Histologic identification of prominent intrapulmonary anasto- Kennedy Shriver National Institute of Child Health and Human
motic vessels in severe congenital diaphragmatic hernia. J Pediatr 166: Development Neonatal Research Network. Integrated genomic analy-
178 –183, 2015. ses in bronchopulmonary dysplasia. J Pediatr 166: 531–537; e513, 2015.
3. Acker SN, Seedorf GJ, Abman SH, Nozik-Grayck E, Partrick DA, 21. Ambalavanan N, Mourani P. Pulmonary hypertension in bronchopul-
Gien J. Pulmonary artery endothelial cell dysfunction and decreased monary dysplasia. Birth Defects Res A Clin Mol Teratol 100: 240 –246,
populations of highly proliferative endothelial cells in experimental 2014.
congenital diaphragmatic hernia. Am J Physiol Lung Cell Mol Physiol 22. Ambalavanan N, Stanishevsky A, Bulger A, Halloran B, Steele C,
305: L943–L952, 2013. Vohra Y, Matalon S. Titanium oxide nanoparticle instillation induces
4. Ackermann M, Houdek JP, Gibney BC, Ysasi A, Wagner W, Belle J, inflammation and inhibits lung development in mice. Am J Physiol Lung
Schittny JC, Enzmann F, Tsuda A, Mentzer SJ, Konerding MA. Cell Mol Physiol 304: L152–L161, 2013.

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00268.2015 • www.ajplung.org


Perspectives
ADVANCES IN LUNG ALVEOLARIZATION AND BPD L1263
23. Anyanwu AC, Bentley JK, Popova AP, Malas O, Alghanem H, 41. Bodempudi V, Hergert P, Smith K, Xia H, Herrera J, Peterson M,
Goldsmith AM, Hershenson MB, Pinsky DJ. Suppression of inflam- Khalil W, Kahm J, Bitterman PB, Henke CA. miR-210 promotes IPF
matory cell trafficking and alveolar simplification by the heme oxygen- fibroblast proliferation in response to hypoxia. Am J Physiol Lung Cell
ase-1 product carbon monoxide. Am J Physiol Lung Cell Mol Physiol Mol Physiol 307: L283–L294, 2014.
306: L749 –L763, 2014. 42. Boucherat O, Montaron S, Berube-Simard FA, Aubin J, Philippidou
24. Aravamudan B, Delmotte P, Thompson M, Vassallo R, Sieck GC, P, Wellik DM, Dasen JS, Jeannotte L. Partial functional redundancy
Pabelick CM, Prakash YS. Response to letter by Dr. Marc Hershenson between Hoxa5 and Hoxb5 paralog genes during lung morphogenesis.
(exposure of airway smooth muscle cells to cigarette smoke extract). Am Am J Physiol Lung Cell Mol Physiol 304: L817–L830, 2013.
J Physiol Lung Cell Mol Physiol 307: L346, 2014. 43. Bouhaddioui W, Provost PR, Tremblay Y. Identification of most stable
25. Aravamudan B, Kiel A, Freeman M, Delmotte P, Thompson M, endogenous control genes for microRNA quantification in the developing
Vassallo R, Sieck GC, Pabelick CM, Prakash YS. Cigarette smoke- mouse lung. PLoS One 9: e111855, 2014.
induced mitochondrial fragmentation and dysfunction in human airway 44. Bozyk PD, Bentley JK, Popova AP, Anyanwu AC, Linn MD, Gold-
smooth muscle. Am J Physiol Lung Cell Mol Physiol 306: L840 –L854, smith AM, Pryhuber GS, Moore BB, Hershenson MB. Neonatal
2014. periostin knockout mice are protected from hyperoxia-induced alveolar
26. Ayala de la Pena F, Kanasaki K, Kanasaki M, Vong S, Rovira C, simplification. PLoS One 7: e31336, 2012.
Kalluri R. Specific activation of K-RasG12D allele in the bladder 45. Brahmajothi MV, Sun NZ, Auten RL. S-nitrosothiol transport via
urothelium results in lung alveolar and vascular defects. PLoS One 9:
PEPT2 mediates biological effects of nitric oxide gas exposure in
e95888, 2014.
macrophages. Am J Respir Cell Mol Biol 48: 230 –239, 2013.
27. Bachiller PR, Cornog KH, Kato R, Buys ES, Roberts JD Jr. Soluble
46. Brahmajothi MV, Tinch BT, Wempe MF, Endou H, Auten RL.
guanylate cyclase modulates alveolarization in the newborn lung. Am J
Hyperoxia inhibits nitric oxide treatment effects in alveolar epithelial
Physiol Lung Cell Mol Physiol 305: L569 –L581, 2013.
cells via effects on L-type amino acid transporter-1. Antioxid Redox
28. Baker CD, Abman SH. Umbilical cord stem cell therapy for broncho-
pulmonary dysplasia: ready for prime time? Thorax 68: 402–404, 2013. Signal 21: 1823–1836, 2014.
29. Baker CD, Seedorf GJ, Wisniewski BL, Black CP, Ryan SL, Balasu- 47. Brandsma CA, Timens W, Jonker MR, Rutgers B, Noordhoek JA,
bramaniam V, Abman SH. Endothelial colony-forming cell condi- Postma DS. Differential effects of fluticasone on extracellular matrix
tioned media promote angiogenesis in vitro and prevent pulmonary production by airway and parenchymal fibroblasts in severe COPD. Am
hypertension in experimental bronchopulmonary dysplasia. Am J Physiol J Physiol Lung Cell Mol Physiol 305: L582–L589, 2013.
Lung Cell Mol Physiol 305: L73–L81, 2013. 48. Brechbuhl HM, Li B, Smith RW, Reynolds SD. Epidermal growth
30. Barkauskas CE, Cronce MJ, Rackley CR, Bowie EJ, Keene DR, factor receptor activity is necessary for mouse basal cell proliferation. Am
Stripp BR, Randell SH, Noble PW, Hogan BL. Type 2 alveolar cells J Physiol Lung Cell Mol Physiol 307: L800 –L810, 2014.
are stem cells in adult lung. J Clin Invest 123: 3025–3036, 2013. 49. Brew N, Hooper SB, Zahra V, Wallace M, Harding R. Mechanical
31. Bartlett JA, Albertolle ME, Wohlford-Lenane C, Pezzulo AA, Zab- ventilation injury and repair in extremely and very preterm lungs. PLoS
ner J, Niles RK, Fisher SJ, McCray PB Jr, Williams KE. Protein One 8: e63905, 2013.
composition of bronchoalveolar lavage fluid and airway surface liquid 50. Britt RD Jr, Velten M, Tipple TE, Nelin LD, Rogers LK. Cycloox-
from newborn pigs. Am J Physiol Lung Cell Mol Physiol 305: L256 – ygenase-2 in newborn hyperoxic lung injury. Free Radic Biol Med 61:
L266, 2013. 502–511, 2013.
32. Belcastro R, Lopez L, Li J, Masood A, Tanswell AK. Chronic lung 51. Buczynski BW, Yee M, Martin KC, Lawrence BP, O’Reilly MA.
injury in the neonatal rat: up-regulation of TGFbeta1 and nitration of Neonatal hyperoxia alters the host response to influenza A virus infection
IGF-R1 by peroxynitrite as likely contributors to impaired alveologen- in adult mice through multiple pathways. Am J Physiol Lung Cell Mol
esis. Free Radic Biol Med 80: 1–11, 2015. Physiol 305: L282–L290, 2013.
33. Berger J, Bhandari V. Animal models of bronchopulmonary dysplasia. 52. Buczynski BW, Yee M, Paige Lawrence B, O’Reilly MA. Lung
The term mouse models. Am J Physiol Lung Cell Mol Physiol 307: development and the host response to influenza A virus are altered by
L936 –L947, 2014. different doses of neonatal oxygen in mice. Am J Physiol Lung Cell Mol
34. Bhandari V. Postnatal inflammation in the pathogenesis of bronchopul- Physiol 302: L1078 –L1087, 2012.
monary dysplasia. Birth Defects Res A Clin Mol Teratol 100: 189 –201, 53. Butler JP. Looking back and looking forward: normal lung growth and
2014. the pathophysiological consequences of ‘misplaced’ growth in disease.
35. Bhargava M, Dey S, Becker T, Steinbach M, Wu B, Lee SM, Higgins Eur J Radiol 83: 2–5, 2014.
L, Kumar V, Bitterman PB, Ingbar DH, Wendt CH. Protein expres- 54. Cai Y, Winn ME, Zehmer JK, Gillette WK, Lubkowski JT, Pilon
sion profile of rat type two alveolar epithelial cells during hyperoxic AL, Kimura S. Preclinical evaluation of human secretoglobin 3A2 in
stress and recovery. Am J Physiol Lung Cell Mol Physiol 305: L604 – mouse models of lung development and fibrosis. Am J Physiol Lung Cell
L614, 2013. Mol Physiol 306: L10 –L22, 2014.
36. Bhattacharya S, Mariani TJ. Systems biology approaches to identify 55. Caminita F, van der Merwe M, Hance B, Krishnan R, Miller S,
developmental bases for lung diseases. Pediatr Res 73: 514 –522, 2013. Buddington K, Buddington RK. A preterm pig model of lung imma-
37. Bhattacharya S, Zhou Z, Yee M, Chu CY, Lopez AM, Lunger VA,
turity and spontaneous infant respiratory distress syndrome. Am J Physiol
Solleti SK, Resseguie E, Buczynski B, Mariani TJ, O’Reilly MA. The
Lung Cell Mol Physiol 308: L118 –L129, 2015.
genome-wide transcriptional response to neonatal hyperoxia identifies
56. Cano E, Carmona R, Munoz-Chapuli R. Wt1-expressing progenitors
Ahr as a key regulator. Am J Physiol Lung Cell Mol Physiol 307:
contribute to multiple tissues in the developing lung. Am J Physiol Lung
L516 –L523, 2014.
38. Bland RD, Ertsey R, Mokres LM, Xu L, Jacobson BE, Jiang S, Cell Mol Physiol 305: L322–L332, 2013.
Alvira CM, Rabinovitch M, Shinwell ES, Dixit A. Mechanical venti- 57. Carpenter TC, Stenmark KR. Predisposition of infants with chronic
lation uncouples synthesis and assembly of elastin and increases apopto- lung disease to respiratory syncytial virus-induced respiratory failure: a
sis in lungs of newborn mice. Prelude to defective alveolar septation vascular hypothesis. Pediatr Infect Dis J 23: S33–S40, 2004.
during lung development? Am J Physiol Lung Cell Mol Physiol 294: 58. Carver BJ, Plosa EJ, Stinnett AM, Blackwell TS, Prince LS. Inter-
L3–L14, 2008. actions between NF-kappaB and SP3 connect inflammatory signaling
39. Bland RD, Mokres LM, Ertsey R, Jacobson BE, Jiang S, Rabinovitch with reduced FGF-10 expression. J Biol Chem 288: 15318 –15325, 2013.
M, Xu L, Shinwell ES, Zhang F, Beasley MA. Mechanical ventilation 59. Castro EC, Parks WT, Galambos C. The ontogeny of human pulmo-
with 40% oxygen reduces pulmonary expression of genes that regulate nary angiotensin-converting enzyme and its aberrant expression may
lung development and impairs alveolar septation in newborn mice. Am J contribute to the pathobiology of bronchopulmonary dysplasia (BPD).
Physiol Lung Cell Mol Physiol 293: L1099 –L1110, 2007. Pediatr Pulmonol 49: 985–990, 2014.
40. Bland RD, Xu L, Ertsey R, Rabinovitch M, Albertine KH, Wynn KA, 60. Chan JK, Vogel CF, Baek J, Kodani SD, Uppal RS, Bein KJ,
Kumar VH, Ryan RM, Swartz DD, Csiszar K, Fong KS. Dysregula- Anderson DS, Van Winkle LS. Combustion derived ultrafine particles
tion of pulmonary elastin synthesis and assembly in preterm lambs with induce cytochrome P-450 expression in specific lung compartments in
chronic lung disease. Am J Physiol Lung Cell Mol Physiol 292: L1370 – the developing neonatal and adult rat. Am J Physiol Lung Cell Mol
L1384, 2007. Physiol 304: L665–L677, 2013.

