Open access peer-reviewed chapter

Perinatal Lung Development: The Lung at Birth

Written By

Jyh-Chang Jean, Lou Ann Scism Brown and Martin Joyce-Brady

Submitted: 08 February 2016 Reviewed: 20 April 2016 Published: 31 August 2016

DOI: 10.5772/63819

From the Edited Volume

Respiratory Management of Newborns

Edited by Hany Aly and Hesham Abdel-Hady

Chapter metrics overview

1,854 Chapter Downloads

View Full Metrics

Abstract

The successful transition from liquid to air breathing at birth is essential in mammalian lung development and the primary biological role of the hypothalamic-pituitary-adrenal axis. At this moment, the lung experiences a major environmental change in oxygen tension as the fluid that filled this organ in utero (pO2 ∼26) is rapidly replaced with ambient air during the first few breaths (pO2 ∼150). This change induces oxidant stress in the lung which is balanced by antioxidant genes that are induced in the late gestational fetal lung to protect against injury. These antioxidant genes impact distinct antioxidant molecules, including glutathione, which can regulate the level of redox stress in lung cells. Cells, such as the alveolar macrophage, are central for host defense and surfactant homeostasis at the newly established air-liquid surface. Yet they are prone to dysfunction with excessive oxidant stress and the newborn lung is susceptible to infection. And yet the rise in alveolar oxygen tension can also serve as a physiologic redox signal that initiates expression of genes that regulate postnatal lung development. Taken together, birth can be viewed as a natural experiment with hyperoxia where the birth process itself serves as an integrator for that level of redox stress that limits lung injury while activating genes required for postnatal lung development.

Keywords

  • lung
  • perinatal
  • birth shock
  • glutathione
  • Kruppel-like factor 4

1. Introduction

The symbols for man (♂), woman (♀), birth (☼), death (†), and infinity (∞) were penned on a blackboard at the opening of the 1960s medical drama “Ben Casey.” These symbols represented a fundamental organization of life in the universe of medicine. Birth was a culmination of a progressive series of molecular and cellular events that began with fertilization, progressed through gastrulation, and then embryogenesis to form the fetus and finally the newborn child. Along the way, an ever enlarging population of cells proliferated, migrated, differentiated, and died and a host of organs with unique functions were constructed on organ-specific extracellular matrices connected to each other by vessels and to the external environment by tubes. Herein, we focus on a select aspect of lung development in the perinatal period. At that time, the fetus experienced a unique transition from a highly protected fluid-filled uterine environment to a highly susceptible gas-filled environment that marked its independent existence as a newborn. The role of the lung as a secretory organ came to an end and its new role as a gas exchanger began. In so doing, the cells of the lung, more so than any other organ, were immediately exposed to an elevated concentration of oxygen.

Decades of lung developmental research have unraveled many features about lung function in the perinatal period. Some highlights include a primary role for hormones in regulating lung cell maturation along with the ontogeny of a surfactant lipoglycoprotein complex to ensure the success of this transition, particularly the hypothalamic-pituitary-adrenal axis [1]. Although some aspects of perinatal lung alveolar, cell surface differentiation is steroid-independent [2]. Identification of regulators that control the progression of lung progenitor cells into distinct lineages to form distal lung alveolar cells [3]. The molecular events occurring during lung development are regulated by transcription factors, and the families involved are extensive [3, 4]. In this review, we will focus on a particular transcription factor family known as the Kruppel-like factors (KLF). Krüppel is a zinc-finger transcription factor described in Drosophila melanogaster as essential for pattern formation and embryogenesis [5]. A related family of mammalian Krüppel-like factors, Klfs, share sequence homology with the DNA-binding domain of Krüppel and serve essential roles in mammalian embryogenesis [6]. Lung Kruppel-like factor, Klf2, has been known to be important in lung development [7]. Another member, Klf5, was shown more recently to play a role in perinatal lung development [8]. Interestingly, Klf5 is known to interact with Klf4, gut enriched Kruppel-like factor, which we will highlight in this review [9].

Advertisement

2. The Kruppel-like family of transcription factors and the lung

Lung development is characterized by a dramatic structural and functional re-organization from an organ of secretion in utero to one of the gas exchanges at birth. This morphologic transition is only partially complete in the newborn murine and human lung. The process of forming new alveoli in the lung occurs largely in the postnatal period. It involves regulation of cell proliferation, differentiation, migration, and apoptosis and affects epithelial, endothelial, and interstitial cell populations within the lung parenchyma and airways. It also involves complex interactions between cells and extracellular matrix proteins [10]. Previous literature described many of the cellular components of these critical postnatal events, and contemporary research has concentrated on the molecular regulators that drive the formation of the gas exchange surface and the mechanical properties of the postnatal lung.

It was known that there was a progressive decline in cell proliferation in mesenchymal cells, including fibroblasts [11] and epithelial cells [12, 13], near the end of gestation, at birth, and during the early postnatal period. The effect was transient as the number of proliferating cells rose dramatically in the late postnatal period with the onset of alveolarization and continued over the next 2–3 weeks [14]. During this time, the interstitium, which was thick and packed with cells at birth, became progressively more thin and sparse with maturation of the alveolar wall [15]. The relative number of interstitial cells actually declined.

Fibroblast differentiation after birth was believed to serve major function in forming alveoli in the late postnatal lung. A population of myofibroblasts, located at the tips of developing septa, expressed α-smooth muscle actin (α-SMA) and produced matrix proteins such as elastin. The location of these cells denoted sites of newly forming alveoli and alveolarization extended from postnatal day 5 through day 20 in the mouse lung. This ability to form new alveoli appeared to be dependent upon the response of the myofibroblast to certain stimuli such as FGF-3, FGF-4 [16], and PDGF-A [17] receptors, as well as deposited matrix protein [18, 19] and transforming growth factor (TGF)-β activity, including a component of the TGF-β signaling pathway, Smad3 [20, 21]. Myofibroblasts are believed to re-emerge de novo in adult lung following tissue injury [22] and are postulated to play a role in repair. Differentiated fibroblasts also produce matrix proteins, including elastin and collagen. The lung actually has very little connective tissue at birth. As a result, lung elastic recoil is limited and the lung is prone to rupture by applied pressure. Production of matrix proteins such as elastin and collagen by interstitial fibroblasts during alveolarization contributes to elasticity and structural integrity to the lung [23]. Interestingly, regulation of cell proliferation and cell differentiation is a role that is reiterated in much of the Kruppel family literature.

Overall, the Kruppel-like family currently consists of over 16 members, and the originally descriptive nomenclature is now being revised into a numerical grouping system based on sequence homology, functional domains and transcriptional activity [24]. For instance, Klf4 is also known as EZF (epithelial zinc finger) and gut-enriched Klf (GKLF). It activates and represses gene transcription and has autoregulatory activity [25]. Klf2 is also known as lung Klf. It is mainly an activator of transcription and plays a role in blood vessel and lung development. Table 1 summarizes selected aspects of a select Klfs and more detailed summaries are available in recent reviews [24, 2628].

