Next Article in Journal
Computational Analysis of Structure-Based Interactions for Novel H1-Antihistamines
Next Article in Special Issue
Mesenchymal Stem Cells Preserve Working Memory in the 3xTg-AD Mouse Model of Alzheimer’s Disease
Previous Article in Journal
The Complex Relationship between Metals and Carbonic Anhydrase: New Insights and Perspectives
Previous Article in Special Issue
Survivin Modulates Squamous Cell Carcinoma-Derived Stem-Like Cell Proliferation, Viability and Tumor Formation in Vivo
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Lung Regeneration: Endogenous and Exogenous Stem Cell Mediated Therapeutic Approaches

1
Academic Unit of Respiratory Medicine, University of Sheffield, Sheffield S10 2RX, UK
2
Department of Respiratory Medicine, University Hospitals of North Midlands, Stoke-on-Trent ST4 6QG, UK
3
Guy Hilton Research Centre, Institute of Science and Technology in Medicine, Keele University, Stoke-on-Trent ST4 7QB, UK
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2016, 17(1), 128; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms17010128
Submission received: 7 December 2015 / Revised: 7 January 2016 / Accepted: 11 January 2016 / Published: 19 January 2016
(This article belongs to the Special Issue Stem Cell Activation in Adult Organism)

Abstract

:
The tissue turnover of unperturbed adult lung is remarkably slow. However, after injury or insult, a specialised group of facultative lung progenitors become activated to replenish damaged tissue through a reparative process called regeneration. Disruption in this process results in healing by fibrosis causing aberrant lung remodelling and organ dysfunction. Post-insult failure of regeneration leads to various incurable lung diseases including chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis. Therefore, identification of true endogenous lung progenitors/stem cells, and their regenerative pathway are crucial for next-generation therapeutic development. Recent studies provide exciting and novel insights into postnatal lung development and post-injury lung regeneration by native lung progenitors. Furthermore, exogenous application of bone marrow stem cells, embryonic stem cells and inducible pluripotent stem cells (iPSC) show evidences of their regenerative capacity in the repair of injured and diseased lungs. With the advent of modern tissue engineering techniques, whole lung regeneration in the lab using de-cellularised tissue scaffold and stem cells is now becoming reality. In this review, we will highlight the advancement of our understanding in lung regeneration and development of stem cell mediated therapeutic strategies in combating incurable lung diseases.

Graphical Abstract

1. Introduction

The tissue turnover of unperturbed adult lung is remarkably slow. However, after injury or insult a specialised group of facultative lung progenitors become activated to replenish damaged tissue through a reparative process called regeneration. Interruption in this process results in healing by fibrosis causing scarring, aberrant lung remodelling and organ dysfunction [1,2]. A highly specialised group of endogenous lung stem/progenitor cells are believed to maintain lung homeostasis and regeneration. In this review we will discuss our latest understanding of the identity of lung stem cells, their activation and proliferation with subsequent differentiation to replenish or repair injured lung tissues. This will include a discussion of lung development and the associated signalling that governs this process. Evidence suggests that in some disease conditions, such as COPD, the embryonic lung developmental process could be reactivated [3,4,5]. Furthermore, for de novo derivation of lung progenitors from pluripotent embryonic stem cells (ESCs) and inducible pluripotent stem cells (iPSC) in vitro, it is essential to understand the molecular pathways that regulate differentiation of adult lung cells from a common embryonic foregut endoderm origin. Exogenous stem cells, such as mesenchymal stem cells (MSCs) and ESCs are involved in research designed to identify clinical applications in lung disease and this will also be investigated.