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00268.2015 • www.ajplung.org


Perspectives
L1264 ADVANCES IN LUNG ALVEOLARIZATION AND BPD

61. Chang DR, Martinez Alanis D, Miller RK, Ji H, Akiyama H, McCrea 81. Datta A, Kim GA, Taylor JM, Gugino SF, Farrow KN, Schumacker
PD, Chen J. Lung epithelial branching program antagonizes alveolar PT, Berkelhamer SK. Mouse lung development and NOX1 induction
differentiation. Proc Natl Acad Sci USA 110: 18042–18051, 2013. during hyperoxia are developmentally regulated and mitochondrial ROS-
62. Chang M, Bany-Mohammed F, Kenney MC, Beharry KD. Effects of dependent. Am J Physiol Lung Cell Mol Physiol 309: L369 –L377, 2015.
a superoxide dismutase mimetic on biomarkers of lung angiogenesis and 82. Davidovich N, Huang J, Margulies SS. Reproducible uniform equibi-
alveolarization during hyperoxia with intermittent hypoxia. Am J Transl axial stretch of precision-cut lung slices. Am J Physiol Lung Cell Mol
Res 5: 594 –607, 2013. Physiol 304: L210 –L220, 2013.
63. Chang YS, Ahn SY, Yoo HS, Sung SI, Choi SJ, Oh WI, Park WS. 83. Dayanim S, Lopez B, Maisonet TM, Grewal S, Londhe VA. Caffeine
Mesenchymal stem cells for bronchopulmonary dysplasia: phase 1 dose- induces alveolar apoptosis in the hyperoxia-exposed developing mouse
escalation clinical trial. J Pediatr 164: 966 –972 e966, 2014. lung. Pediatr Res 75: 395–402, 2014.
64. Chang YS, Choi SJ, Ahn SY, Sung DK, Sung SI, Yoo HS, Oh WI, 84. del Cerro MJ, Sabate Rotes A, Carton A, Deiros L, Bret M, Cordeiro
Park WS. Timing of umbilical cord blood derived mesenchymal stem M, Verdu C, Barrios MI, Albajara L, Gutierrez-Larraya F. Pulmo-
cells transplantation determines therapeutic efficacy in the neonatal nary hypertension in bronchopulmonary dysplasia: clinical findings,
hyperoxic lung injury. PLoS One 8: e52419, 2013. cardiovascular anomalies and outcomes. Pediatr Pulmonol 49: 49 –59,
65. Chao CM, El Agha E, Tiozzo C, Minoo P, Bellusci S. A breath of fresh 2014.
air on the mesenchyme: impact of impaired mesenchymal development 85. Delaney C, Gien J, Roe G, Isenberg N, Kailey J, Abman SH.
on the pathogenesis of bronchopulmonary dysplasia. Front Med (Laus- Serotonin contributes to high pulmonary vascular tone in a sheep model
anne) 2: 27, 2015. of persistent pulmonary hypertension of the newborn. Am J Physiol Lung
66. Chen B, Nelin VE, Locy ML, Jin Y, Tipple TE. Thioredoxin-1 Cell Mol Physiol 304: L894 –L901, 2013.
mediates hypoxia-induced pulmonary artery smooth muscle cell prolif- 86. Desai TJ, Brownfield DG, Krasnow MA. Alveolar progenitor and stem
eration. Am J Physiol Lung Cell Mol Physiol 305: L389 –L395, 2013. cells in lung development, renewal and cancer. Nature 507: 190 –194,
67. Chen F, Marquez H, Kim YK, Qian J, Shao F, Fine A, Cruikshank 2014.
WW, Quadro L, Cardoso WV. Prenatal retinoid deficiency leads to 87. Dodson RB, Morgan MR, Galambos C, Hunter KS, Abman SH.
airway hyperresponsiveness in adult mice. J Clin Invest 124: 801–811, Chronic intrauterine pulmonary hypertension increases main pulmonary
2014. artery stiffness and adventitial remodeling in fetal sheep. Am J Physiol
68. Chen X, Walther FJ, Sengers RM, Laghmani EH, Salam A, Folkerts Lung Cell Mol Physiol 307: L822–L828, 2014.
G, Pera T, Wagenaar GT. Metformin attenuates hyperoxia-induced 88. Drummond S, Ramachandran S, Torres E, Huang J, Hehre D,
lung injury in neonatal rats by reducing the inflammatory response. Am Suguihara C, Young KC. CXCR4 blockade attenuates hyperoxia-
J Physiol Lung Cell Mol Physiol 309: L262–L270, 2015. induced lung injury in neonatal rats. Neonatology 107: 304 –311, 2015.
69. Chetty A, Bennett M, Dang L, Nakamura D, Cao GJ, Mujahid S, 89. Duong-Quy S, Hua-Huy T, Pham H, Tang X, Mercier JC, Baud O,
Volpe M, Herman I, Becerra SP, Nielsen HC. Pigment epithelium- Dinh-Xuan AT. Early inhaled nitric oxide at high dose enhances rat lung
derived factor mediates impaired lung vascular development in neonatal development after birth. Nitric Oxide 38: 8 –16, 2014.
hyperoxia. Am J Respir Cell Mol Biol 52: 295–303, 2015. 90. Dye BR, Hill DR, Ferguson MA, Tsai YH, Nagy MS, Dyal R, Wells
70. Cho HY, Kleeberger SR. Association of Nrf2 with airway pathogenesis: JM, Mayhew CN, Nattiv R, Klein OD, White ES, Deutsch GH,
lessons learned from genetic mouse models. Arch Toxicol. In press. Spence JR. In vitro generation of human pluripotent stem cell derived
71. Choi CW, Kim BI, Mason SN, Potts-Kant EN, Brahmajothi MV, lung organoids. Elife 24: 4, 2015.
Auten RL. Intra-amniotic LPS amplifies hyperoxia-induced airway hy- 91. Egger C, Gerard C, Vidotto N, Accart N, Cannet C, Dunbar A,
perreactivity in neonatal rats. Pediatr Res 74: 11–18, 2013. Tigani B, Piaia A, Jarai G, Jarman E, Schmid HA, Beckmann N.
72. Choo-Wing R, Syed MA, Harijith A, Bowen B, Pryhuber G, Janer C, Lung volume quantified by MRI reflects extracellular-matrix deposition
Andersson S, Homer RJ, Bhandari V. Hyperoxia and interferon- and altered pulmonary function in bleomycin models of fibrosis: effects
gamma-induced injury in developing lungs occur via cyclooxygenase-2 of SOM230. Am J Physiol Lung Cell Mol Physiol 306: L1064 –L1077,
and the endoplasmic reticulum stress-dependent pathway. Am J Respir 2014.
Cell Mol Biol 48: 749 –757, 2013. 92. El Agha E, Bellusci S. Walking along the fibroblast growth factor 10
73. Collins JJ, Kallapur SG, Knox CL, Kemp MW, Kuypers E, Zim- route: a key pathway to understand the control and regulation of epithe-
mermann LJ, Newnham JP, Jobe AH, Kramer BW. Repeated intra- lial and mesenchymal cell-lineage formation during lung development
uterine exposures to inflammatory stimuli attenuated transforming and repair after injury. Scientifica (Cairo) 2014: 538379, 2014.
growth factor-beta signaling in the ovine fetal lung. Neonatology 104: 93. El Agha E, Herold S, Al Alam D, Quantius J, MacKenzie B, Carraro
49 –55, 2013. G, Moiseenko A, Chao CM, Minoo P, Seeger W, Bellusci S. Fgf10-
74. Collins JJ, Kunzmann S, Kuypers E, Kemp MW, Speer CP, Newn- positive cells represent a progenitor cell population during lung devel-
ham JP, Kallapur SG, Jobe AH, Kramer BW. Antenatal glucocorti- opment and postnatally. Development 141: 296 –306, 2014.
coids counteract LPS changes in TGF-␤ pathway and caveolin-1 in ovine 94. Eldredge LC, Treuting PM, Manicone AM, Ziegler SF, Parks WC,
fetal lung. Am J Physiol Lung Cell Mol Physiol 304: L438 –L444, 2013. McGuire JK. CD11b mononuclear cells mitigate hyperoxia-induced
75. Cornett B, Snowball J, Varisco BM, Lang R, Whitsett J, Sinner D. lung injury in neonatal mice. Am J Respir Cell Mol Biol. In press.
Wntless is required for peripheral lung differentiation and pulmonary 95. Elremaly W, Mohamed I, Mialet-Marty T, Rouleau T, Lavoie JC.
vascular development. Dev Biol 379: 38 –52, 2013. Ascorbylperoxide from parenteral nutrition induces an increase of redox
76. Coste K, Beurskens LW, Blanc P, Gallot D, Delabaere A, Blanchon potential of glutathione and loss of alveoli in newborn guinea pig lungs.
L, Tibboel D, Labbe A, Rottier RJ, Sapin V. Metabolic disturbances of Redox Biol 2: 725–731, 2014.
the vitamin A pathway in human diaphragmatic hernia. Am J Physiol 96. Enomoto M, Jain A, Pan J, Shifrin Y, Van Vliet T, McNamara PJ,
Lung Cell Mol Physiol 308: L147–L157, 2015. Jankov RP, Belik J. Newborn rat response to single vs. combined
77. Counter WB, Wang IQ, Farncombe TH, Labiris NR. Airway and cGMP-dependent pulmonary vasodilators. Am J Physiol Lung Cell Mol
pulmonary vascular measurements using contrast-enhanced micro-CT in Physiol 306: L207–L215, 2014.
rodents. Am J Physiol Lung Cell Mol Physiol 304: L831–L843, 2013. 97. Eurlings IM, Dentener MA, Mercken EM, de Cabo R, Bracke KR,
78. Cuna A, Halloran B, Faye-Petersen O, Kelly D, Crossman DK, Cui Vernooy JH, Wouters EF, Reynaert NL. A comparative study of
X, Pandit K, Kaminski N, Bhattacharya S, Ahmad A, Mariani TJ, matrix remodeling in chronic models for COPD; mechanistic insights
Ambalavanan N. Alterations in gene expression and DNA methylation into the role of TNF-␣. Am J Physiol Lung Cell Mol Physiol 307:
during murine and human lung alveolar septation. Am J Respir Cell Mol L557–L565, 2014.
Biol 53: 60 –73, 2015. 98. Fediuk J, Sikarwar AS, Nolette N, Dakshinamurti S. Thromboxane-
79. Czövek D, Peták F, Donati Y, Belin X, Pache JC, Barazzone Argi- induced actin polymerization in hypoxic neonatal pulmonary arterial
roffo C, Habre W. Prevention of hyperoxia-induced bronchial hyperre- myocytes involves Cdc42 signaling. Am J Physiol Lung Cell Mol Physiol
activity by sildenafil and vasoactive intestinal peptide: impact of pre- 307: L877–L887, 2014.
served lung function and structure. Respir Res 15: 81, 2014. 99. Fehrholz M, Hutten M, Kramer BW, Speer CP, Kunzmann S.
80. D’Angio CT, Ryan RM. Animal models of bronchopulmonary dyspla- Amplification of steroid-mediated SP-B expression by physiological
sia. The preterm and term rabbit models. Am J Physiol Lung Cell Mol levels of caffeine. Am J Physiol Lung Cell Mol Physiol 306: L101–L109,
Physiol 307: L959 –L969, 2014. 2014.

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00268.2015 • www.ajplung.org