The Klfs are a subset of a larger family that includes Sp1 and Sp1-like proteins with Sp1 being the founding member of the family. Sp1 and Sp-1 like proteins are grouped together by phylogenetic tree analysis of protein sequences (group I) and the Klfs divide into groups II and III [24]. In general, Sp1 and Sp1-like members preferentially recognize the GT box in promoters (5′-GGTGTGGGG-3′), whereas the Klf members preferentially recognize the CA box (5′-CACCC-3′). However, competition among members for these sites and post-translational modifications that affect DNA-binding site affinity is an active area of investigation. It is believed that this diversity of protein members allows for a precise regulation of gene expression under a variety of physiological conditions. The ability of the Klf family to regulate cell proliferation, survival, and differentiation of different cell lineages suggests a potentially broad role in normal development, response to injury, and carcinogenesis.

Klf #  Alternate name Transcriptional activity Cell functions Phylogenetic grouping Expression sites Expressed in lung
Klf2 Lung KLF Activator Blood vessel, lung development, T cell survival II Blood vessels, hematopoietic cells + [29, 30]
Klf4 Gut KLF Activator/ repressor Anti-proliferation
Survival
II Gut, epithelium, fibroblasts + [31, 32]
Klf5 BTEB2 Activator Cell growth II + [7, 33]
Klf6 COPEB Activator Putative tumor suppressor II + [34]
Klf9 BTEB1 Activator/ repressor Neurite outgrowth
Carcinogen  metabolism
III Ubiquitous + [35]
Klf13 BTEB3 Activator/ repressor Anti-proliferation
Carcinogen  metabolism
III + [36]

Table 1.

Select characteristics for some members of the Krüppel-like family.

Klf4 was initially characterized in two independent laboratories. One identified it as a novel zinc-finger protein expressed in differentiated epithelial cells, hence named epithelial zinc finger (EZF) [31], and transiently in certain mesenchymal cells. The other identified it as a gut-enriched Krüppel-like factor expressed during growth arrest, hence GKLF [32]. In cultured fibroblasts, expression was abundant in growth-arrested cells and barely detectable in rapidly proliferating cells as the protein inhibited DNA synthesis. The message transcript was most abundant in colon followed by testis, lung, and small intestine by Northern blot and in epithelial cells of the colon as they migrated from the base to the top of the crypt [32]. Message was not detectable in the mouse embryo by Northern blot until day 15.5, but was evident in mesenchymal cells of the first branchial arch at day 11.5 and the metanephric kidney at day 12.5 by in situ hybridization. At day 15.5, the situ signal was detected in epithelial cells of the colon and the tongue and by the newborn period in epithelial cells of the esophagus and the stomach as well [31]. Message was also evident in skin of adult mice [31]. More recently, Klf4 expression has been defined in corneal epithelial cells [37]. This new gene was hypothesized to play a role in epithelial cells in many organs during differentiation in development and in mesenchymal cells during early formation of the skeleton and kidney.

These original studies demonstrated that Kl4 was an important regulator of cell proliferation and cell differentiation. In keeping with this concept, serum deprivation, DNA damage, and contact inhibition were associated with decreased proliferation and increased Klf4 expression [32, 38], while GI neoplastic lesions [39, 40] and cancer cell lines [41] were associated with high levels of proliferation but decreased levels of Klf4 expression. In tissues, Klf4 was expressed largely in post-mitotic, terminally differentiated epithelial cells [31, 32]. Transcriptional profiling of Klf4 confirmed this dual role in cell cycle regulation and epithelial cell differentiation [42]. Cell cycle inhibitors were upregulated by Klf4, including the cdk inhibitor p21WAF1/CIP1, and cell cycle activators were downregulated, including cyclinD1 [38, 43]. Targeted elimination of the Klf4 gene in mice revealed its critical role in terminal differentiation of epithelial cells in the skin in the perinatal period [44]. Loss of barrier function was lethal within 24 h of birth due to the rapid loss of fluids. An independent Klf4 gene ablation experiment not only confirmed this observation but also showed an alteration in the terminal differentiation program of epithelial goblet cells in the colon [45].

In keeping with the known expression of Klf4 in mesenchymal cells, several studies demonstrated regulation of this gene in non-epithelial cell types. Human Klf4 was originally cloned from a human umbilical vein cDNA library after the initial description of mouse Klf4 [46]. More recently, Klf4 induction was observed in a genomic analysis of immediate/early responses to shear stress in human coronary artery endothelial cells [47]. Shear stress inhibits endothelial cell proliferation, and Klf4 was postulated to provide a mechanism for this effect. Studies with cardiac fibroblasts identified Klf4 induction by DNA microarray and linked its expression with several cell cycle and differentiation-associated genes that were induced with a sudden change from hypoxic to normoxic conditions in vitro to model “perceived hyperoxia” of ischemia-reperfusion. Klf4 was proposed to play a role in activating pathways for growth-arrest and fibroblast differentiation in these cells in response to oxygen-induced injury [48]. Klf4 has also been shown to mediate redox-sensitive inhibition of proliferation in vascular smooth muscle cells [49]. Klf4 induction in response oxidant exposure was dependent on hydroxyl radical production, intracellular calcium, p38 MAP kinase activation, and protein synthesis, although a transcriptional mechanism for its induction was not defined. The Klf4 response was rapid, detectable within 30 min, and associated with induction of p21WAF/Cip1, p27, and p53, known Klf4 target genes and inhibitors of cell proliferation. While Klf4 expression was known previously to respond to changes in the cell environment, this study identified oxidants as an additional extracellular signaling stimulus for Kl4 induction, and linked induction was activation of the MAPK signal transduction pathway.

Prior to our investigation, little was known about a role for Klf4 in lung development. Nonetheless, lung Klf4 expression had been reported in the lung in several different studies. The original characterizations of Klf4 noted expression in the adult [32] and the newborn lung [31]. Klf4 induction was also described in newborn compared to 4-week-old mice, a time of active lung cell proliferation and differentiation [50]. Early and transient Klf4 protein induction was described in the lung within 2 h following pneumonectomy although the cellular site was not determined [51]. Klf4 mRNA expression was induced in the airway transcriptome in response to cigarette smoke [52]. Klf4 mRNA was induced in alveolar epithelial type 1-like cells in response to ozone exposure [53]. Klf4 has very recently been shown to directly regulate TGF-β induced myofibroblast differentiation in isolated adult rat lung fibroblasts by inhibiting expression of α-SMA [54]. In this model, a Klf4 protein–protein interaction with Smad3 blocks Smad3 binding to a Smad3-binding element in the α-SMA promoter. This direct linkage of Klf4 with Smad3 reveals a novel mechanism by which Klf4 regulates TGF-β-induced myofibroblast gene expression in adult lung.

With this background in mind, our finding that lung Klf4 mRNA was dramatically upregulated with birth in a normal oxygen environment and attenuation of this induction by birth in a more hypoxic environment led us to hypothesize that Klf4 was regulated in a redox-responsive fashion in lung mesenchymal cells and that its target genes regulated fibroblast proliferation and differentiation events associated with postnatal lung development [55].