2. Lung Development and Associated Signalling Pathways

Mammalian lung development is a complex but continuous process that starts at approximately embryonic day 9 (E9.0) in mice, and at an undefined early time in human, and continues through infancy [6]. The early embryonic and pseudoglandular stages elaborate the conducting airways, and in later stage canalicular, saccular and alveolar development takes place through reduction of mesenchyme and formation of pulmonary vasculature to form a thin air-blood barrier. A new-born infant possesses approximately 50 million alveoli but will add another 250 million alveoli through infancy increasing surface area from approximately 3 to 70 m2. In a completely developed lung there are more than 40 different cell types, with different functions [7].
During embryonic weeks 3 to 7, the human lung originates as a ventral diverticulum arising from the foregut endoderm, which bifurcates and gives rise laterally to the two primary bronchial buds (Figure 1). The lung buds grow into surrounding splanchnic mesoderm and branch repeatedly, giving rise to the future respiratory tree. This primitive lung bud is lined by epithelium derived from foregut endoderm, which later differentiates into specialised cells that line both the conducting and respiratory airways [7,8]. On the other hand, mesenchymal cells condensed around the primitive airways differentiate into blood vessels, smooth muscle, cartilage, and other connective tissues of the lung [9]. During embryonic weeks 7 to 16, an extensive branching morphogenesis takes place through rapid proliferation of the primitive airways. This bronchial tree branching pattern does not change after this stage and remains more or less unchanged throughout the adult life [10]. This is the pseudoglandular stage. Here, the most peripheral structures including the terminal bronchioles, respiratory bronchioles and alveolar ducts form. The cartilage begins to form around the larger airways and smooth muscle forms around airways and major blood vessels. In this phase, the airways are lined by columnar epithelium with few ciliated and goblet cells [10,11].
During the canalicular and saccular stages (embryonic weeks 16 to 35), the pulmonary acinar units form which contain alveolar ducts, alveolar sacs and alveoli. A gradual decrease in mesenchymal tissue results in close apposition of the pulmonary vasculature to the alveolar walls. At this time, type I alveolar epithelial cell (AECI) and type II AEC (AECII) are differentiated from the cuboidal epithelial cells in the most peripheral parts of the lung. Lamellar bodies associated with surfactant synthesis begin to appear in the cytoplasm of AECII cells and AECI cells begin their flattening process and attenuate to provide an air-blood interface [11]. This alveolar development continues after birth and is completed through infancy [12].
During lung organogenesis, the proximodistal patterning, branching morphogenesis, alveolarisation and cellular differentiation are strictly regulated by the surrounding mesenchyme and a number of key growth factor-directed signalling pathways [13]. Cumulative data from several recombination experiments reveal that mesenchyme is essential for initial lung budding and subsequent branching of foregut endoderm, where the governing stimulation is carried out by the qualitative and quantitative differential distribution of surrounding mesenchyme [14,15,16,17]. This reciprocal endoderm-mesoderm cross-talk is likely mediated by numerus soluble growth factors and transcription factors originating from both epithelial and mesenchymal cells (Figure 1) [18,19,20]. Mouse embryonic lung organ culture experiments demonstrate that the quantity of mesenchyme specifies epithelial cell fate during differentiation. That is, a small amount of mesenchyme is able to direct cell differentiation toward a bronchiolar phenotype; whereas, an increased amount of the same mesenchyme is able to induce the epithelial cells to differentiate toward an alveolar phenotype [21]. Furthermore, tissue recombination experiments show that distal lung mesenchyme can reprogram rat tracheal epithelial cells to an AECII cell phenotype, and conversely, proximal mesenchyme can induce distal lung epithelium towards a tracheal epithelial cell phenotype [22,23]. These findings highlight the significance of the epithelial-mesenchymal interaction in coordinating the precise temporospatial pattern of lung development.
Figure 1. Epithelial-mesenchymal cross-talk and governing signalling during early development and branching morphogenesis of lung. Factors are represented only at sites where the expression is most abundant. Fibroblast growth factor-10 (FGF-10) is highly expressed in the distal mesenchyme and acts as a chemotactic focus for the epithelium during lung budding. FGF-10 also regulates Sox9 expression in the distal epithelial progenitors and induces bone morphogenetic protein (BMP)-4 expression. Sox2 expression in the proximal epithelium is under regulation of histone deacetylases 1/2 (HDAC1/2) signalling. FGF-10 expression in the mesenchyme is regulated by Wnt/β-catenin signalling (red arrow). A high concentration of BMP-4 signal also serves to locally inhibit endoderm proliferation, thereby inducing the lateral outgrowth of new airway branches. Sonic hedgehog (Shh) at the distal tips functions to downregulate FGF-10 expression in the mesenchyme, which limits local budding. Transforming growth factor-β (TGF-β) signalling also prevents local budding, by decreasing endodermal proliferation and by stimulating synthesis of matrix components at branch points. Solid arrows indicate sources from and influences on cells/molecules; dotted arrow indicates direction of patterning.