Perspectives
ADVANCES IN LUNG ALVEOLARIZATION AND BPD L1265
100. Fehrholz M, Speer CP, Kunzmann S. Caffeine and rolipram affect postnatal hyperoxia: closer to clinical features? Ann Anat 195: 351–358,
Smad signalling and TGF-beta1 stimulated CTGF and transgelin expres- 2013.
sion in lung epithelial cells. PLoS One 9: e97357, 2014. 121. Gortner L, Shen J, Tutdibi E. Sexual dimorphism of neonatal lung
101. Fineschi S, De Cunto G, Facchinetti F, Civelli M, Imbimbo BP, development. Klin Padiatr 225: 64 –69, 2013.
Carnini C, Villetti G, Lunghi B, Stochino S, Gibbons DL, Hayday A, 122. Greer RM, Miller JD, Okoh VO, Halloran BA, Prince LS. Epithelial-
Lungarella G, Cavarra E. Receptor for advanced glycation end prod- mesenchymal co-culture model for studying alveolar morphogenesis.
ucts contributes to postnatal pulmonary development and adult lung Organogenesis 10: 340 –349, 2014.
maintenance program in mice. Am J Respir Cell Mol Biol 48: 164 –171, 123. Grzesik BA, Vohwinkel CU, Morty RE, Mayer K, Herold S, Seeger
2013. W, Vadasz I. Efficient gene delivery to primary alveolar epithelial cells
102. Firsova AB, Cole TJ, Mollard R. Transient vascular and long-term by nucleofection. Am J Physiol Lung Cell Mol Physiol 305: L786 –L794,
alveolar deficits following a hyperoxic injury to neonatal mouse lung. 2013.
BMC Pulm Med 14: 59, 2014. 124. Gupta A, Perez M, Lee KJ, Taylor JM, Farrow KN. SOD2 activity is
103. Floen MJ, Forred BJ, Bloom EJ, Vitiello PF. Thioredoxin-1 redox not impacted by hyperoxia in murine neonatal pulmonary artery smooth
signaling regulates cell survival in response to hyperoxia. Free Radic muscle cells and mice. Int J Mol Sci 16: 6373–6390, 2015.
Biol Med 75: 167–177, 2014. 125. Hadchouel A, Franco-Montoya ML, Delacourt C. Altered lung devel-
104. Friedmacher F, Fujiwara N, Hofmann AD, Takahashi H, Alvarez opment in bronchopulmonary dysplasia. Birth Defects Res A Clin Mol
LA, Gosemann JH, Puri P. Prenatal retinoic acid increases lipofibro- Teratol 100: 158 –167, 2014.
blast expression in hypoplastic rat lungs with experimental congenital 126. Hagood JS. Beyond the genome: epigenetic mechanisms in lung remod-
diaphragmatic hernia. J Pediatr Surg 49: 876 –881; discussion 881, 2014. eling. Physiology (Bethesda) 29: 177–185, 2014.
105. Friedmacher F, Fujiwara N, Hofmann AD, Takahashi H, Gosemann 127. Hagood JS, Ambalavanan N. Systems biology of lung development and
JH, Puri P. Evidence for decreased lipofibroblast expression in hyp- regeneration: current knowledge and recommendations for future re-
oplastic rat lungs with congenital diaphragmatic hernia. Pediatr Surg Int search. Wiley Interdiscip Rev Syst Biol Med 5: 125–133, 2013.
30: 1023–1029, 2014. 128. Han W, Guo C, Liu Q, Yu B, Liu Z, Yang J, Deng C. Aberrant elastin
106. Friedmacher F, Gosemann JH, Fujiwara N, Alvarez LA, Cor- remodeling in the lungs of O2-exposed newborn mice; primarily results
cionivoschi N, Puri P. Spatiotemporal alterations in Sprouty-2 expres- from perturbed interaction between integrins and elastin. Cell Tissue Res
sion and tyrosine phosphorylation in nitrofen-induced pulmonary hyp- 359: 589 –603, 2015.
oplasia. J Pediatr Surg 48: 2219 –2225, 2013. 129. Hanna G, Fontanella A, Palmer G, Shan S, Radiloff DR, Zhao Y,
107. Friedmacher F, Gosemann JH, Takahashi H, Corcionivoschi N, Puri Irwin D, Hamilton K, Boico A, Piantadosi CA, Blueschke G, Dew-
P. Decreased pulmonary c-Cbl expression and tyrosine phosphorylation hirst M, McMahon T, Schroeder T. Automated measurement of blood
in the nitrofen-induced rat model of congenital diaphragmatic hernia. flow velocity and direction and hemoglobin oxygen saturation in the rat
Pediatr Surg Int 29: 19 –24, 2013. lung using intravital microscopy. Am J Physiol Lung Cell Mol Physiol
108. Friedmacher F, Hofmann AD, Takahashi H, Takahashi T, Gos- 304: L86 –L91, 2013.
emann JH, Puri P. Disruption of THY-1 signaling in alveolar lipofi-
130. Harijith A, Pendyala S, Reddy NM, Bai T, Usatyuk PV, Berdyshev E,
broblasts in experimentally induced congenital diaphragmatic hernia.
Gorshkova I, Huang LS, Mohan V, Garzon S, Kanteti P, Reddy SP,
Pediatr Surg Int 30: 129 –135, 2014.
Raj JU, Natarajan V. Sphingosine kinase 1 deficiency confers protec-
109. Friedmacher F, Hofmann AD, Takahashi T, Takahashi H, Kutasy B,
tion against hyperoxia-induced bronchopulmonary dysplasia in a murine
Puri P. Prenatal administration of all-trans retinoic acid upregulates
model: role of S1P signaling and Nox proteins. Am J Pathol 183:
leptin signaling in hypoplastic rat lungs with experimental congenital
1169 –1182, 2013.
diaphragmatic hernia. Pediatr Surg Int 30: 1183–1190, 2014.
131. Herriges M, Morrisey EE. Lung development: orchestrating the gen-
110. Fung ME, Thebaud B. Stem cell-based therapy for neonatal lung
eration and regeneration of a complex organ. Development 141: 502–513,
disease: it is in the juice. Pediatr Res 75: 2–7, 2014.
111. Galambos C, Sims-Lucas S, Abman SH. Histologic evidence of in- 2014.
trapulmonary anastomoses by three-dimensional reconstruction in severe 132. Herriges MJ, Swarr DT, Morley MP, Rathi KS, Peng T, Stewart
bronchopulmonary dysplasia. Ann Am Thorac Soc 10: 474 –481, 2013. KM, Morrisey EE. Long noncoding RNAs are spatially correlated with
112. Galambos C, Sims-Lucas S, Abman SH. Three-dimensional recon- transcription factors and regulate lung development. Genes Dev 28:
struction identifies misaligned pulmonary veins as intrapulmonary shunt 1363–1379, 2014.
vessels in alveolar capillary dysplasia. J Pediatr 164: 192–195, 2014. 133. Herring MJ, Avdalovic MV, Quesenberry CL, Putney LF, Tyler NK,
113. Galambos C, Sims-Lucas S, Ali N, Gien J, Dishop MK, Abman SH. Ventimiglia FF, St George JA, Hyde DM. Accelerated structural
Intrapulmonary vascular shunt pathways in alveolar capillary dysplasia decrements in the aging female rhesus macaque lung compared with
with misalignment of pulmonary veins. Thorax 70: 84 –85, 2015. males. Am J Physiol Lung Cell Mol Physiol 304: L125–L134, 2013.
114. Galinsky R, Polglase GR, Hooper SB, Black MJ, Moss TJ. The 134. Herring MJ, Putney LF, Wyatt G, Finkbeiner WE, Hyde DM.
consequences of chorioamnionitis: preterm birth and effects on develop- Growth of alveoli during postnatal development in humans based on
ment. J Pregnancy 2013: 412831, 2013. stereological estimation. Am J Physiol Lung Cell Mol Physiol 307:
115. Galvis LA, Holik AZ, Short KM, Pasquet J, Lun AT, Blewitt ME, L338 –L344, 2014.
Smyth IM, Ritchie ME, Asselin-Labat ML. Repression of Igf1 expres- 135. Hershenson MB, Aghili S, Punjabi N, Hernandez C, Ray DW,
sion by Ezh2 prevents basal cell differentiation in the developing lung. Garland A, Glagov S, Solway J. Hyperoxia-induced airway hyperre-
Development 142: 1458 –1469, 2015. sponsiveness and remodeling in immature rats. Am J Physiol Lung Cell
116. Gammon ST, Foje N, Brewer EM, Owers E, Downs CA, Budde MD, Mol Physiol 262: L263–L269, 1992.
Leevy WM, Helms MN. Preclinical anatomical, molecular, and func- 136. Hershenson MB, Garland A, Kelleher MD, Zimmermann A, Her-
tional imaging of the lung with multiple modalities. Am J Physiol Lung nandez C, Solway J. Hyperoxia-induced airway remodeling in immature
Cell Mol Physiol 306: L897–L914, 2014. rats. Correlation with airway responsiveness. Am Rev Respir Dis 146:
117. Ghosh MC, Gorantla V, Makena PS, Luellen C, Sinclair SE, 1294 –1300, 1992.
Schwingshackl A, Waters CM. Insulin-like growth factor-I stimulates 137. Hilgendorff A, O’Reilly MA. Bronchopulmonary dysplasia early
differentiation of ATII cells to ATI-like cells through activation of changes leading to long-term consequences. Front Med (Lausanne) 2: 2,
Wnt5a. Am J Physiol Lung Cell Mol Physiol 305: L222–L228, 2013. 2015.
118. Gien J, Tseng N, Seedorf G, Roe G, Abman SH. Peroxisome prolif- 138. Hilgendorff A, Parai K, Ertsey R, Navarro E, Jain N, Carandang F,
erator activated receptor-␥-Rho-kinase interactions contribute to vascular Peterson J, Mokres L, Milla C, Preuss S, Alcazar MA, Khan S,
remodeling after chronic intrauterine pulmonary hypertension. Am J Masumi J, Ferreira-Tojais N, Mujahid S, Starcher B, Rabinovitch
Physiol Lung Cell Mol Physiol 306: L299 –L308, 2014. M, Bland R. Lung matrix and vascular remodeling in mechanically
119. Goldblatt H, Benischek M. Vitamin A deficiency and metaplasia. J Exp ventilated elastin haploinsufficient newborn mice. Am J Physiol Lung
Med 46: 699 –707, 1927. Cell Mol Physiol 308: L464 –L478, 2015.
120. Gortner L, Monz D, Mildau C, Shen J, Kasoha M, Laschke MW, 139. Hilgendorff A, Reiss I, Ehrhardt H, Eickelberg O, Alvira CM.
Roolfs T, Schmiedl A, Meier C, Tutdibi E. Bronchopulmonary dys- Chronic lung disease in the preterm infant. Lessons learned from animal
plasia in a double-hit mouse model induced by intrauterine hypoxia and models. Am J Respir Cell Mol Biol 50: 233–245, 2014.

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00268.2015 • www.ajplung.org