Advertisement

3. Oxygen as a regulator of Kruppel-like factor 4 in the newborn lung

This birth shock hypothesis was studied in time-pregnant mice by comparing findings in the lung with those with those in the liver, an organ that is not exposed to the same changes in environmental oxygen as the lung right at birth. And changes in newborn organs at 2, 6, 12, and 24 h after birth were compared with those of the corresponding fetal organ at day 21 of gestation, the time just before birth. Changes in gene expression at birth were examined with microarrays, and these data revealed an acute change in the level of expression of 157 genes within 2 h of birth in room air in the lung. The number of gene changes steadily declined at 6, 12, and 24 h thereafter. Most of the lung gene changes through 6 h involved transient induction or repression of expression. Less than 30% of the changes involved genes in common to any two successive time points, suggesting the presence of sequential waves of new gene expression over time. The full implication of this pattern is yet to be fully understood. Nonetheless, four functional categories of gene expression were overrepresented in these microarray data at 2 h after birth in room air: transcriptional regulators, structural genes, apoptosis-related genes, and antioxidants. Glutathione disulfide was analyzed to determine whether there was a change in the local redox state after birth in room air. This oxidation product accumulated fourfold within 2 h of birth and steadily decreased thereafter. Glutathione was always in excess of glutathione disulfide at all times points indicating a surfeit of antioxidant buffering capacity. But the lung is exposed to a transient oxidant stress and a change in the local redox state with the rise in environmental oxygen at birth.

Contrast these lung findings with those in the liver after birth in room air. The liver exhibited no accumulation of glutathione disulfide at the 2 h time point and no sign of oxidant stress at any of the early time points thereafter. These data suggest that the liver may not be subjected to the same magnitude of change in environmental oxygen tension as the lung at birth or it may follow a very different time course. In addition, there were far fewer changes in gene expression in the liver at 2 h after birth when compared to the lung, and no functional categories of gene expression were found to be overrepresented in the liver gene microarray dataset. There were a few gene expression levels that did change in common between these two organs. These could represent a shared response to the birth process itself. But we interpreted the majority of unique changes in the lung as a lung-specific response related to the change in alveolar oxygen content at birth.

To explore this, we examined a group of time-pregnant mice exposed to 10% oxygen using a glove bag at the 21st day of gestation. Some of these hypoxic mice were killed after 12 h to harvest fetal lung and assess baseline fetal measures at the end of gestation, whereas others were monitored and newborn lung was collected again at 2, 6, and 12 h after birth. Of note here, all dams, fetuses, and newborns survived in the 10% oxygen environment, and this exposure did not significantly alter baseline gene expression in the fetal lung as determined with microarrays. This survival pattern suggested that changes in blood flow, lung stretch, nutrient supply, and fluid resorption were similar to that of mice birthed in room air and that these factors were less likely to differentially impact gene expression. Birth into 10% oxygen, however, did impact the level of redox stress in the lung at birth as well as the number of gene expression changes. Accumulation of glutathione disulfide was attenuated by half compared to the normal lung. And there was a 60% decrease in the number of gene changes at 2 h, compared to that in room air with elimination of any overrepresented functional category of gene expression. It was as if birth into an environment of hypoxia lacked any change in gene expression at all and retained a pattern like that of the fetal lung. The transient rise and fall in glutathione disulfide in the newborn lung in room air, despite the induction of several antioxidants in the late fetal lung and the lung at birth which buffered some but not all such stress [5659], supports our hypothesis that a measured degree of redox stress, a birth shock, may serve a function in the lung at birth which we surmise is signalling expression of lung genes required for the onset of postnatal lung development [55].

The most provocative piece of data in our study was finding that the gene exhibiting the greatest change in expression in the lung at birth in room air was a transcription factor. And that transcription factor was Kruppel-like factor 4. Klf4 was previously referred to as gut Kruppel-like factor (GKLF), a tissue-specific gene that inhibited cell proliferation and stimulated differentiation in the gut, particularly in the epithelial cells of the crypts where there is a tight link between growth arrest and differentiation. Klf4 regulates cell proliferation by inhibiting cell-cycle progression and inducing growth arrest [31, 32, 60]. Mesenchymal and epithelial cell proliferation is known to progressively decline in the lung during the perinatal period [2931], and a transcription factor like Klf4 is a reasonable candidate to regulate this process. Klf4 is known to induce growth arrest in cells through the cell-cycle inhibitor p21WAF/Cip1, and the absence of Klf4 in the Klf4 knockout mouse is associated with a corresponding decrease in the level of p21WAF/Cip1 mRNA expression in the late fetal lung and persistent cell proliferation in fibroblasts and airway epithelial cells at birth [55].

Klf4 has previously been described in the mesenchyme during development [31]. We generated Klf4 knockout and Klf4 expressing fibroblasts to demonstrate that smooth muscle actin, fibronectin, tenascin C, and the alpha 1 chain of Type 1 collagen are Klf4 target genes and that fibroblast connective tissue gene expression is down-regulated in the perinatal lung of the Klf4 knockout mouse [55]. Impaired connective tissue gene expression has at least two distinct biological implications for postnatal lung development. First, lack of collagen synthesis decreases the tensile strength of the postnatal lung and increases the risk for rupture under pressure [61] and emphysema [62]. Second, lack of fibronectin, type 1 collagen, tenascin C, and smooth muscle actin expression can impair myofibroblast differentiation and postnatal alveogenesis [6366]. Interestingly, Klf4 our study confirmed differential regulation of smooth muscle actin gene expression in the myofibroblast and the vascular smooth muscle cell as Klf4 deficiency in the Klf4 knockout mouse lead to a loss of expression in the myofibroblast (positive regulator) but preservation of expression in the vascular smooth cell (negative regulator). This negative regulatory function in vascular smooth muscle cells was already known [67].

Finally, cell death was also increased in the Klf4-deficient lung of the Klf4 knockout mouse at birth. An expanding literature now describes Klf4 as an inhibitor of apoptosis [6]. Apoptosis is present even in the normal lung at birth and may play a physiologic role in postnatal lung development [68, 69]. We found an excess of apoptosis in the Klf4-deficient lung of the Klf4 knockout mouse, and this could well contribute to a loss of cell population and gene expression at birth. We also noticed that apoptosis exhibited a cell-specific rather than a general nature, as it was not increased in vascular smooth muscle cells within large blood vessels [55]. Taken together, we found good correlation between the gene categories that were overrepresented in the gene changes 2 h after birth and Klf4 as a potential regulator of these gene categories.

Is Klf4 expression in lung fibroblasts regulated by hyperoxia? We showed that it is and the mechanism is transcriptional in nature and independent of protein synthesis [55]. Further details of the mechanism await more studies. But Klf4 is known to be activated by oxidants, heat, chemical and mechanical forces, and nutrient deprivation [6]. Lack of Klf4 induction can be associated with stress [70] and the Klf4-deficient lung of the Klf4 knockout mouse at birth exhibited signs of stress such as increased degree of apoptosis and p53 expression and dramatic induction of p21CIP1//Waf1 independent of Klf4. The full implications of Klf4 expression in the perinatal period will require further study in vitro using Klf4 deficient and sufficient cell cultures and tissue explants, but ultimate proof will require a fibroblast-specific conditional deletion of Klf4 in vivo [71, 72]. But our study was the first to draw attention to a physiologic role for oxygen as a regulator of this transcription factor for fibroblast and myofibroblast differentiation in normal postnatal lung development [55]. The role of Klf4 in regulating these fibroblast populations during lung development is likely to be recapitulated in the adult lung during repair of lung injury and to involve interactions with other Kruppel-like family members. The newborn lung at birth could provide a window into the responsiveness of an organism to changes in the environment of oxygen and oxidant stress.