Perspectives
L1266 ADVANCES IN LUNG ALVEOLARIZATION AND BPD

140. Hillman NH, Kemp MW, Noble PB, Kallapur SG, Jobe AH. Sus- 160. Joza S, Wang J, Tseu I, Ackerley C, Post M. Fetal, but not postnatal,
tained inflation at birth did not protect preterm fetal sheep from lung deletion of semaphorin-neuropilin-1 signaling affects murine alveolar
injury. Am J Physiol Lung Cell Mol Physiol 305: L446 –L453, 2013. development. Am J Respir Cell Mol Biol 49: 627–636, 2013.
141. Hogan BL, Barkauskas CE, Chapman HA, Epstein JA, Jain R, Hsia 161. Kallapur SG, Kramer BW, Jobe AH. Ureaplasma and BPD. Semin
CC, Niklason L, Calle E, Le A, Randell SH, Rock J, Snitow M, Perinatol 37: 94 –101, 2013.
Krummel M, Stripp BR, Vu T, White ES, Whitsett JA, Morrisey EE. 162. Kapere Ochieng J, Schilders K, Kool H, Buscop-van Kempen M,
Repair and regeneration of the respiratory system: complexity, plasticity, Boerema-De Munck A, Grosveld F, Wijnen R, Tibboel D, Rottier RJ.
and mechanisms of lung stem cell function. Cell Stem Cell 15: 123–138, Differentiated type II pneumocytes can be reprogrammed by ectopic
2014. Sox2 expression. PLoS One 9: e107248, 2014.
142. Hogmalm A, Bry M, Strandvik B, Bry K. IL-1␤ expression in the 163. Kawamura T, Wakabayashi N, Shigemura N, Huang CS, Masutani
distal lung epithelium disrupts lung morphogenesis and epithelial cell K, Tanaka Y, Noda K, Peng X, Takahashi T, Billiar TR, Okumura
differentiation in fetal mice. Am J Physiol Lung Cell Mol Physiol 306: M, Toyoda Y, Kensler TW, Nakao A. Hydrogen gas reduces hyperoxic
L23–L34, 2014. lung injury via the Nrf2 pathway in vivo. Am J Physiol Lung Cell Mol
143. Hou A, Fu J, Yang H, Zhu Y, Pan Y, Xu S, Xue X. Hyperoxia Physiol 304: L646 –L656, 2013.
stimulates the transdifferentiation of type II alveolar epithelial cells in 164. Kho AT, Liu K, Visner G, Martin T, Boudreault F. Identification of
newborn rats. Am J Physiol Lung Cell Mol Physiol 308: L861–L872, dedifferentiation and redevelopment phases during postpneumonectomy
2015. lung growth. Am J Physiol Lung Cell Mol Physiol 305: L542–L554,
144. Hou Y, Liu M, Husted C, Chen C, Thiagarajan K, Johns JL, Rao SP, 2013.
Alvira CM. Activation of the nuclear factor ␬-B pathway during post- 165. Khoshgoo N, Kholdebarin R, Iwasiow BM, Keijzer R. MicroRNAs
natal lung inflammation preserves alveolarization by suppressing macro- and lung development. Pediatr Pulmonol 48: 317–323, 2013.
phage inflammatory protein-2. Am J Physiol Lung Cell Mol Physiol 309: 166. Kim SC, Kellett T, Wang S, Nishi M, Nagre N, Zhou B, Flodby P,
L593–L604, 2015. Shilo K, Ghadiali SN, Takeshima H, Hubmayr RD, Zhao X. TRIM72
145. Hsia CC. Signals and mechanisms of compensatory lung growth. J Appl is required for effective repair of alveolar epithelial cell wounding. Am J
Physiol 97: 1992–1998, 2004. Physiol Lung Cell Mol Physiol 307: L449 –L459, 2014.
146. Huang Y, Kapere Ochieng J, Kempen MB, Munck AB, Swagemak- 167. King G, Damas JE, Cake MH, Berryman D, Maker GL. Influence of
ers S, van Ijcken W, Grosveld F, Tibboel D, Rottier RJ. Hypoxia glucocorticoids, neuregulin-1␤, and sex on surfactant phospholipid se-
inducible factor 3alpha plays a critical role in alveolarization and distal cretion from type II cells. Am J Physiol Lung Cell Mol Physiol 306:
epithelial cell differentiation during mouse lung development. PLoS One L292–L298, 2014.
8: e57695, 2013. 168. Kinsella JP, Abman SH. Inhaled nitric oxide in the premature newborn.
147. Hudak BB, Zhang LY, Kleeberger SR. Inter-strain variation in sus- J Pediatr 151: 10 –15, 2007.
ceptibility to hyperoxic injury of murine airways. Pharmacogenetics 3: 169. Kinsella JP, Greenough A, Abman SH. Bronchopulmonary dysplasia.
135–143, 1993. Lancet 367: 1421–1431, 2006.
170. Knaapi J, Kiviranta R, Laine J, Kaapa P, Lukkarinen H. Cathepsin
148. Huleihel L, Ben-Yehudah A, Milosevic J, Yu G, Pandit K, Sakamoto
K overexpression modifies lung development in newborn mice. Pediatr
K, Yousef H, LeJeune M, Coon TA, Redinger CJ, Chensny L, Manor
Pulmonol 50: 164 –172, 2015.
E, Schatten G, Kaminski N. Let-7d microRNA affects mesenchymal
171. Kobayashi T, Kim H, Liu X, Sugiura H, Kohyama T, Fang Q, Wen
phenotypic properties of lung fibroblasts. Am J Physiol Lung Cell Mol
FQ, Abe S, Wang X, Atkinson JJ, Shipley JM, Senior RM, Rennard
Physiol 306: L534 –L542, 2014.
SI. Matrix metalloproteinase-9 activates TGF-␤ and stimulates fibroblast
149. Hummler SC, Rong M, Chen S, Hehre D, Alapati D, Wu S. Targeting
contraction of collagen gels. Am J Physiol Lung Cell Mol Physiol 306:
glycogen synthase kinase-3beta to prevent hyperoxia-induced lung injury
L1006 –L1015, 2014.
in neonatal rats. Am J Respir Cell Mol Biol 48: 578 –588, 2013.
172. Kool H, Mous D, Tibboel D, de Klein A, Rottier RJ. Pulmonary
150. Hupa KL, Schmiedl A, Pabst R, Von Horsten S, Stephan M. Maternal
vascular development goes awry in congenital lung abnormalities. Birth
deprivation decelerates postnatal morphological lung development of Defects Res C Embryo Today 102: 343–358, 2014.
F344 rats. Anat Rec (Hoboken) 297: 317–326, 2014. 173. Koskinen A, Lukkarinen H, Laine J, Ahotupa M, Kaapa P, Soukka
151. Hussein O, Walters B, Stroetz R, Valencia P, McCall D, Hubmayr H. Delay in rat lung alveolarization after the combined exposure of
RD. Biophysical determinants of alveolar epithelial plasma membrane maternal hyperglycemia and postnatal hyperoxia. Pediatr Pulmonol 49:
wounding associated with mechanical ventilation. Am J Physiol Lung 179 –188, 2014.
Cell Mol Physiol 305: L478 –L484, 2013. 174. Kotton DN, Morrisey EE. Lung regeneration: mechanisms, applications
152. Ito Y, Correll K, Schiel JA, Finigan JH, Prekeris R, Mason RJ. Lung and emerging stem cell populations. Nat Med 20: 822–832, 2014.
fibroblasts accelerate wound closure in human alveolar epithelial cells 175. Kourembanas S. Expanding the pool of stem cell therapy for lung
through hepatocyte growth factor/c-Met signaling. Am J Physiol Lung growth and repair. Circulation 129: 2091–2093, 2014.
Cell Mol Physiol 307: L94 –L105, 2014. 176. Kroon AA, Delriccio V, Tseu I, Kavanagh BP, Post M. Mechanical
153. Jagarapu J, Kelchtermans J, Rong M, Chen S, Hehre D, Hummler S, ventilation-induced apoptosis in newborn rat lung is mediated via FasL/
Faridi MH, Gupta V, Wu S. Leukadherin-1 attenuates hyperoxia- Fas pathway. Am J Physiol Lung Cell Mol Physiol 305: L795–L804,
induced lung injury in neonatal rats. Am J Respir Cell Mol Biol. In press. 2013.
154. Jain D, Bancalari E. Bronchopulmonary dysplasia: clinical perspective. 177. Kroon AA, Wang J, Post M. Alterations in expression of elastogenic
Birth Defects Res A Clin Mol Teratol 100: 134 –144, 2014. and angiogenic genes by different conditions of mechanical ventilation in
155. Jain R, Barkauskas CE, Takeda N, Bowie EJ, Aghajanian H, Wang newborn rat lung. Am J Physiol Lung Cell Mol Physiol 308: L639 –L649,
Q, Padmanabhan A, Manderfield LJ, Gupta M, Li D, Li L, Trivedi 2015.
CM, Hogan BL, Epstein JA. Plasticity of Hopx(⫹) type I alveolar cells 178. Kumarasamy A, Schmitt I, Nave AH, Reiss I, van der Horst I, Dony
to regenerate type II cells in the lung. Nat Commun 6: 6727, 2015. E, Roberts JD Jr, de Krijger RR, Tibboel D, Seeger W, Schermuly
156. James ML, Ross AC, Nicola T, Steele C, Ambalavanan N. VARA RT, Eickelberg O, Morty RE. Lysyl oxidase activity is dysregulated
attenuates hyperoxia-induced impaired alveolar development and lung during impaired alveolarization of mouse and human lungs. Am J Respir
function in newborn mice. Am J Physiol Lung Cell Mol Physiol 304: Crit Care Med 180: 1239 –1252, 2009.
L803–L812, 2013. 179. Kunzmann S, Collins JJ, Kuypers E, Kramer BW. Thrown off
157. Jin Y, Jin Y, Chen B, Tipple TE, Nelin LD. Arginase II is a target of balance: the effect of antenatal inflammation on the developing lung and
miR-17-5p and regulates miR-17-5p expression in human pulmonary immune system. Am J Obstet Gynecol 208: 429 –437, 2013.
artery smooth muscle cells. Am J Physiol Lung Cell Mol Physiol 307: 180. Lacaze-Masmonteil T. That chorioamnionitis is a risk factor for bron-
L197–L204, 2014. chopulmonary dysplasia–the case against. Paediatr Respir Rev 15: 53–
158. Jobe AH. The new bronchopulmonary dysplasia. Curr Opin Pediatr 23: 55, 2014.
167–172, 2011. 181. LaFemina MJ, Sutherland KM, Bentley T, Gonzales LW, Allen L,
159. Jobe AH. What is BPD in 2012 and what will BPD become? Early Hum Chapin CJ, Rokkam D, Sweerus KA, Dobbs LG, Ballard PL, Frank
Dev, 88 Suppl 2: , 2012. JA. Claudin-18 deficiency results in alveolar barrier dysfunction and

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00268.2015 • www.ajplung.org


Perspectives
ADVANCES IN LUNG ALVEOLARIZATION AND BPD L1267
impaired alveologenesis in mice. Am J Respir Cell Mol Biol 51: 550 – 203. Lykkedegn S, Sorensen GL, Beck-Nielsen SS, Christesen HT. The
558, 2014. impact of vitamin D on fetal and neonatal lung maturation. A systematic
182. Lahm T, Tuder RM, Petrache I. Progress in solving the sex hormone review. Am J Physiol Lung Cell Mol Physiol 308: L587–L602, 2015.
paradox in pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 204. Maduekwe ET, Buczynski BW, Yee M, Rangasamy T, Stevens TP,
307: L7–L26, 2014. Lawrence BP, O’Reilly MA. Cumulative neonatal oxygen exposure
183. Lal CV, Schwarz MA. Vascular mediators in chronic lung disease of predicts response of adult mice infected with influenza A virus. Pediatr
infancy: role of endothelial monocyte activating polypeptide II (EMAP Pulmonol. In press.
II). Birth Defects Res A Clin Mol Teratol 100: 180 –188, 2014. 205. Madurga A, Golec A, Pozarska A, Ishii I, Mižíková I, Nardiello C,
184. Lange AW, Haitchi HM, LeCras TD, Sridharan A, Xu Y, Wert SE, Vadasz I, Herold S, Mayer K, Reichenberger F, Fehrenbach H,
James J, Udell N, Thurner PJ, Whitsett JA. Sox17 is required for Seeger W, Morty RE. The H2S-generating enzymes cystathionine
normal pulmonary vascular morphogenesis. Dev Biol 387: 109 –120, ␤-synthase and cystathionine ␥-lyase play a role in vascular development
2014. during normal lung alveolarization. Am J Physiol Lung Cell Mol Physiol
185. Lawson WE, Blackwell TS. ␤-Catenin and CCNs in lung epithelial 309: L710 –L724, 2015.
repair. Am J Physiol Lung Cell Mol Physiol 304: L579 –L581, 2013. 206. Madurga A, Mižíková I, Ruiz-Camp J, Morty RE. Recent advances in
186. Lee AH, Dhaliwal R, Kantores C, Ivanovska J, Gosal K, McNamara late lung development and the pathogenesis of bronchopulmonary dys-
PJ, Letarte M, Jankov RP. Rho-kinase inhibitor prevents bleomycin- plasia. Am J Physiol Lung Cell Mol Physiol 305: L893–L905, 2013.
induced injury in neonatal rats independent of effects on lung inflamma- 207. Madurga A, Mižíková I, Ruiz-Camp J, Vadász I, Herold S, Mayer K,
tion. Am J Respir Cell Mol Biol 50: 61–73, 2014. Fehrenbach H, Seeger W, Morty RE. Systemic hydrogen sulfide
187. Lee HJ, Lee YJ, Choi CW, Lee JA, Kim EK, Kim HS, Kim BI, Choi administration partially restores normal alveolarization in an experimen-
JH. Rosiglitazone, a peroxisome proliferator-activated receptor-gamma tal animal model of bronchopulmonary dysplasia. Am J Physiol Lung
agonist, restores alveolar and pulmonary vascular development in a rat Cell Mol Physiol 306: L684 –L697, 2014.
model of bronchopulmonary dysplasia. Yonsei Med J 55: 99 –106, 2014. 208. Mahoney JE, Mori M, Szymaniak AD, Varelas X, Cardoso WV. The
188. Lee MK, Smith SM, Banerjee MM, Li C, Minoo P, Volpe MV, hippo pathway effector Yap controls patterning and differentiation of
Nielsen HC. The p66Shc adapter protein regulates the morphogenesis airway epithelial progenitors. Dev Cell 30: 137–150, 2014.
and epithelial maturation of fetal mouse lungs. Am J Physiol Lung Cell 209. Mailleux AA, Moshai EF, Crestani B. Sonic Hedgehog signaling in
Mol Physiol 306: L316 –L325, 2014. pulmonary fibrosis: a spiky issue? Am J Physiol Lung Cell Mol Physiol
189. Lewis KJ, Tibbitt MW, Zhao Y, Branchfield K, Sun X, Balasubra- 304: L391–L393, 2013.
maniam V, Anseth KS. In vitro model alveoli from photodegradable 210. Malpighi M. De Pulmonibus Epistola Altera. Bononiae, Typis Jo:
microsphere templates. Biomater Sci 3: 821–832, 2015. Baptistae Ferronii, 1661.
190. Li C, Li M, Li S, Xing Y, Yang CY, Li A, Borok Z, De Langhe S, 211. Malpighi M. De Pulmonibus Observationes Anatomicae. Bononiae.
Minoo P. Progenitors of secondary crest myofibroblasts are developmen- Typis Jo: Baptistae Ferronii, 1661.
tally committed in early lung mesoderm. Stem Cells 33: 999 –1012, 2015. 212. Mammoto T, Jiang E, Jiang A, Mammoto A. Extracellular matrix
191. Li C, Mpollo MS, Gonsalves CS, Tahara SM, Malik P, Kalra VK.
structure and tissue stiffness control postnatal lung development through
Peroxisome proliferator-activated receptor-alpha-mediated transcription
the lipoprotein receptor-related protein 5/Tie2 signaling system. Am J
of miR-199a2 attenuates endothelin-1 expression via hypoxia-inducible
Respir Cell Mol Biol 49: 1009 –1018, 2013.
factor-1alpha. J Biol Chem 289: 36031–36047, 2014.
213. Mandell E, Seedorf G, Gien J, Abman SH. Vitamin D treatment
192. Li J, Masood A, Yi M, Lau M, Belcastro R, Ivanovska J, Jankov RP,
improves survival and infant lung structure after intra-amniotic endotoxin
Tanswell AK. The IGF-I/IGF-R1 pathway regulates postnatal lung
exposure in rats: potential role for the prevention of bronchopulmonary
growth and is a nonspecific regulator of alveologenesis in the neonatal
dysplasia. Am J Physiol Lung Cell Mol Physiol 306: L420 –L428, 2014.
rat. Am J Physiol Lung Cell Mol Physiol 304: L626 –L637, 2013.
214. Mangalmurti NS, Friedman JL, Wang LC, Stolz D, Muthukumaran
193. Lim R, Fedulov AV, Kobzik L. Maternal stress during pregnancy
G, Siegel DL, Schmidt AM, Lee JS, Albelda SM. The receptor for
increases neonatal allergy susceptibility: role of glucocorticoids. Am J
Physiol Lung Cell Mol Physiol 307: L141–L148, 2014. advanced glycation end products mediates lung endothelial activation by
194. Lim R, Muljadi R, Koulaeva E, Vosdoganes P, Chan ST, Acharya R, RBCs. Am J Physiol Lung Cell Mol Physiol 304: L250 –L263, 2013.
Gurusinghe S, Ritvos O, Pasternack A, Wallace EM. Activin A 215. Manzano RM, Mascaretti RS, Carrer V, Haddad LB, Fernandes AR,
contributes to the development of hyperoxia-induced lung injury in Reyes AM, Rebello CM. A hyperoxic lung injury model in premature
neonatal mice. Pediatr Res 77: 749 –756, 2015. rabbits: the influence of different gestational ages and oxygen concen-
195. Limjunyawong N, Fallica J, Horton MR, Mitzner W. Measurement of trations. PLoS One 9: e95844, 2014.
the pressure-volume curve in mouse lungs. J Vis Exp: 52376, 2015. 216. Mao Q, Chu S, Ghanta S, Padbury JF, De Paepe ME. Ex vivo
196. Lingappan K, Jiang W, Wang L, Couroucli XI, Barrios R, Moorthy expanded human cord blood-derived hematopoietic progenitor cells in-
B. Sex-specific differences in hyperoxic lung injury in mice: implications duce lung growth and alveolarization in injured newborn lungs. Respir
for acute and chronic lung disease in humans. Toxicol Appl Pharmacol Res 14: 37, 2013.
272: 281–290, 2013. 217. Martin CR, Zaman MM, Gilkey C, Salguero MV, Hasturk H,
197. Liu S, Young SM, Varisco BM. Dynamic expression of chymotrypsin- Kantarci A, Van Dyke TE, Freedman SD. Resolvin D1 and lipoxin A4
like elastase 1 over the course of murine lung development. Am J Physiol improve alveolarization and normalize septal wall thickness in a neonatal
Lung Cell Mol Physiol 306: L1104 –L1116, 2014. murine model of hyperoxia-induced lung injury. PLoS One 9: e98773,
198. Lizotte PP, Hanford LE, Enghild JJ, Nozik-Grayck E, Giles BL, 2014.
Oury TD. Developmental expression of the receptor for advanced 218. Martin RJ, Fanaroff AA. The preterm lung and airway: past, present,
glycation end-products (RAGE) and its response to hyperoxia in the and future. Pediatr Neonatol 54: 228 –234, 2013.
neonatal rat lung. BMC Dev Biol 7: 15, 2007. 219. Martin YN, Manlove L, Dong J, Carey WA, Thompson MA, Pa-
199. Londhe VA, Maisonet TM, Lopez B, Shin BC, Huynh J, Devaskar belick CM, Pandya HC, Martin RJ, Wigle DA, Prakash YS. Hyper-
SU. Retinoic acid rescues alveolar hypoplasia in the calorie-restricted oxia-induced changes in estradiol metabolism in postnatal airway smooth
developing rat lung. Am J Respir Cell Mol Biol 48: 179 –187, 2013. muscle. Am J Physiol Lung Cell Mol Physiol 308: L141–L146, 2015.
200. Lu K, Reddy R, Berika M, Warburton D, El-Hashash AH. Abroga- 220. Masood A, Yi M, Lau M, Belcastro R, Li J, Kantores C, Pace-Asciak
tion of Eya1/Six1 disrupts the saccular phase of lung morphogenesis and CR, Jankov RP, Tanswell AK. Cyclooxygenase-2 inhibition partially
causes remodeling. Dev Biol 382: 110 –123, 2013. protects against 60% O2 -mediated lung injury in neonatal rats. Pediatr
201. Luo Y, El Agha E, Turcatel G, Chen H, Chiu J, Warburton D, Pulmonol 49: 991–1002, 2014.
Bellusci S, Qian BP, Menke DB, Shi W. Mesenchymal adenomatous 221. MasoudiMotlagh M, Sepehr R, Sheibani N, Sorenson CM, Ranji M.
polyposis coli plays critical and diverse roles in regulating lung devel- Optical cryoimaging of mitochondrial redox state in bronchopulmonary-
opment. BMC Biol 13: 42, 2015. dysplasia injury models in mice lungs. Quant Imaging Med Surg 5:
202. Lv Y, Tang LL, Wei JK, Xu XF, Gu W, Fu LC, Zhang LY, Du LZ. 159 –162, 2015.
Decreased Kv1.5 expression in intrauterine growth retardation rats with 222. Mayor RS, Finch KE, Zehr J, Morselli E, Neinast MD, Frank AP,
exaggerated pulmonary hypertension. Am J Physiol Lung Cell Mol Hahner LD, Wang J, Rakheja D, Palmer BF, Rosenfeld CR, Savani
Physiol 305: L856 –L865, 2013. RC, Clegg DJ. Maternal high fat diet is associated with impaired fetal