Advertisement

4. Glutathione in perinatal lung antioxidant defense and macrophage function

In the alveolar space, alveolar macrophage (AM) surveillance is the first line of defense in the immune response as they ingest and clear pathogens, release cytokines and chemokines to recruit other immune cells, serve as antigen presenting cells, and interact with other alveolar cells to fine-tune the immune response. In order to fulfill their roles as sensors, transmitters, and responders to inflammation, AMs respond to endogenous and exogenous danger signals by changing their repertoire of surface receptor expression and functional phenotype to promote either host defense, wound healing, or immune regulation. This plasticity in their biological responses, including differentiation, phenotype, immune functions, and cellular interactions, is determined in large part by their extracellular milieu [73]. However, the phenotype is not permanent and pathogens or changes in the underlying microenvironment can promote a change between a predominance of one set of characteristics to another phenotype. Depending on the different stimuli within the microenvironment within the alveolar space, alveolar macrophages are activated via distinct pathways that produce opposing effects on macrophage receptor expression, cytokine expression, and phagocytic function [74]. In response to interferon-γ (IFN-γ) or microbial products, macrophages become classically activated (M1) macrophages and produce inflammatory cytokines and reactive intermediates of oxygen and nitrogen. In response to interleukin-4 or -13, macrophages become alternatively activated (M2) and produce anti-inflammatory mediators such as the interleukin-1 receptor antagonist and TGF-β, promote tissue repair and remodeling, and become tolerant to stimulation by endotoxin in order to protect against overwhelming systemic inflammation. However, this immunosuppression is associated with a decreased capacity to clear microbes, and many diseases have abnormal shifts in the AM phenotype and immune responses that limit the ability of the cells to become innate immune effectors. Whether newborn AMs are skewed toward a M1 or M2 phenotype is unclear, but studies in a mouse model suggest that AM maturation in the fetal lung may include features of both pro-inflammatory and alternative activation paradigms [75].

Newborn infants, particularly those born preterm, face immunological challenges after birth because of the developmental stage of their immune systems. Upon birth, there is an age-dependent maturation of the intrapulmonary inflammatory responses with decreased AM immune responses such as phagocytosis and secretion of inflammatory mediators with potential immunoregulatory consequences [76, 77]. The functionally immature status of the newborn AM is suggested to be critical in the recurrent problem of early infancy pulmonary infections and morbidity [78]. At day 3 of life for premature newborns, the alveolar cells are more likely to have greater numbers of mononuclear phagocytes in the airway but fewer of these cells are functionally mature AMs [79]. This suggests that newborn AMs are functionally immature which may lead to increased susceptibility to lung infections. Furthermore, the degree of postnatal immune-suppression correlates with gestational age and is a predisposing factor for late infections [80].

The growth factor GM-CSF, granulocyte-macrophage colony stimulating factor, and its transcription factors PU.1 and Bach2 are increased after birth and necessary for AM differentiation and homeostasis making them critical to the AM immune response [81]. In newborn mice, the pleotropic peroxisome proliferator-activated receptor-γ (PPARγ) modulates several transcription factors and genes associated with AM differentiation and immune functions. However, PPARγ expression in fetal lung monocytes is dependent on the GM-CSF pathway suggesting that GM-CSF has a lung-specific role in the perinatal development of AMs through the induction of PPARγ in fetal monocytes. Therefore, factors that suppress GM-CSF or PPARγ expression in the newborn AM also suppress differentiation and the development of a mature immune response as summarized I recent reviews [8285].

In in vivo studies, ROS induces upregulation of TGF-β expression [86], which is also a characteristic of a M2 phenotype [74]. Once TGF-β is activated, it induces the GSH depletion and increases intracellular ROS production. Children with severe asthma have significantly higher concentration of TGF-β in the epithelial lining fluid, which is associated with increased TGF-β and increased oxidative stress in the AMs [87]. In a mouse model, intranasal instillation of an adenovirus expressing constitutively active TGF-β results in suppression of the expression of both catalytic and modifier subunits of glutamate-cysteine ligase (GCL), the rate-limiting enzyme in de novo glutathione synthesis, decreased glutathione concentration, and increased protein and lipid peroxidation in mouse lung [88]. Similarly, M2 macrophages have robust increases in TGF-β1 production and secretion [74], thereby creating a vicious cycle of ROS generation, TGF-β production and signaling, and a M2 phenotype [89]. Indeed, mounting evidence suggests that AMs, particularly M2 AMs, are the predominant source of TGF-β in lung fibrosis and depleting M2-polarized macrophages during the progressive phase of fibrosis reduces TGF-β and collagen deposition [90]. In contrast, glutathione precursors induce proteolysis of the TGF-β type II receptor and disintegration of TGF-β1 [86]. Therefore, perturbations in glutathione availability and its redox status in the epithelial lining fluid are important modulators of the AM phenotype and immune functions.

In addition a role for gestational development in the AM phenotype and immune response, the role of fetal exposures and subsequent changes in the microenvironment on the AM phenotype must also be considered. In animal models, in utero ethanol exposure also promotes oxidant stress in the newborn lung as evidenced by decreased concentrations of the reduced moiety and increased concentration of the oxidized glutathione moiety in the alveolus and neonatal AM [91]. This oxidation of the glutathione redox state in the alveolar space was associated with increased expression of TGF-β and markers of a M2 phenotype in the newborn AM [9294]. Impaired AM phagocytosis decreased the neonatal lung’s defense against experimental Group B Strep as well as systemic sepsis [92]. However, addition of the glutathione precursor S-adenosyl-methionine (SAM-e) to the dam’s diet restored the AM immune responses as well as decreased the experimental Group B Strep pneumonia and systemic sepsis in the newborn with fetal ethanol exposure. A central role for glutathione availability in the alveolar space in the AM immune functions was further supported by the ability of intranasal delivery of glutathione in the newborn to reverse the immune dysfunction associated with fetal ethanol exposure. These studies highlight the vital importance of glutathione availability in the alveolar space for AM immune functions and the neonatal lung’s defense against bacterial infection [92].

Studies in the adult of chronic ethanol ingestion highlight the role of the glutathione redox state and oxidative stress in AM expression of TGF-β, GM-CSF/PU.1 signaling, PPARγ expression, immune functions, and risk of experimental pneumonia [89, 9598]. In addition to GM-CSF, expression and activity of PU.1 are also dependent on Nrf2 and the antioxidant response element which are downregulated by alcohol-induced oxidant stress [99]. Alcohol exposure also upregulates KLF4 in AMs and treatments with GM-CSF or TGF-β enhance or dampen KLF4 expression and binding, respectively [100]. Treatment with siRNA against KLF4 normalizes the effects on expression of GM-CSF and TGF-β as well as AM immune functions. A potential dynamic interactive role for chronic oxidant stress on TGF-β, GM-CSF, PU.1, Nrf2, and KLF4 on the newborn AM phenotype and immune functions remains to be determined.

The most common form of newborn chronic lung disease, bronchopulmonary dysplasia (BPD), is thought to be caused by oxidative disruption of lung morphogenesis. Following birth, various risk factors, including pulmonary or systemic infections, high concentrations of oxygen inspiration, and mechanical ventilation, may act synergistically, amplifying the inflammation. At birth, premature newborns have significantly lower glutathione concentrations than term newborns, and the lowest glutathione levels are associated with the infants with the most severe airways problems and required high oxygen support [101104]. In intubated newborns, exhaled breath condensate samples correlated with tracheal aspirate samples in terms of reduced, oxidized, and total glutathione (GSH + GSSG) [105]. Decreased glutathione and subsequent oxidative stress in this microenvironment of the lining fluid was also observed in the AMs obtained from the tracheal aspirate. This glutathione deficiency and oxidative stress associated with the immature lung are likely to promote changes in the AM phenotype and immune functions, thereby increasing the risk of infection. This increased oxidative stress may contribute to the significant increased expression of TGF-β in the AMs of premature newborns [106, 107]. Further research is needed to specifically decipher the unique and complex mechanisms underlying AM maturation and pulmonary immune regulation at baseline and in response to oxidative stress. Pulmonary immune regulation in the premature neonate may be similar to the adult or it may be dramatically different because of the immaturity of the immune system.

Advertisement

5. Is birth an integrator that balances oxidant stress and antioxidant defense?

We have presented our work and ideas about the lung at birth and its experience of a major change in the oxygen environment. Clearly antioxidant gene induction is part of the developmental program in the late fetal lung and provides protection against oxidant stress. Other antioxidant genes are induced at birth in response to oxidant stress as an added source of protection. These activities are essential to a cell like the alveolar macrophage which must now populate the gas-filled alveolar space, protect against inhaled pathogens, and regulate surfactant homeostasis. Nonetheless, the normal lung does not buffer all the oxidant stress it encounters at birth. Rather a transient but measurable degree of oxidant stress persists and we propose that this level serves a physiological function in our birth shock hypothesis. As the local redox state of lung cells changes, this signals the lung to initiate an alternative course for postnatal lung development. It is in effect a “natural experiment with hyperoxia” as the lung cannot predict the magnitude of the change in the oxygen environment that will ensue. It survives by balancing a level of redox stress that limits lung injury, while permitting activation of genes required for postnatal lung development. Birth then may serve as an integrator of these two competing ends. Our observations about the transcription factor Klf4 (a birth shock protein), fibroblast and myofibroblast and macrophage cell populations and the transcriptional regulators we described affecting cell proliferation, cell differentiation, and cell apoptosis in our studies are likely only the beginning of understanding this hypothesis. We believe that study of an event such as this in the newborn lung may provide new insight and strategies to assess, support, and manipulate the oxygen environment and oxidant stress to enable a more normal development for the premature lung at birth and repair of oxidant-induced injury in the developing and the mature lung.

Advertisement

Acknowledgments

Dr. Joyce-Brady was funded by NHLBI PO1 HL47049 and an Ignition Award from Boston University Office of Technology Development. Dr. Brown is funded by National Institutes on Alcohol Abuse and Alcoholism P50 AA-135757 and National Heart Lung And Blood Institute 1RO1 HL 125042 and 5R34 HL 117351.