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00268.2015 • www.ajplung.org


Perspectives
L1268 ADVANCES IN LUNG ALVEOLARIZATION AND BPD

lung development. Am J Physiol Lung Cell Mol Physiol 309: L360 – Z, Whitsett JA, Gail D, Blaisdell CJ, Lin QS. Molecular determinants
L368, 2015. of lung development. Ann Am Thorac Soc 10: S12–S16, 2013.
223. McCulley D, Wienhold M, Sun X. The pulmonary mesenchyme directs 243. Morty RE, Königshoff M, Eickelberg O. Transforming growth factor-
lung development. Curr Opin Genet Dev 32: 98 –105, 2015. beta signaling across ages: from distorted lung development to chronic
224. McDougall AR, Hooper SB, Zahra VA, Cole TJ, Lo CY, Doran T, obstructive pulmonary disease. Proc Am Thorac Soc 6: 607–613, 2009.
Wallace MJ. Trop2 regulates motility and lamellipodia formation in 244. Morty RE, Matalon S. Real-time visualization of lung function: from
cultured fetal lung fibroblasts. Am J Physiol Lung Cell Mol Physiol 305: micro to macro. Am J Physiol Lung Cell Mol Physiol 304: L1–L3, 2013.
L508 –L521, 2013. 245. Mourani PM, Abman SH. Pulmonary vascular disease in bronchopul-
225. McDougall AR, Tolcos M, Hooper SB, Cole TJ, Wallace MJ. Trop2: monary dysplasia: pulmonary hypertension and beyond. Curr Opin
from development to disease. Dev Dyn 244: 99 –109, 2015. Pediatr 25: 329 –337, 2013.
226. McGowan SE. Paracrine cellular and extracellular matrix interactions 246. Mühlfeld C, Hegermann J, Wrede C, Ochs M. A review of recent
with mesenchymal progenitors during pulmonary alveolar septation. developments and applications of morphometry/stereology in lung re-
Birth Defects Res A Clin Mol Teratol 100: 227–239, 2014. search. Am J Physiol Lung Cell Mol Physiol 309: L526 –L536, 2015.
227. McGowan SE, McCoy DM. Platelet-derived growth factor-A and Sonic 247. Mühlfeld C, Knudsen L, Ochs M. Stereology and morphometry of lung
Hedgehog signaling direct lung fibroblast precursors during alveolar tissue. Methods Mol Biol 931: 367–390, 2013.
septal formation. Am J Physiol Lung Cell Mol Physiol 305: L229 –L239, 248. Mühlfeld C, Ochs M. Quantitative microscopy of the lung: a problem-
2013. based approach. Part 2: Stereological parameters and study designs in
228. McGowan SE, McCoy DM. Regulation of fibroblast lipid storage and various diseases of the respiratory tract. Am J Physiol Lung Cell Mol
myofibroblast phenotypes during alveolar septation in mice. Am J Physiol 305: L205–L221, 2013.
Physiol Lung Cell Mol Physiol 307: L618 –L631, 2014. 249. Mujahid S, Logvinenko T, Volpe MV, Nielsen HC. miRNA regulated
229. McGrath-Morrow SA, Hayashi M, Aherrera A, Lopez A, Malinina pathways in late stage murine lung development. BMC Dev Biol 13: 13,
A, Collaco JM, Neptune E, Klein JD, Winickoff JP, Breysse P, 2013.
Lazarus P, Chen G. The effects of electronic cigarette emissions on 250. Murphy SR, Oslund KL, Hyde DM, Miller LA, Van Winkle LS,
systemic cotinine levels, weight and postnatal lung growth in neonatal Schelegle ES. Ozone-induced airway epithelial cell death, the neuro-
mice. PLoS One 10: e0118344, 2015. kinin-1 receptor pathway, and the postnatal developing lung. Am J
230. McGrath-Morrow SA, Lauer T, Collaco JM, Lopez A, Malhotra D, Physiol Lung Cell Mol Physiol 307: L471–L481, 2014.
Alekseyev YO, Neptune E, Wise R, Biswal S. Transcriptional responses 251. Narasaraju T, Shukla D, More S, Huang C, Zhang L, Xiao X, Liu L.
of neonatal mouse lung to hyperoxia by Nrf2 status. Cytokine 65: 4 –9, Role of microRNA-150 and glycoprotein nonmetastatic melanoma pro-
2014. tein B in angiogenesis during hyperoxia-induced neonatal lung injury.
231. McKenna S, Michaelis KA, Agboke F, Liu T, Han K, Yang G, Am J Respir Cell Mol Biol 52: 253–261, 2015.
Dennery PA, Wright CJ. Sustained hyperoxia-induced NF-␬B activa- 252. Narayanan M, Beardsmore CS, Owers-Bradley J, Dogaru CM,
tion improves survival and preserves lung development in neonatal mice. Mada M, Ball I, Garipov RR, Kuehni CE, Spycher BD, Silverman M.
Am J Physiol Lung Cell Mol Physiol 306: L1078 –L1089, 2014.
Catch-up alveolarization in ex-preterm children: evidence from (3)He
232. McKleroy W, Lee TH, Atabai K. Always cleave up your mess:
magnetic resonance. Am J Respir Crit Care Med 187: 1104 –1109, 2013.
targeting collagen degradation to treat tissue fibrosis. Am J Physiol Lung
253. Nave AH, Mižíková I, Niess G, Steenbock H, Reichenberger F,
Cell Mol Physiol 304: L709 –L721, 2013.
Talavera ML, Veit F, Herold S, Mayer K, Vadász I, Weissmann N,
233. McQualter JL, McCarty RC, Van der Velden J, O’Donoghue RJ,
Seeger W, Brinckmann J, Morty RE. Lysyl oxidases play a causal role
Asselin-Labat ML, Bozinovski S, Bertoncello I. TGF-beta signaling in
in vascular remodeling in clinical and experimental pulmonary arterial
stromal cells acts upstream of FGF-10 to regulate epithelial stem cell
hypertension. Arterioscler Thromb Vasc Biol 34: 1446 –1458, 2014.
growth in the adult lung. Stem Cell Res 11: 1222–1233, 2013.
254. Nichols JL, Gladwell W, Verhein KC, Cho HY, Wess J, Suzuki O,
234. Mehta A, Dobersch S, Dammann RH, Bellusci S, Ilinskaya ON,
Braun T, Barreto G. Validation of Tuba1a as appropriate internal Wiltshire T, Kleeberger SR. Genome-wide association mapping of
control for normalization of gene expression analysis during mouse lung acute lung injury in neonatal inbred mice. FASEB J 28: 2538 –2550,
development. Int J Mol Sci 16: 4492–4511, 2015. 2014.
235. Minoo P, Wolkenhauer O, Guttentag S. Systems biology and pediatric 255. Nickles HT, Sumkauskaite M, Wang X, Wegner I, Puderbach M,
research. Pediatr Res 73: 499 –501, 2013. Kuebler WM. Mechanical ventilation causes airway distension with
236. Miranda LF, Rodrigues CO, Ramachandran S, Torres E, Huang J, proinflammatory sequelae in mice. Am J Physiol Lung Cell Mol Physiol
Klim J, Hehre D, McNiece I, Hare JM, Suguihara CY, Young KC. 307: L27–L37, 2014.
Stem cell factor improves lung recovery in rats following neonatal 256. Nijmeh H, Balasubramaniam V, Burns N, Ahmad A, Stenmark KR,
hyperoxia-induced lung injury. Pediatr Res 74: 682–688, 2013. Gerasimovskaya EV. High proliferative potential endothelial colony-
237. Mirza H, Ziegler J, Ford S, Padbury J, Tucker R, Laptook A. forming cells contribute to hypoxia-induced pulmonary artery vasa va-
Pulmonary hypertension in preterm infants: prevalence and association sorum neovascularization. Am J Physiol Lung Cell Mol Physiol 306:
with bronchopulmonary dysplasia. J Pediatr 165: 909 –914 e901, 2014. L661–L671, 2014.
238. Mižíková I, Ruiz-Camp J, Steenbock H, Madurga A, Vadász I, 257. Nold MF, Mangan NE, Rudloff I, Cho SX, Shariatian N, Samaras-
Herold S, Mayer K, Seeger W, Brinckmann J, Morty RE. Collagen inghe TD, Skuza EM, Pedersen J, Veldman A, Berger PJ, Nold-Petry
and elastin cross-linking is altered during aberrant late lung development CA. Interleukin-1 receptor antagonist prevents murine bronchopulmo-
associated with hyperoxia. Am J Physiol Lung Cell Mol Physiol 308: nary dysplasia induced by perinatal inflammation and hyperoxia. Proc
L1145–L1158, 2015. Natl Acad Sci USA 110: 14384 –14389, 2013.
239. Monz D, Tutdibi E, Mildau C, Shen J, Kasoha M, Laschke MW, 258. Northway WH Jr, Rosan RC, Porter DY. Pulmonary disease following
Roolfs T, Schmiedl A, Tschernig T, Bieback K, Gortner L. Human respirator therapy of hyaline-membrane disease. Bronchopulmonary dys-
umbilical cord blood mononuclear cells in a double-hit model of bron- plasia. N Engl J Med 276: 357–368, 1967.
chopulmonary dysplasia in neonatal mice. PLoS One 8: e74740, 2013. 259. Ntokou A, Klein F, Dontireddy D, Becker S, Bellusci S, Richardson
240. Morales E, Sakurai R, Husain S, Paek D, Gong M, Ibe B, Li Y, WD, Szibor M, Braun T, Morty RE, Seeger W, Voswinckel R,
Husain M, Torday JS, Rehan VK. Nebulized PPARgamma agonists: a Ahlbrecht K. Characterization of the platelet-derived growth factor
novel approach to augment neonatal lung maturation and injury repair in receptor ␣-positive cell lineage during murine late lung development. Am
rats. Pediatr Res 75: 631–640, 2014. J Physiol Lung Cell Mol Physiol. In press.
241. Mori M, Mahoney JE, Stupnikov MR, Paez-Cortez JR, Szymaniak 260. Null DM, Alvord J, Leavitt W, Wint A, Dahl MJ, Presson AP, Lane
AD, Varelas X, Herrick DB, Schwob J, Zhang H, Cardoso WV. RH, DiGeronimo RJ, Yoder BA, Albertine KH. High-frequency nasal
Notch3-Jagged signaling controls the pool of undifferentiated airway ventilation for 21 d maintains gas exchange with lower respiratory
progenitors. Development 142: 258 –267, 2015. pressures and promotes alveolarization in preterm lambs. Pediatr Res 75:
242. Morrisey EE, Cardoso WV, Lane RH, Rabinovitch M, Abman SH, 507–516, 2014.
Ai X, Albertine KH, Bland RD, Chapman HA, Checkley W, Epstein 261. O’Reilly M, Hansbro PM, Horvat JC, Beckett EL, Harding R, Sozo
JA, Kintner CR, Kumar M, Minoo P, Mariani TJ, McDonald DM, F. Bronchiolar remodeling in adult mice following neonatal exposure to
Mukouyama YS, Prince LS, Reese J, Rossant J, Shi W, Sun X, Werb hyperoxia: relation to growth. Anat Rec (Hoboken) 297: 758 –769, 2014.

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00268.2015 • www.ajplung.org


Perspectives
ADVANCES IN LUNG ALVEOLARIZATION AND BPD L1269
262. O’Reilly M, Harding R, Sozo F. Altered small airways in aged mice required for lung branching morphogenesis and alveolarization. Devel-
following neonatal exposure to hyperoxic gas. Neonatology 105: 39 –45, opment 141: 4751–4762, 2014.
2014. 282. Podolin PL, Foley JP, Carpenter DC, Bolognese BJ, Logan GA, Long
263. O’Reilly M, Sozo F, Harding R. Impact of preterm birth and broncho- E 3rd, Harrison OJ, Walsh PT. T cell depletion protects against
pulmonary dysplasia on the developing lung: long-term consequences for alveolar destruction due to chronic cigarette smoke exposure in mice. Am
respiratory health. Clin Exp Pharmacol Physiol 40: 765–773, 2013. J Physiol Lung Cell Mol Physiol 304: L312–L323, 2013.
264. O’Reilly M, Thébaud B. Animal models of bronchopulmonary dyspla- 283. Poonyagariyagorn HK, Metzger S, Dikeman D, Mercado AL, Ma-
sia. The term rat models. Am J Physiol Lung Cell Mol Physiol 307: linina A, Calvi C, McGrath-Morrow S, Neptune ER. Superoxide
L948 –L958, 2014. dismutase 3 dysregulation in a murine model of neonatal lung injury. Am
265. O’Reilly M, Thébaud B. The promise of stem cells in bronchopulmo- J Respir Cell Mol Biol 51: 380 –390, 2014.
nary dysplasia. Semin Perinatol 37: 79 –84, 2013. 284. Popova AP, Bentley JK, Cui TX, Richardson MN, Linn MJ, Lei J,
266. Ochieng JK, Schilders K, Kool H, Boerema-De Munck A, Buscop- Chen Q, Goldsmith AM, Pryhuber GS, Hershenson MB. Reduced
Van Kempen M, Gontan C, Smits R, Grosveld FG, Wijnen RM, platelet-derived growth factor receptor expression is a primary feature of
Tibboel D, Rottier RJ. Sox2 regulates the emergence of lung basal cells human bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol
by directly activating the transcription of Trp63. Am J Respir Cell Mol Physiol 307: L231–L239, 2014.
Biol 51: 311–322, 2014. 285. Porzionato A, Zaramella P, Macchi V, Sarasin G, Di Giulio C, Rigon
267. Ochs M, Mühlfeld C. Quantitative microscopy of the lung: a problem- A, Grisafi D, Dedja A, Chiandetti L, De Caro R. Cyclosporine and
based approach. Part 1: Basic principles of lung stereology. Am J Physiol hyperoxia-induced lung damage in neonatal rats. Respir Physiol Neuro-
Lung Cell Mol Physiol 305: L15–L22, 2013. biol 187: 41–46, 2013.
268. Oka T, Xu J, Kaiser RA, Melendez J, Hambleton M, Sargent MA, 286. Prakash YS. Airway smooth muscle in airway reactivity and remodel-
Lorts A, Brunskill EW, Dorn GW 2nd, Conway SJ, Aronow BJ, ing: what have we learned? Am J Physiol Lung Cell Mol Physiol 305:
Robbins J, Molkentin JD. Genetic manipulation of periostin expression L912–L933, 2013.
reveals a role in cardiac hypertrophy and ventricular remodeling. Circ 287. Prakash YS, Matalon S. Nanoparticles and the lung: friend or foe? Am
Res 101: 313–321, 2007. J Physiol Lung Cell Mol Physiol 306: L393–L396, 2014.
269. Papin JF, Wolf RF, Kosanke SD, Jenkins JD, Moore SN, Anderson 288. Raffay TM, Locy ML, Hill CL, Jindal NS, Rogers LK, Welty SE,
MP, Welliver RC Sr. Infant baboons infected with respiratory syncytial Tipple TE. Neonatal hyperoxic exposure persistently alters lung secre-
virus develop clinical and pathological changes that parallel those of toglobins and annexin A1. Biomed Res Int 2013: 408485, 2013.
human infants. Am J Physiol Lung Cell Mol Physiol 304: L530 –L539, 289. Rafii S, Cao Z, Lis R, Siempos II, Chavez D, Shido K, Rabbany SY,
2013. Ding BS. Platelet-derived SDF-1 primes the pulmonary capillary vascu-
270. Park HS, Park JW, Kim HJ, Choi CW, Lee HJ, Kim BI, Chun YS. lar niche to drive lung alveolar regeneration. Nat Cell Biol 17: 123–136,
Sildenafil alleviates bronchopulmonary dysplasia in neonatal rats by 2015.
activating the hypoxia-inducible factor signaling pathway. Am J Respir 290. Rajavelu P, Chen G, Xu Y, Kitzmiller JA, Korfhagen TR, Whitsett
Cell Mol Biol 48: 105–113, 2013. JA. Airway epithelial SPDEF integrates goblet cell differentiation and
271. Parra-Robles J, Wild JM. On the use of 3He diffusion magnetic pulmonary Th2 inflammation. J Clin Invest 125: 2021–2031, 2015.
resonance as evidence of neo-alveolarization during childhood and ado- 291. Ramachandran S, Krishnamurthy S, Jacobi AM, Wohlford-Lenane
lescence. Am J Respir Crit Care Med 189: 501–502, 2014. C, Behlke MA, Davidson BL, McCray PB Jr. Efficient delivery of
272. Patel RM, Leong T, Carlton DP, Vyas-Read S. Early caffeine therapy RNA interference oligonucleotides to polarized airway epithelia in vitro.
and clinical outcomes in extremely preterm infants. J Perinatol 33: Am J Physiol Lung Cell Mol Physiol 305: L23–L32, 2013.
134 –140, 2013. 292. Ramachandran S, Suguihara C, Drummond S, Chatzistergos K,
273. Peng T, Morrisey EE. Development of the pulmonary vasculature: Klim J, Torres E, Huang J, Hehre D, Rodrigues CO, McNiece IK,
Current understanding and concepts for the future. Pulm Circ 3: 176 – Hare JM, Young KC. Bone marrow-derived c-kit⫹ cells attenuate
178, 2013. neonatal hyperoxia-induced lung injury. Cell Transplant 24: 85–95,
274. Pereira-Terra P, Kholdebarin R, Higgins M, Iwasiow BM, Correia- 2015.
Pinto J, Keijzer R. Lower NPAS3 expression during the later stages of 293. Ramani M, Bradley WE, Dell’Italia LJ, Ambalavanan N. Early
abnormal lung development in rat congenital diaphragmatic hernia. exposure to hyperoxia or hypoxia adversely impacts cardiopulmonary
Pediatr Surg Int 31: 659 –663, 2015. development. Am J Respir Cell Mol Biol 52: 594 –602, 2015.
275. Pérez-Rial S, Del Puerto-Nevado L, Girón-Martínez Á, Terrón- 294. Ratner V, Sosunov SA, Niatsetskaya ZV, Utkina-Sosunova IV, Ten
Expósito R, Díaz-Gil JJ, González-Mangado N, Peces-Barba G. Liver VS. Mechanical ventilation causes pulmonary mitochondrial dysfunction
growth factor treatment reverses emphysema previously established in a and delayed alveolarization in neonatal mice. Am J Respir Cell Mol Biol
cigarette smoke exposure mouse model. Am J Physiol Lung Cell Mol 49: 943–950, 2013.
Physiol 307: L718 –L726, 2014. 295. Ravikumar P, Ye J, Zhang J, Pinch SN, Hu MC, Kuro-o M, Hsia CC,
276. Pham H, Vottier G, Pansiot J, Duong-Quy S, Bollen B, Dalous J, Moe OW. ␣-Klotho protects against oxidative damage in pulmonary
Gallego J, Mercier JC, Dinh-Xuan AT, Bonnin P, Charriaut-Mar- epithelia. Am J Physiol Lung Cell Mol Physiol 307: L566 –L575, 2014.
langue C, Baud O. Inhaled NO prevents hyperoxia-induced white matter 296. Razavi H, Stewart SE, Xu C, Sawada H, Zarafshar SY, Taylor CA,
damage in neonatal rats. Exp Neurol 252: 114 –123, 2014. Rabinovitch M, Feinstein JA. Chronic effects of pulmonary artery
277. Pieretti AC, Ahmed AM, Roberts JD Jr, Kelleher CM. A novel in stenosis on hemodynamic and structural development of the lungs. Am J
vitro model to study alveologenesis. Am J Respir Cell Mol Biol 50: Physiol Lung Cell Mol Physiol 304: L17–L28, 2013.
459 –469, 2014. 297. Rehan VK, Liu J, Sakurai R, Torday JS. Perinatal nicotine-induced
278. Pierro M, Ionescu L, Montemurro T, Vadivel A, Weissmann G, transgenerational asthma. Am J Physiol Lung Cell Mol Physiol 305:
Oudit G, Emery D, Bodiga S, Eaton F, Peault B, Mosca F, Lazzari L, L501–L507, 2013.
Thebaud B. Short-term, long-term and paracrine effect of human um- 298. Rehan VK, Sugano S, Wang Y, Santos J, Romero S, Dasgupta C,
bilical cord-derived stem cells in lung injury prevention and repair in Keane MP, Stahlman MT, Torday JS. Evidence for the presence of
experimental bronchopulmonary dysplasia. Thorax 68: 475–484, 2013. lipofibroblasts in human lung. Exp Lung Res 32: 379 –393, 2006.
279. Pietrzyk JJ, Kwinta P, Wollen EJ, Bik-Multanowski M, Madetko- 299. Richter J, Toelen J, Vanoirbeek J, Kakigano A, Dekoninck P,
Talowska A, Gunther CC, Jagla M, Tomasik T, Saugstad OD. Gene Verbeken E, Deprest J. Functional assessment of hyperoxia-induced
expression profiling in preterm infants: new aspects of bronchopulmo- lung injury after preterm birth in the rabbit. Am J Physiol Lung Cell Mol
nary dysplasia development. PLoS One 8: e78585, 2013. Physiol 306: L277–L283, 2014.
280. Pisarcik S, Maylor J, Lu W, Yun X, Undem C, Sylvester JT, 300. Rieger-Fackeldey E, Park MS, Schanbacher BL, Joshi MS, Chicoine
Semenza GL, Shimoda LA. Activation of hypoxia-inducible factor-1 in LG, Nelin LD, Bauer JA, Welty SE, Smith CV. Lung development
pulmonary arterial smooth muscle cells by endothelin-1. Am J Physiol alterations in newborn mice after recovery from exposure to sublethal
Lung Cell Mol Physiol 304: L549 –L561, 2013. hyperoxia. Am J Pathol 184: 1010 –1016, 2014.
281. Plosa EJ, Young LR, Gulleman PM, Polosukhin VV, Zaynagetdinov 301. Rios H, Koushik SV, Wang H, Wang J, Zhou HM, Lindsley A,
R, Benjamin JT, Im AM, van der Meer R, Gleaves LA, Bulus N, Han Rogers R, Chen Z, Maeda M, Kruzynska-Frejtag A, Feng JQ,
W, Prince LS, Blackwell TS, Zent R. Epithelial beta1 integrin is Conway SJ. Periostin null mice exhibit dwarfism, incisor enamel de-