References

  1. 1. Muglia L, Jacobson L, Dikkes P, Majzoub JA. Corticotropin-releasing hormone deficiency reveals major fetal but not adult glucocorticoid need. Nature 1995;373:427–32.
  2. 2. Joyce-Brady MF, Brody JS. Ontogeny of pulmonary alveolar epithelial markers of differentiation. Dev Biol 1990;137:331–48.
  3. 3. Treutlein B, Brownfield DG, Wu AR, Neff NF, Mantalas GL, Espinoza FH, et al. Reconstructing lineage hierarchies of the distal lung epithelium using single-cell RNA-seq. Nature 2014 May 15;509(7500):371–5.
  4. 4. Maeda Y, Dave V, Whitsett JA. Transcriptional control of lung morphogenesis. Physiol Rev 2007 Jan;87(1):219–44.
  5. 5. St JD, Nusslein-Volhard C. The origin of pattern and polarity in the Drosophila embryo. Cell 1992 Jan 24;68(2):201–19.
  6. 6. McConnell BB, Yang VW. Mammalian Kruppel-like factors in health and diseases. Physiol Rev 2010 Oct;90(4):1337–81.
  7. 7. Wani MA, Wert SE, Lingrel JB. Lung Kruppel-like factor, a zinc finger transcription factor, is essential for normal lung development. J Biol Chem 1999 Jul 23;274(30):21180–5.
  8. 8. Wan H, Luo F, Wert SE, Zhang L, Xu Y, Ikegami M, et al. Kruppel-like factor 5 is required for perinatal lung morphogenesis and function. Development 2008 Aug;135(15):2563–72.
  9. 9. Bieker JJ. Kruppel-like factors: three fingers in many pies. J Biol Chem 2001 Sep 14;276(37):34355–8.
  10. 10. Brody JS, Thurlbeck W. Development, growth, and aging of the lung. Handbook of Physiology. The Respiratory System. Bethesda: American Physiological Society; 1986. p. 355–86.
  11. 11. Kauffman SL, Burri PH, Weibel ER. The postnatal growth of the rat lung. II. Autoradiography. Anat Rec 1974 Sep;180(1):63–76.
  12. 12. Adamson IY, King GM. Epithelial-mesenchymal interactions in postnatal rat lung growth. Exp Lung Res 1985;8(4):261–74.
  13. 13. O'Hare KH, Townes PL. Morphogenesis of albino rat lung: an autoradiographic analysis of the embryological origin of the type I and II pulmonary epithelial cells. J Morphol 1970;132(1):69–87.
  14. 14. Burri PH. The postnatal growth of the rat lung. 3. Morphology. Anat Rec 1974 Sep;180(1):77–98.
  15. 15. Vaccaro C, Brody JS. Ultrastructure of developing alveoli. Anat Rec 1978;192:467–80.
  16. 16. Weinstein M, Xu X, Ohyama K, Deng CX. FGFR-3 and FGFR-4 function cooperatively to direct alveogenesis in the murine lung. Development 1998 Sep;125(18):3615–23.
  17. 17. Bostrom H, Willetts K, Pekny M, Leveen P, Lindahl P, Hedstrand H, et al. PDGF-A signaling is a critical event in lung alveolar myofibroblast development and alveogenesis. Cell 1996 Jun 14;85(6):863–73.
  18. 18. Nakamura T, Lozano PR, Ikeda Y, Iwanaga Y, Hinek A, Minamisawa S, et al. Fibulin-5/DANCE is essential for elastogenesis in vivo. Nature 2002 Jan 10;415(6868):171–5.
  19. 19. Yanagisawa H, Davis EC, Starcher BC, Ouchi T, Yanagisawa M, Richardson JA, et al. Fibulin-5 is an elastin-binding protein essential for elastic fibre development in vivo. Nature 2002 Jan 10;415(6868):168–71.
  20. 20. Chen H, Sun J, Buckley S, Chen C, Warburton D, Wang XF, et al. Abnormal mouse lung alveolarization caused by Smad3 deficiency is a developmental antecedent of centrilobular emphysema. Am J Physiol Lung Cell Mol Physiol 2005 Apr;288(4):L683–L691.
  21. 21. Morris DG, Huang X, Kaminski N, Wang Y, Shapiro SD, Dolganov G, et al. Loss of integrin alpha(v)beta6-mediated TGF-beta activation causes Mmp12-dependent emphysema. Nature 2003 Mar 13;422(6928):169–73.
  22. 22. Rishikof DC, Lucey EC, Kuang PP, Snider GL, Goldstein RH. Induction of the myofibroblast phenotype following elastolytic injury to mouse lung. Histochem Cell Biol 2006 May;125(5):527–34.
  23. 23. Nardell EA, Brody JS. Determinants of mechanical properties of rat lung during postnatal development. J Appl Physiol 1982 Jul;53(1):140–8.
  24. 24. Kaczynski J, Cook T, Urrutia R. Sp1-and Kruppel-like transcription factors. Genome Biology 2003;4(2):206.
  25. 25. Mahatan C, Kaestner K, Geiman D, Yang V. Characterization of the structure and regulation of the murine gene encoding gut-enriched Kruppel-like factor (Kruppel-like factor 4). Nuc Acids Res 1999;27(23):4562–9.
  26. 26. Lomberk G, Urrutia R. The family feud: turning off Sp1 by Sp1-like KLF proteins. Biochemical Journal 2005 Nov 15;392:1–11.
  27. 27. Philipsen S, Suske G. A tale of three fingers: the family of mammalian Sp/XKLF transcription factors. Nucleic Acids Res 1999 Aug 1;27(15):2991–3000.
  28. 28. Wei D, Kanai M, Huang S, Xie K. Emerging role of KLF4 in human gastrointestinal cancer. Carcinogenesis 2006 Jan;27(1):23–31.
  29. 29. Mariani TJ, Reed JJ, Shapiro SD. Expression profiling of the developing mouse lung: Insights into the establishment of the extracellular matrix. Am J Respir Cell Mol Biol 2002;26:541–8.
  30. 30. Wani MA, Wert SE, Lingrel JB. Lung Kruppel-like factor, a zinc finger transcription factor, is essential for normal lung development. J Biol Chem 1999 Jul 23;274(30):21180–5.
  31. 31. Garrett-Sinha L, Eberspaecher H, Seldin M, de Crombrugghe B. A gene for a novel zinc-finger protein expressed in differentiated epithelial cells and transiently in certain mesenchymal cells. J Biol Chem 1996;271:31384–90.
  32. 32. Shields J, Christy R, Yang V. Identification and characterization of a gene encoding a gut-enriched Kruppel-like factor expressed during growth arrest. J Biol Chem 1996;271(33):20009–17.
  33. 33. Ohnishi KTMKYSSKYT. Matrix metalloproteinase-mediated extracellular matrix protein degradation in human pulmonary emphysema. Lab Invest 1998;78(9):1077–87.
  34. 34. Laub F, Aldabe R, Ramirez F, Friedman S. Embryonic expression of Kruppel-like factor 6 in neural and non-neural tissues. Mechanisms of Development 2001 Aug;106(1–2):167–70.
  35. 35. Imataka H, Sogawa K, Yasumoto K, Kikuchi Y, Sasano K, Kobayashi A, et al. Two regulatory proteins that bind to the basic transcription element (BTE), a GC box sequence in the promoter region of the rat P-4501A1 gene. EMBO J 1992 Oct;11(10):3663–71.
  36. 36. Scohy S, Gabant P, Van RT, Hertveldt V, Dreze PL, Van VP, et al. Identification of KLF13 and KLF14 (SP6), novel members of the SP/XKLF transcription factor family. Genomics 2000 Nov 15;70(1):93–101.
  37. 37. Chiambaretta F, De GF, Turet G, Marceau G, Gain P, Dastugue B, et al. Cell and tissue specific expression of human Kruppel-like transcription factors in human ocular surface. Mol Vis 2004 Nov 23;10:901–9.
  38. 38. Zhang W, Geiman DE, Shields JM, Dang DT, Mahatan CS, Kaestner KH, et al. The gut-enriched Kruppel-like factor (Kruppel-like factor 4) mediates the transactivating effect of p53 on the p21WAF1/Cip1 promoter. J Biol Chem 2000 Jun 16;275(24):18391–8.
  39. 39. Dang DT, Bachman KE, Mahatan CS, Dang LH, Giardiello FM, Yang VW. Decreased expression of the gut-enriched Kruppel-like factor gene in intestinal adenomas of multiple intestinal neoplasia mice and in colonic adenomas of familial adenomatous polyposis patients. FEBS Lett 2000 Jul 7;476(3):203–7.