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00268.2015 • www.ajplung.org


Perspectives
L1270 ADVANCES IN LUNG ALVEOLARIZATION AND BPD

fects, and an early-onset periodontal disease-like phenotype. Mol Cell 321. Sun H, Choo-Wing R, Fan J, Leng L, Syed MA, Hare AA, Jorgensen
Biol 25: 11131–11144, 2005. WL, Bucala R, Bhandari V. Small molecular modulation of macro-
302. Rossor T, Greenough A. Advances in paediatric pulmonary vascular phage migration inhibitory factor in the hyperoxia-induced mouse model
disease associated with bronchopulmonary dysplasia. Expert Rev Respir of bronchopulmonary dysplasia. Respir Res 14: 27, 2013.
Med 9: 35–43, 2015. 322. Sun H, Choo-Wing R, Sureshbabu A, Fan J, Leng L, Yu S, Jiang D,
303. Roth-Kleiner M, Berger TM, Gremlich S, Tschanz SA, Mund SI, Noble P, Homer RJ, Bucala R, Bhandari V. A critical regulatory role
Post M, Stampanoni M, Schittny JC. Neonatal steroids induce a for macrophage migration inhibitory factor in hyperoxia-induced injury
down-regulation of tenascin-C and elastin and cause a deceleration of the in the developing murine lung. PLoS One 8: e60560, 2013.
first phase and an acceleration of the second phase of lung alveolariza- 323. Sureshbabu A, Syed MA, Boddupalli CS, Dhodapkar MV, Homer
tion. Histochem Cell Biol 141: 75–84, 2014. RJ, Minoo P, Bhandari V. Conditional overexpression of TGFbeta1
304. Rozycki HJ. Potential contribution of type I alveolar epithelial cells to promotes pulmonary inflammation, apoptosis and mortality via TGF-
chronic neonatal lung disease. Front Pediatr 2: 45, 2014. betaR2 in the developing mouse lung. Respir Res 16: 4, 2015.
305. Sakurai R, Villarreal P, Husain S, Liu J, Sakurai T, Tou E, Torday 324. Sutsko RP, Young KC, Ribeiro A, Torres E, Rodriguez M, Hehre D,
JS, Rehan VK. Curcumin protects the developing lung against long-term Devia C, McNiece I, Suguihara C. Long-term reparative effects of
hyperoxic injury. Am J Physiol Lung Cell Mol Physiol 305: L301–L311, mesenchymal stem cell therapy following neonatal hyperoxia-induced
2013. lung injury. Pediatr Res 73: 46 –53, 2013.
306. Salaets T, Richter J, Brady P, Jimenez J, Nagatomo T, Deprest J,
325. Suzuki T, Suzuki S, Fujino N, Ota C, Yamada M, Suzuki T, Yamaya
Toelen J. Transcriptome analysis of the preterm rabbit lung after seven
M, Kondo T, Kubo H. c-Kit immunoexpression delineates a putative
days of hyperoxic exposure. PLoS One 10: e0136569, 2015.
endothelial progenitor cell population in developing human lungs. Am J
307. Sallon C, Soulet D, Provost PR, Tremblay Y. Automated high perfor-
Physiol Lung Cell Mol Physiol 306: L855–L865, 2014.
mance analysis of lung morphometry. Am J Respir Cell Mol Biol 53:
326. Syed MA, Bhandari V. Hyperoxia exacerbates postnatal inflammation-
149 –158, 2015.
308. Schmidt B, Roberts RS, Davis P, Doyle LW, Barrington KJ, Ohlsson induced lung injury in neonatal BRP-39 null mutant mice promoting the
A, Solimano A, Tin W; Caffeine for Apnea of Prematurity Trial M1 macrophage phenotype. Mediators Inflamm 2013: 457189, 2013.
Group. Long-term effects of caffeine therapy for apnea of prematurity. 327. Tahedl D, Wirkes A, Tschanz SA, Ochs M, Muhlfeld C. How
N Engl J Med 357: 1893–1902, 2007. common is the lipid body-containing interstitial cell in the mammalian
309. Schneider JP, Ochs M. Alterations of mouse lung tissue dimensions lung? Am J Physiol Lung Cell Mol Physiol 307: L386 –L394, 2014.
during processing for morphometry: a comparison of methods. Am J 328. Tan W, Madhavan K, Hunter KS, Park D, Stenmark KR. Vascular
Physiol Lung Cell Mol Physiol 306: L341–L350, 2014. stiffening in pulmonary hypertension: cause or consequence? (2013
310. Schulz MH, Pandit KV, Lino Cardenas CL, Ambalavanan N, Ka- Grover Conference Series). Pulm Circ 4: 560 –580, 2014.
minski N, Bar-Joseph Z. Reconstructing dynamic microRNA-regulated 329. Tang JR, Michaelis KA, Nozik-Grayck E, Seedorf GJ, Hartman-
interaction networks. Proc Natl Acad Sci USA 110: 15686 –15691, 2013. Filson M, Abman SH, Wright CJ. The NF-kappaB inhibitory proteins
311. Schumacker PT, Gillespie MN, Nakahira K, Choi AM, Crouser ED, IkappaBalpha and IkappaBbeta mediate disparate responses to inflam-
Piantadosi CA, Bhattacharya J. Mitochondria in lung biology and mation in fetal pulmonary endothelial cells. J Immunol 190: 2913–2923,
pathology: more than just a powerhouse. Am J Physiol Lung Cell Mol 2013.
Physiol 306: L962–L974, 2014. 330. Teng RJ, Du J, Afolayan AJ, Eis A, Shi Y, Konduri GG. AMP kinase
312. Schwartze JT, Becker S, Sakkas E, Wujak LA, Niess G, Usemann J, activation improves angiogenesis in pulmonary artery endothelial cells
Reichenberger F, Herold S, Vadasz I, Mayer K, Seeger W, Morty with in utero pulmonary hypertension. Am J Physiol Lung Cell Mol
RE. Glucocorticoids recruit Tgfbr3 and Smad1 to shift transforming Physiol 304: L29 –L42, 2013.
growth factor-beta signaling from the Tgfbr1/Smad2/3 axis to the Acvrl1/ 331. Thakur VS, Liang YW, Lingappan K, Jiang W, Wang L, Barrios R,
Smad1 axis in lung fibroblasts. J Biol Chem 289: 3262–3275, 2014. Zhou G, Guntupalli B, Shivanna B, Maturu P, Welty SE, Moorthy B,
313. Sen P, Dharmadhikari AV, Majewski T, Mohammad MA, Kalin TV, Couroucli XI. Increased susceptibility to hyperoxic lung injury and
Zabielska J, Ren X, Bray M, Brown HM, Welty S, Thevananther S, alveolar simplification in newborn rats by prenatal administration of
Langston C, Szafranski P, Justice MJ, Kalinichenko VV, Gambin A, benzo[a]pyrene. Toxicol Lett 230: 322–332, 2014.
Belmont J, Stankiewicz P. Comparative analyses of lung transcriptomes 332. Thane K, Ingenito EP, Hoffman AM. Lung regeneration and transla-
in patients with alveolar capillary dysplasia with misalignment of pul- tional implications of the postpneumonectomy model. Transl Res 163:
monary veins and in Foxf1 heterozygous knockout mice. PLoS One 9: 363–376, 2014.
e94390, 2014. 333. Thomas W, Speer CP. Chorioamnionitis is essential in the evolution of
314. Setlakwe EL, Lemos KR, Lavoie-Lamoureux A, Duguay JD, Lavoie bronchopulmonary dysplasia–the case in favour. Paediatr Respir Rev 15:
JP. Airway collagen and elastic fiber content correlates with lung 49 –52, 2014.
function in equine heaves. Am J Physiol Lung Cell Mol Physiol 307: 334. Thunqvist P, Gustafsson P, Norman M, Wickman M, Hallberg J.
L252–L260, 2014. Lung function at 6 and 18 months after preterm birth in relation to
315. Shenberger JS, Shew RL, Johnson DE. Hyperoxia-induced airway severity of bronchopulmonary dysplasia. Pediatr Pulmonol 50: 978 –986,
remodeling and pulmonary neuroendocrine cell hyperplasia in the wean-
2014.
ling rat. Pediatr Res 42: 539 –544, 1997.
335. Tibboel J, Groenman FA, Selvaratnam J, Wang J, Tseu I, Huang Z,
316. Singh SP, Gundavarapu S, Smith KR, Chand HS, Saeed AI, Mishra
Caniggia I, Luo D, van Tuyl M, Ackerley C, de Jongste JC, Tibboel
NC, Hutt J, Barrett EG, Husain M, Harrod KS, Langley RJ, Sopori
D, Post M. Hypoxia-inducible factor-1 stimulates postnatal lung devel-
ML. Gestational exposure of mice to secondhand cigarette smoke causes
bronchopulmonary dysplasia blocked by the nicotinic receptor antagonist opment but does not prevent O2-induced alveolar injury. Am J Respir
mecamylamine. Environ Health Perspect 121: 957–964, 2013. Cell Mol Biol 52: 448 –458, 2015.
317. Sluiter I, Veenma D, van Loenhout R, Rottier R, de Klein A, Keijzer 336. Tibboel J, Joza S, Reiss I, de Jongste JC, Post M. Amelioration of
R, Post M, Tibboel D. Etiological and pathogenic factors in congenital hyperoxia-induced lung injury using a sphingolipid-based intervention.
diaphragmatic hernia. Eur J Pediatr Surg 22: 345–354, 2012. Eur Respir J 42: 776 –784, 2013.
318. Srisuma S, Bhattacharya S, Simon DM, Solleti SK, Tyagi S, Starcher 337. Tipple TE. The thioredoxin system in neonatal lung disease. Antioxid
B, Mariani TJ. Fibroblast growth factor receptors control epithelial- Redox Signal 21: 1916 –1925, 2014.
mesenchymal interactions necessary for alveolar elastogenesis. Am J 338. Tiwari KK, Chu C, Couroucli X, Moorthy B, Lingappan K. Differ-
Respir Crit Care Med 181: 838 –850, 2010. ential concentration-specific effects of caffeine on cell viability, oxida-
319. Stenmark KR, Abman SH. Lung vascular development: implications tive stress, and cell cycle in pulmonary oxygen toxicity in vitro. Biochem
for the pathogenesis of bronchopulmonary dysplasia. Annu Rev Physiol Biophys Res Commun 450: 1345–1350, 2014.
67: 623–661, 2005. 339. Tremblay Y, Provost PR. Major enzymes controlling the androgenic
320. Stouch AN, Zaynagetdinov R, Barham WJ, Stinnett AM, Slaughter pressure in the developing lung. J Steroid Biochem Mol Biol 137: 93–98,
JC, Yull FE, Hoffman HM, Blackwell TS, Prince LS. IkappaB kinase 2013.
activity drives fetal lung macrophage maturation along a non-M1/M2 340. Truog WE. Inhaled nitric oxide for the prevention of bronchopulmonary
paradigm. J Immunol 193: 1184 –1193, 2014. dysplasia. Expert Opin Pharmacother 8: 1505–1513, 2007.