  40. 40. Ton-That H, Kaestner KH, Shields JM, Mahatanankoon CS, Yang VW. Expression of the gut-enriched Kruppel-like factor gene during development and intestinal tumorigenesis. FEBS Lett 1997 Dec 15;419(2–3):239–43.
  41. 41. Dang DT, Mahatan CS, Dang LH, Agboola IA, Yang VW. Expression of the gut-enriched Kruppel-like factor (Kruppel-like factor 4) gene in the human colon cancer cell line RKO is dependent on CDX2. Oncogene 2001 Aug 9;20(35):4884–90.
  42. 42. Chen X, Whitney EM, Gao SY, Yang VW. Transcriptional profiling of Kruppel-like factor 4 reveals a function in cell cycle regulation and epithelial differentiation. J Mol Biol 2003 Feb 21;326(3):665–77.
  43. 43. Shie JL, Chen ZY, Fu M, Pestell RG, Tseng CC. Gut-enriched Kruppel-like factor represses cyclin D1 promoter activity through Sp1 motif. Nucleic Acids Res 2000 Aug 1;28(15):2969–76.
  44. 44. Segre J, Bauer C, Fuchs E. Klf4 is a transcription factor required for establishing the barrier function of the skin. Nature Genetics 1999;22(4):356–60.
  45. 45. Katz J, Perreault N, Goldstein B, Lee C, Labosky P, Yang V, et al. The zinc-finger transcription factor Klf4 is required for terminal differentiation of goblet cells in the colon. Development 2002;129(11):2619–28.
  46. 46. Yet SF, McA'Nulty MM, Folta SC, Yen HW, Yoshizumi M, Hsieh CM, et al. Human EZF, a Kruppel-like zinc finger protein, is expressed in vascular endothelial cells and contains transcriptional activation and repression domains. J Biol Chem 1998 Jan 9;273(2):1026–31.
  47. 47. Peters DG, Zhang XC, Benos PV, Heidrich-O'Hare E, Ferrell RE. Genomic analysis of immediate/early response to shear stress in human coronary artery endothelial cells. Physiol Genomics 2002 Dec 26;12(1):25–33.
  48. 48. Roy S, Khanna S, Wallace WA, Lappalainen J, Rink C, Cardounel AJ, et al. Characterization of perceived hyperoxia in isolated primary cardiac fibroblasts and in the reoxygenated heart. J Biol Chem 2003 Nov 21;278(47):47129–35.
  49. 49. Nickenig G, Baudler S, Muller C, Werner C, Werner N, Welzel H, et al. Redox-sensitive vascular smooth muscle cell proliferation is mediated by GKLF and Id3 in vitro and in vivo. FASEB J 2002 Jul;16(9):1077–86.
  50. 50. Bonner ALWYM. Gene expression signatures identify novel regulatory pathways during murine lung development: implications for lung tumorigenesis. J Medical Genetics 2003;40:408–17.
  51. 51. Landesberg L, Ramalingam R, Lee K, Rosengart T, Crystal R. Upregulation of transcription factors in lung in the early phase of postpneumonectomy lung growth. Amer J Physiol Lung Cell Mol Physiol 2001;281:L1138–L1149.
  52. 52. Spira A, Beane J, Shah V, Liu G, Schembri F, Yang X, et al. Effects of cigarette smoke on the human airway epithelial cell transcriptome. Proc Natl Acad Sci USA 2004 Jul 6;101(27):10143–8.
  53. 53. Wang J, Wang S, Manzer R, McConville G, Mason RJ. Ozone induces oxidative stress in rat alveolar type II and type I-like cells. Free Radic Biol Med 2006 Jun 1;40(11):1914–28.
  54. 54. Hu B, Wu Z, Liu T, Ullenbruch MR, Jin H, Phan SH. Gut-enriched Kruppel-like factor interaction with Smad3 inhibits myofibroblast differentiation. Am J Respir Cell Mol Biol 2006 Jul 20. 2007;36(1);78–84.
  55. 55. Jean JC, George E, Kaestner KH, Brown LA, Spira A, Joyce-Brady M. Transcription factor Klf4, induced in the lung by oxygen at birth, regulates perinatal fibroblast and myofibroblast differentiation. PLoS One 2013;8(1):e54806.
  56. 56. Chen Y, Frank L. Differential gene expression of antioxidant enzymes in the perinatal rat lung. Pediatric Research 1993;34:27–31.
  57. 57. Das KC, Guo X-L, White CW. Induction of thioredoxin and thioredoxin reductase gene expression in lungs of newborn primates by oxygen. Am J Physiol 1999;276:530–9.
  58. 58. Das KC, Phl PMB, Guo X, White CW. Induction of Peroxiredoxin Gene Expression by Oxygen In Lungs of Newborn Primates. Am J Respir Cell Mol Biol 2001;In press.
  59. 59. Land SC, Wilson SM. Redox regulation of lung development and perinatal lung epithelial function. Antioxid Redox Signal 2005;7(1–2):92–107.
  60. 60. Chen X, Johns D, Geiman D, Marban E, Dang D, Hamlin G, et al. Kruppel-like factor 4 (gut-enriched Kruppel-like factor) inhibits cell proliferation by blocking G1/S progression of the cell cycle. J Biol Chem 2001;276(32):30423–8.
  61. 61. Nardell EA, Brody JS. Determinants of mechanical properties of rat lung during postnatal development. J Appl Physiol Respir Environ Exerc Physiol 1982 Jul;53(1):140–8.
  62. 62. Foronjy R, D'Armiento J. The role of collagenase in emphysema. Respir Res 2001;2(6):348–52.
  63. 63. Hinz B, Gabbiani G. Cell-matrix and cell-cell contacts of myofibroblasts: role in connective tissue remodeling. Thromb Haemost 2003 Dec;90(6):993–1002.
  64. 64. Lindahl P, Karlsson L, Hellstrom M, Gebre-Medhin S, Willetts K, Heath JK, et al. Alveogenesis failure in PDGF-A-deficient mice is coupled to lack of distal spreading of alveolar smooth muscle cell progenitors during lung development. Development 1997 Oct;124(20):3943–53.
  65. 65. McGowan SE. Extracellular matrix and the regulation of lung development and repair. FASEB J 1992;6:2895–904.
  66. 66. Wagner TE, Frevert CW, Herzog EL, Schnapp LM. Expression of the integrin subunit alpha8 in murine lung development. J Histochem Cytochem 2003 Oct;51(10):1307–15.
  67. 67. Liu Y, Sinha S, McDonald OG, Shang Y, Hoofnagle MH, Owens GK. Kruppel-like factor 4 abrogates myocardin-induced activation of smooth muscle gene expression. J Biol Chem 2005 Mar 11;280(10):9719–27.
  68. 68. Scavo LM, Ertsey R, Chapin CJ, Allen L, Kitterman JA. Apoptosis in the development of rat and human fetal lungs. Am J Respir Cell Mol Biol 1998 Jan;18(1):21–31.
  69. 69. Schittny JC, Djonov V, Fine A, Burri P. Programmed Cell Death Contributes to Postnatal Lung Development. Amer J Respir Cell Mol Biol 1998;18:786–93.
  70. 70. Toledano MB. The guardian recruits cops: the p53-p21 axis delegates prosurvival duties to the Keap1-Nrf2 stress pathway. Mol Cell 2009 Jun 26;34(6):637–9.
  71. 71. Katz JP, Perreault N, Goldstein BG, Actman L, McNally SR, Silberg DG, et al. Loss of Klf4 in mice causes altered proliferation and differentiation and precancerous changes in the adult stomach. Gastroenterology 2005 Apr;128(4):935–45.
  72. 72. Swamynathan SK, Katz JP, Kaestner KH, Ashery-Padan R, Crawford MA, Piatigorsky J. Conditional deletion of the mouse Klf4 gene results in corneal epithelial fragility, stromal edema, and loss of conjunctival goblet cells. Mol Cell Biol 2007 Jan;27(1):182–94.
  73. 73. Aggarwal NR, King LS, D'Alessio FR. Diverse macrophage populations mediate acute lung inflammation and resolution. Am J Physiol Lung Cell Mol Physiol 2014 Apr 15;306(8):L709–L725.
  74. 74. Martinez FO, Gordon S. The M1 and M2 paradigm of macrophage activation: time for reassessment. F1000Prime Rep 2014;6:13.
  75. 75. Stouch AN, Zaynagetdinov R, Barham WJ, Stinnett AM, Slaughter JC, Yull FE, et al. IkappaB kinase activity drives fetal lung macrophage maturation along a non-M1/M2 paradigm. J Immunol 2014 Aug 1;193(3):1184–93.