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00268.2015 • www.ajplung.org


Perspectives
ADVANCES IN LUNG ALVEOLARIZATION AND BPD L1271
341. Tschanz SA, Salm LA, Roth-Kleiner M, Barre SF, Burri PH, 359. Warburton D, Shi W, Xu B. TGF-␤-Smad3 signaling in emphysema
Schittny JC. Rat lungs show a biphasic formation of new alveoli during and pulmonary fibrosis: an epigenetic aberration of normal development?
postnatal development. J Appl Physiol 117: 89 –95, 2014. Am J Physiol Lung Cell Mol Physiol 304: L83–L85, 2013.
342. Vadivel A, Alphonse RS, Etches N, van Haaften T, Collins JJ, 360. Warburton D, Tefft D, Mailleux A, Bellusci S, Thiery JP, Zhao J,
O’Reilly M, Eaton F, Thébaud B. Hypoxia-inducible factors promote Buckley S, Shi W, Driscoll B. Do lung remodeling, repair, and regen-
alveolar development and regeneration. Am J Respir Cell Mol Biol 50: eration recapitulate respiratory ontogeny? Am J Respir Crit Care Med
96 –105, 2014. 164: S59 –S62, 2001.
343. Vadivel A, Alphonse RS, Ionescu L, Machado DS, O’Reilly M, Eaton 361. Weibel ER. A retrospective of lung morphometry: from 1963 to present.
F, Haromy A, Michelakis ED, Thébaud B. Exogenous hydrogen Am J Physiol Lung Cell Mol Physiol 305: L405–L408, 2013.
sulfide (H2S) protects alveolar growth in experimental O2-induced neo- 362. Weichelt U, Cay R, Schmitz T, Strauss E, Sifringer M, Buhrer C,
natal lung injury. PLoS One 9: e90965, 2014. Endesfelder S. Prevention of hyperoxia-mediated pulmonary inflamma-
344. Veenma DC, de Klein A, Tibboel D. Developmental and genetic tion in neonatal rats by caffeine. Eur Respir J 41: 966 –973, 2013.
aspects of congenital diaphragmatic hernia. Pediatr Pulmonol 47: 534 – 363. West AR, Zaman N, Cole DJ, Walker MJ, Legant WR, Boudou T,
545, 2012. Chen CS, Favreau JT, Gaudette GR, Cowley EA, Maksym GN.
345. Velten M, Britt RD Jr, Heyob KM, Tipple TE, Rogers LK. Maternal Development and characterization of a 3D multicell microtissue culture
dietary docosahexaenoic acid supplementation attenuates fetal growth model of airway smooth muscle. Am J Physiol Lung Cell Mol Physiol
304: L4 –L16, 2013.
restriction and enhances pulmonary function in a newborn mouse model
364. West JB. Marcello Malpighi and the discovery of the pulmonary capil-
of perinatal inflammation. J Nutr 144: 258 –266, 2014.
laries and alveoli. Am J Physiol Lung Cell Mol Physiol 304: L383–L390,
346. Vogel ER, Britt RD Jr, Trinidad MC, Faksh A, Martin RJ, Mac-
2013.
Farlane PM, Pabelick CM, Prakash YS. Perinatal oxygen in the
365. Whitehead GS, Burch LH, Berman KG, Piantadosi CA, Schwartz
developing lung. Can J Physiol Pharmacol 93: 119 –127, 2015.
DA. Genetic basis of murine responses to hyperoxia-induced lung injury.
347. Vogel ER, VanOosten SK, Holman MA, Hohbein DD, Thompson Immunogenetics 58: 793–804, 2006.
MA, Vassallo R, Pandya HC, Prakash YS, Pabelick CM. Cigarette 366. Whitsett JA, Weaver TE. Alveolar development and disease. Am J
smoke enhances proliferation and extracellular matrix deposition by Respir Cell Mol Biol 53: 1–7, 2015.
human fetal airway smooth muscle. Am J Physiol Lung Cell Mol Physiol 367. Williams AE, Chambers RC. The mercurial nature of neutrophils: still
307: L978 –L986, 2014. an enigma in ARDS? Am J Physiol Lung Cell Mol Physiol 306: L217–
348. Volckaert T, De Langhe SP. Wnt and FGF mediated epithelial-mesen- L230, 2014.
chymal crosstalk during lung development. Dev Dyn 244: 342–366, 368. Witsch TJ, Niess G, Sakkas E, Likhoshvay T, Becker S, Herold S,
2015. Mayer K, Vadász I, Roberts JD Jr, Seeger W, Morty RE. Transglu-
349. Vosdoganes P, Lim R, Koulaeva E, Chan ST, Acharya R, Moss TJ, taminase 2: a new player in bronchopulmonary dysplasia? Eur Respir J
Wallace EM. Human amnion epithelial cells modulate hyperoxia-in- 44: 109 –121, 2014.
duced neonatal lung injury in mice. Cytotherapy 15: 1021–1029, 2013. 369. Witsch TJ, Turowski P, Sakkas E, Niess G, Becker S, Herold S,
350. Vyas-Read S, Wang W, Kato S, Colvocoresses-Dodds J, Fifadara Mayer K, Vadász I, Roberts JD Jr, Seeger W, Morty RE. Deregula-
NH, Gauthier TW, Helms MN, Carlton DP, Brown LA. Hyperoxia tion of the lysyl hydroxylase matrix cross-linking system in experimental
induces alveolar epithelial-to-mesenchymal cell transition. Am J Physiol and clinical bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol
Lung Cell Mol Physiol 306: L326 –L340, 2014. Physiol 306: L246 –L259, 2014.
351. Wagenaar GT, Laghmani el H, de Visser YP, Sengers RM, Steendijk 370. Wolf D, Tseng N, Seedorf G, Roe G, Abman SH, Gien J. Endothelin-1
P, Baelde HJ, Walther FJ. Ambrisentan reduces pulmonary arterial decreases endothelial PPAR␥ signaling and impairs angiogenesis after
hypertension but does not stimulate alveolar and vascular development in chronic intrauterine pulmonary hypertension. Am J Physiol Lung Cell
neonatal rats with hyperoxic lung injury. Am J Physiol Lung Cell Mol Mol Physiol 306: L361–L371, 2014.
Physiol 304: L264 –L275, 2013. 371. Xia H, Ren X, Bolte CS, Ustiyan V, Zhang Y, Shah TA, Kalin TV,
352. Wagenaar GT, Laghmani el H, Fidder M, Sengers RM, de Visser Whitsett JA, Kalinichenko VV. Foxm1 regulates resolution of hyper-
YP, de Vries L, Rink R, Roks AJ, Folkerts G, Walther FJ. Agonists oxic lung injury in newborns. Am J Respir Cell Mol Biol 52: 611–621,
of MAS oncogene and angiotensin II type 2 receptors attenuate cardio- 2015.
pulmonary disease in rats with neonatal hyperoxia-induced lung injury. 372. Xie W, Lynch TJ, Liu X, Tyler SR, Yu S, Zhou X, Luo M, Kusner
Am J Physiol Lung Cell Mol Physiol 305: L341–L351, 2013. DM, Sun X, Yi Y, Zhang Y, Goodheart MJ, Parekh KR, Wells JM,
353. Wagenaar GT, Sengers RM, Laghmani el H, Chen X, Lindeboom Xue HH, Pevny LH, Engelhardt JF. Sox2 modulates Lef-1 expression
MP, Roks AJ, Folkerts G, Walther FJ. Angiotensin II type 2 receptor during airway submucosal gland development. Am J Physiol Lung Cell
ligand PD123319 attenuates hyperoxia-induced lung and heart injury at Mol Physiol 306: L645–L660, 2014.
a low dose in newborn rats. Am J Physiol Lung Cell Mol Physiol 307: 373. Xu W, Liu C, Kaartinen V, Chen H, Lu CH, Zhang W, Luo Y, Shi
L261–L272, 2014. W. TACE in perinatal mouse lung epithelial cells promotes lung saccular
354. Walkup LL, Tkach JA, Higano NS, Thomen RP, Fain SB, Merhar formation. Am J Physiol Lung Cell Mol Physiol 305: L953–L963, 2013.
374. Yang G, Biswasa C, Lin QS, La P, Namba F, Zhuang T, Muthu M,
SL, Fleck RJ, Amin RS, Woods JC. Quantitative magnetic resonance
Dennery PA. Heme oxygenase-1 regulates postnatal lung repair after
imaging of bronchopulmonary dysplasia in the NICU environment. Am J
hyperoxia: role of beta-catenin/hnRNPK signaling. Redox Biol 1: 234 –
Respir Crit Care Med. In press.
243, 2013.
355. Wang H, Jafri A, Martin RJ, Nnanabu J, Farver C, Prakash YS,
375. Yang Y, Qiu J, Kan Q, Zhou XG, Zhou XY. MicroRNA expression
MacFarlane PM. Severity of neonatal hyperoxia determines structural profiling studies on bronchopulmonary dysplasia: a systematic review
and functional changes in developing mouse airway. Am J Physiol Lung and meta-analysis. Genet Mol Res 12: 5195–5206, 2013.
Cell Mol Physiol 307: L295–L301, 2014. 376. Yao H, Sundar IK, Ahmad T, Lerner C, Gerloff J, Friedman AE,
356. Wang H, St Julien KR, Stevenson DK, Hoffmann TJ, Witte JS, Phipps RP, Sime PJ, McBurney MW, Guarente L, Rahman I. SIRT1
Lazzeroni LC, Krasnow MA, Quaintance CC, Oehlert JW, Jelliffe- protects against cigarette smoke-induced lung oxidative stress via a
Pawlowski LL, Gould JB, Shaw GM, O’Brodovich HM. A genome- FOXO3-dependent mechanism. Am J Physiol Lung Cell Mol Physiol
wide association study (GWAS) for bronchopulmonary dysplasia. Pedi- 306: L816 –L828, 2014.
atrics 132: 290 –297, 2013. 377. Yoder BA, Coalson JJ. Animal models of bronchopulmonary dysplasia.
357. Warburton D, Bellusci S, De Langhe S, Del Moral PM, Fleury V, The preterm baboon models. Am J Physiol Lung Cell Mol Physiol 307:
Mailleux A, Tefft D, Unbekandt M, Wang K, Shi W. Molecular L970 –L977, 2014.
mechanisms of early lung specification and branching morphogenesis. 378. Yoo HS, Chang YS, Kim JK, Ahn SY, Kim ES, Sung DK, Jeon GW,
Pediatr Res 57: 26R–37R, 2005. Hwang JH, Shim JW, Park WS. Antenatal betamethasone attenuates
358. Warburton D, El-Hashash A, Carraro G, Tiozzo C, Sala F, Rogers intrauterine infection-aggravated hyperoxia-induced lung injury in neo-
O, De Langhe S, Kemp PJ, Riccardi D, Torday J, Bellusci S, Shi W, natal rats. Pediatr Res 73: 726 –733, 2013.
Lubkin SR, Jesudason E. Lung organogenesis. Curr Top Dev Biol 90: 379. Young SM, Liu S, Joshi R, Batie MR, Kofron M, Guo J, Woods JC,
73–158, 2010. Varisco BM. Localization and stretch-dependence of lung elastase ac-

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00268.2015 • www.ajplung.org


Perspectives
L1272 ADVANCES IN LUNG ALVEOLARIZATION AND BPD

tivity in development and compensatory growth. J Appl Physiol 118: 385. Zemans RL, McClendon J, Aschner Y, Briones N, Young SK, Lau
921–931, 2015. LF, Kahn M, Downey GP. Role of ␤-catenin-regulated CCN matricel-
380. Ysasi AB, Belle JM, Gibney BC, Fedulov AV, Wagner W, AkiraT- lular proteins in epithelial repair after inflammatory lung injury. Am J
suda, Konerding MA, Mentzer SJ. Effect of unilateral diaphragmatic Physiol Lung Cell Mol Physiol 304: L415–L427, 2013.
paralysis on postpneumonectomy lung growth. Am J Physiol Lung Cell 386. Zhang H, Sweezey NB, Kaplan F. LGL1 modulates proliferation,
Mol Physiol 305: L439 –L445, 2013. apoptosis, and migration of human fetal lung fibroblasts. Am J Physiol
381. Yu XM, Wang L, Li JF, Liu J, Li J, Wang W, Wang J, Wang C. Lung Cell Mol Physiol 308: L391–L402, 2015.
Wnt5a inhibits hypoxia-induced pulmonary arterial smooth muscle cell 387. Zhang S, Patel A, Chu C, Jiang W, Wang L, Welty SE, Moorthy B,
proliferation by downregulation of ␤-catenin. Am J Physiol Lung Cell Shivanna B. Aryl hydrocarbon receptor is necessary to protect fetal
Mol Physiol 304: L103–L111, 2013. human pulmonary microvascular endothelial cells against hyperoxic
382. Yurt M, Liu J, Sakurai R, Gong M, Husain SM, Siddiqui MA,
injury: Mechanistic roles of antioxidant enzymes and RelB. Toxicol Appl
Husain M, Villarreal P, Akcay F, Torday JS, Rehan VK. Vitamin D
Pharmacol 286: 92–101, 2015.
supplementation blocks pulmonary structural and functional changes in a
388. Zhang X, Xu J, Wang J, Gortner L, Zhang S, Wei X, Song J, Zhang
rat model of perinatal vitamin D deficiency. Am J Physiol Lung Cell Mol
Physiol 307: L859 –L867, 2014. Y, Li Q, Feng Z. Reduction of microRNA-206 contributes to the
383. Zana-Taieb E, Butruille L, Franco-Montoya ML, Lopez E, Vernier development of bronchopulmonary dysplasia through up-regulation of
F, Grandvuillemin I, Evain-Brion D, Deruelle P, Baud O, Delacourt fibronectin 1. PLoS One 8: e74750, 2013.
C, Jarreau PH. Effect of two models of intrauterine growth restriction 389. Zhao L, Yee M, O’Reilly MA. Transdifferentiation of alveolar epithelial
on alveolarization in rat lungs: morphometric and gene expression type II to type I cells is controlled by opposing TGF-␤ and BMP
analysis. PLoS One 8: e78326, 2013. signaling. Am J Physiol Lung Cell Mol Physiol 305: L409 –L418, 2013.
384. Zana-Taieb E, Pham H, Franco-Montoya ML, Jacques S, Le- 390. Zou W, Zou Y, Zhao Z, Li B, Ran P. Nicotine-induced epithelial-
tourneur F, Baud O, Jarreau PH, Vaiman D. Impaired alveolarization mesenchymal transition via Wnt/␤-catenin signaling in human airway
and intra-uterine growth restriction in rats: a postnatal genome-wide epithelial cells. Am J Physiol Lung Cell Mol Physiol 304: L199 –L209,
analysis. J Pathol 235: 420 –430, 2015. 2013.

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00268.2015 • www.ajplung.org

You might also like