  76. 76. Islam MA, Uddin MJ, Tholen E, Tesfaye D, Looft C, Schellander K, et al. Age-related changes in phagocytic activity and production of pro-inflammatory cytokines by lipopolysaccharide stimulated porcine alveolar macrophages. CYTOKINE 2012 Dec;60(3):707–17.
  77. 77. Kurkjian C, Hollifield M, Lines JL, Rogosky A, Empey KM, Qureshi M, et al. Alveolar macrophages in neonatal mice are inherently unresponsive to Pneumocystis murina infection. Infect Immun 2012 Aug;80(8):2835–46.
  78. 78. Martin TR, Ruzinski JT, Wilson CB, Skerrett SJ. Effects of endotoxin in the lungs of neonatal rats: age-dependent impairment of the inflammatory response. J Infect Dis 1995 Jan;171(1):134–44.
  79. 79. Prince LR, Maxwell NC, Gill SK, Dockrell DH, Sabroe I, McGreal EP, et al. Macrophage phenotype is associated with disease severity in preterm infants with chronic lung disease. PLoS One 2014;9(8):e103059.
  80. 80. Palojarvi A, Petaja J, Siitonen S, Janer C, Andersson S. Low monocyte HLA-DR expression as an indicator of immunodepression in very low birth weight infants. Pediatr Res 2013 Apr;73(4 Pt 1):469–75.
  81. 81. Iwabuchi H, Kawasaki T, Yamamoto T, Uchiyama M, Nakata K, Naito M. Expression of PU.1 and terminal differentiation of alveolar macrophages in newborn rats. Cell Tissue Res 2007 Jul;329(1):71–9.
  82. 82. Divangahi M, King IL, Pernet E. Alveolar macrophages and type I IFN in airway homeostasis and immunity. Trends Immunol 2015 May;36(5):307–14.
  83. 83. Hussell T, Bell TJ. Alveolar macrophages: plasticity in a tissue-specific context. Nat Rev Immunol 2014 Feb;14(2):81–93.
  84. 84. Morales-Nebreda L, Misharin AV, Perlman H, Budinger GR. The heterogeneity of lung macrophages in the susceptibility to disease. Eur Respir Rev 2015 Sep;24(137):505–9.
  85. 85. Schneider C, Nobs SP, Kurrer M, Rehrauer H, Thiele C, Kopf M. Induction of the nuclear receptor PPAR-gamma by the cytokine GM-CSF is critical for the differentiation of fetal monocytes into alveolar macrophages. Nat Immunol 2014 Nov;15(11):1026–37.
  86. 86. Koli K, Myllarniemi M, Keski-Oja J, Kinnula VL. Transforming growth factor-beta activation in the lung: focus on fibrosis and reactive oxygen species. Antioxid Redox Signal 2008 Feb;10(2):333–42.
  87. 87. Brown SD, Baxter KM, Stephenson ST, Esper AM, Brown LA, Fitzpatrick AM. Airway TGF-beta1 and oxidant stress in children with severe asthma: association with airflow limitation. J Allergy Clin Immunol 2012 Feb;129(2):388–96, 396.
  88. 88. Liu RM, Vayalil PK, Ballinger C, Dickinson DA, Huang WT, Wang S, et al. Transforming growth factor beta suppresses glutamate-cysteine ligase gene expression and induces oxidative stress in a lung fibrosis model. Free Radic Biol Med 2012 Aug 1;53(3):554–63.
  89. 89. Brown SD, Brown LA. Ethanol (EtOH)-induced TGF-beta1 and reactive oxygen species production are necessary for EtOH-induced alveolar macrophage dysfunction and induction of alternative activation. Alcohol Clin Exp Res 2012 Nov;36(11):1952–62.
  90. 90. Murthy S, Larson-Casey JL, Ryan AJ, He C, Kobzik L, Carter AB. Alternative activation of macrophages and pulmonary fibrosis are modulated by scavenger receptor, macrophage receptor with collagenous structure. FASEB J 2015 Aug;29(8):3527–36.
  91. 91. Ping XD, Harris FL, Brown LA, Gauthier TW. In vivo dysfunction of the term alveolar macrophage after in utero ethanol exposure. Alcohol Clin Exp Res 2007 Feb;31(2):308–16.
  92. 92. Gauthier TW, Young PA, Gabelaia L, Tang SM, Ping XD, Harris FL, et al. In utero ethanol exposure impairs defenses against experimental group B streptococcus in the term Guinea pig lung. Alcohol Clin Exp Res 2009 Feb;33(2):300–6.
  93. 93. Gauthier TW, Ping XD, Gabelaia L, Brown LA. Delayed neonatal lung macrophage differentiation in a mouse model of in utero ethanol exposure. Am J Physiol Lung Cell Mol Physiol 2010 Jul;299(1):L8–16.
  94. 94. Konomi JV, Harris FL, Ping XD, Gauthier TW, Brown LA. Zinc insufficiency mediates ethanol-induced alveolar macrophage dysfunction in the pregnant female mouse. Alcohol Alcohol 2015 Jan;50(1):30–8.
  95. 95. Joshi PC, Applewhite L, Ritzenthaler JD, Roman J, Fernandez AL, Eaton DC, et al. Chronic ethanol ingestion in rats decreases granulocyte-macrophage colony-stimulating factor receptor expression and downstream signaling in the alveolar macrophage. J Immunol 2005 Nov 15;175(10):6837–45.
  96. 96. Yeligar SM, Harris FL, Hart CM, Brown LA. Ethanol induces oxidative stress in alveolar macrophages via upregulation of NADPH oxidases. J Immunol 2012 Apr 15;188(8):3648–57.
  97. 97. Yeligar SM, Harris FL, Hart CM, Brown LA. Glutathione attenuates ethanol-induced alveolar macrophage oxidative stress and dysfunction by downregulating NADPH oxidases. Am J Physiol Lung Cell Mol Physiol 2014 Mar 1;306(5):L429–L441.
  98. 98. Yeligar SM, Mehta AJ, Harris FL, Brown LA, Hart CM. PPARgamma regulates chronic alcohol-induced alveolar macrophage dysfunction. Am J Respir Cell Mol Biol 2015 Dec 17. 2015 Dec17-DOI 10.1165/rcmb.2015-0077OC.
  99. 99. Staitieh BS, Egea EE, Fan X, Azih N, Neveu W, Guidot DM. Activation of alveolar macrophages with interferon-gamma promotes antioxidant defenses via the Nrf2-ARE pathway. J Clin Cell Immunol 2015 Oct;6(5).
  100. 100. Curry-McCoy TV, Guidot DM, Joshi PC. Chronic alcohol ingestion in rats decreases Kruppel-like factor 4 expression and intracellular zinc in the lung. Alcohol Clin Exp Res 2013 Mar;37(3):361–71.
  101. 101. Collard KJ, Godeck S, Holley JE, Quinn MW. Pulmonary antioxidant concentrations and oxidative damage in ventilated premature babies. Arch Dis Child Fetal Neonatal Ed 2004 Sep;89(5):F412–F416.
  102. 102. Grigg J, Barber A, Silverman M. Bronchoalveolar lavage fluid glutathione in intubated premature infants. Arch Dis Child 1993 Jul;69(1 Spec No):49–51.
  103. 103. Jain A, Mehta T, Auld PA, Rodrigues J, Ward RF, Schwartz MK, et al. Glutathione metabolism in newborns: evidence for glutathione deficiency in plasma, bronchoalveolar lavage fluid, and lymphocytes in prematures. Pediatr Pulmonol 1995 Sep;20(3):160–6.
  104. 104. Reise JA, Taylor GW, Fardy CH, Silverman M. Glutathione and neonatal lung disease. Clin Chim Acta 1997 Sep 8;265(1):113–9.
  105. 105. Rosso MI, Roark S, Taylor E, Ping X, Ward JM, Roche K, et al. Exhaled breath condensate in intubated neonates—a window into the lung's glutathione status. Respir Res 2014;15:1.
  106. 106. Kwong KY, Niang S, Literat A, Zhu NL, Ramanathan R, Jones CA, et al. Expression of transforming growth factor beta (TGF-b1) by human preterm lung inflammatory cells. Life Sci 2006 Nov 17;79(25):2349–56.
  107. 107. Strandjord TP, Clark JG, Guralnick DE, Madtes DK. Immunolocalization of transforming growth factor-alpha, epidermal growth factor (EGF), and EGF-receptor in normal and injured developing human lung. Pediatr Res 1995 Dec;38(6):851–6.

Written By

Jyh-Chang Jean, Lou Ann Scism Brown and Martin Joyce-Brady

Submitted: 08 February 2016 Reviewed: 20 April 2016 Published: 31 August 2016