Figure 1. Epithelial-mesenchymal cross-talk and governing signalling during early development and branching morphogenesis of lung. Factors are represented only at sites where the expression is most abundant. Fibroblast growth factor-10 (FGF-10) is highly expressed in the distal mesenchyme and acts as a chemotactic focus for the epithelium during lung budding. FGF-10 also regulates Sox9 expression in the distal epithelial progenitors and induces bone morphogenetic protein (BMP)-4 expression. Sox2 expression in the proximal epithelium is under regulation of histone deacetylases 1/2 (HDAC1/2) signalling. FGF-10 expression in the mesenchyme is regulated by Wnt/β-catenin signalling (red arrow). A high concentration of BMP-4 signal also serves to locally inhibit endoderm proliferation, thereby inducing the lateral outgrowth of new airway branches. Sonic hedgehog (Shh) at the distal tips functions to downregulate FGF-10 expression in the mesenchyme, which limits local budding. Transforming growth factor-β (TGF-β) signalling also prevents local budding, by decreasing endodermal proliferation and by stimulating synthesis of matrix components at branch points. Solid arrows indicate sources from and influences on cells/molecules; dotted arrow indicates direction of patterning.
Ijms 17 00128 g001
At least 11 different epithelial cell types have been identified in the conducting and respiratory portions of the lung [24]. The basal, secretory, and ciliated cells are the major cell types constituting the pseudostratified epithelium of the proximal tracheobronchial area while AECI and AECII cells make up the alveolar epithelium [25]. The onset of cell differentiation is signalled by various gene products including surfactant protein (SP)-A, SP-B, SP-C, SP-D and club (Clara) cells secretory protein (CCSP) [26,27,28,29]. During early embryonic development, the undifferentiated epithelium co-expresses several lineage markers including SP-A, CCSP and neuroendocrine cell markers. But later, during the pseudoglandular phase, this epithelium differentiates into different lineages and becomes restricted to proximal and distal airways [30]. Transcriptional analysis reveals that the surfactant proteins and CCSP genes share common mechanisms for gene regulation and expression in respiratory epithelial cells. Nkx2.1 (also known as thyroid transcription factor-1 (TTF-1)), a homeodomain protein, plays a pivotal role in several phases of lung development, including epithelial cell lineage determination [31,32,33,34]. Nkx2.1 binds and activates SP-A, SP-B, SP-C, and CCSP promoter elements as well as Nkx2.1 itself [35,36,37,38,39]. During development of the respiratory epithelium, Nkx2.1 appears to be the earliest marker, with onset of expression at the time of lung bud formation from the foregut endoderm [40]. Expression of Nkx2.1 is present throughout the foetal lung including respiratory epithelial cells of the trachea, bronchi, and developing respiratory tubules, but its expression is most robust in distal alveolar epithelium [24,40,41]. Null mutation of Nkx2.1 in mice exhibit tracheoesophageal fistulas and fail to form bronchiolar and alveolar structures [32,33,34]. Pulmonary-specific gene expression including SP-B, SP-C, and CCSP is extinguished within transgenic lungs, which do, however, contain ciliated and mucus-secreting cells [34]. Thus, Nkx2.1 is recognised as a “master gene” in maintaining the lung morphogenesis as well as cytodifferentiation of certain epithelial cell lineages [24]. However, targeted gene mutation studies confer that while Nkx2.1 is not required for initial specification of lung primordia it is essential for pulmonary development and cell differentiation [33,42].
The precise regulatory function of Nkx2.1 in pulmonary cytodifferentiation is not well understood; however, study reveals that Nkx2.1 has multiple binding sites for both ubiquitous and specific transcription factors, including those of the hepatocyte nuclear factor (HNF) and GATA zinc finger families [43,44,45]. GATA and HNF play crucial role for the development of the foregut endoderm [46,47,48]. Multiple studies have identified HNF-3 binding sites in the SP-A, SP-B, and CCSP promoter regions [35,49,50]. The HNF-3β null mutation results in an early embryonic lethal phenotype with primitive foregut deformities, resulting in agenesis of lung and other foregut derivatives [51]. While Nkx2.1, GATA and HNF play crucial role in cytodifferentiation and specification of cell fate, the Homeobox (Hox) genes regulate the patterning of the lung during embryonic development. Hox genes act as transcription factors and are consistently expressed throughout the lung during development and maintain proximal-distal orientation of the lung as well as branching morphogenesis [52,53,54]. Hoxb-3 and Hoxb-4 genes are expressed both in the proximal and distal mesenchyme of the entire developing lung; whereas, Hoxb-2 and Hoxb-5 are restricted within the mesenchyme of distal lung buds (Figure 1) [52]. Hoxb-3 transactivates the Nkx2.1 promoter, which suggests that Hoxb-3 could regulate proximal-distal lung patterning in an Nkx2.1 depended manner [24,31]. Mouse embryonic lung culture experimentation has demonstrated that retinoic acid induces Hoxa-5, Hoxb-5 and Hoxb-6 gene expression; whereas, Hoxb-5 is negatively regulated by epidermal growth factor (EGF) and transforming growth factor-β (TGF-β) [55,56]. Retinoic acid has been demonstrated to facilitate the growth of proximal airways and gene expression at the expense of distal structures in a dose-dependent manner; therefore, it is probable that Hox genes mediate the retinoic acid-induced alteration in lung patterning [57,58].
Bone morphogenetic protein (BMP)-4, a member of the TGF-β family proteins, is also implicated in the control of the proximal-distal patterning of the lung and in branching morphogenesis [58,59]. BMP-4 gene expression is restricted to the tips of distal buds and to the adjacent mesenchyme, which locally inhibits endoderm proliferation and forces the outgrowth of lateral branches (Figure 1) [58]. Moreover, inhibition of BMP signalling results in complete proximalization of the respiratory epithelium, including ciliated cells in the most distal portions of lungs. Therefore, it is hypothesised that BMP proteins provide a concentration gradient to regulate proximal versus distal lung endoderm differentiation [59]. Endodermal cells located at the periphery of the lung, which are exposed to high levels of BMP-4, maintain a distal identity while cells below a certain threshold of the BMP-4 signal initiate a proximal differentiation program [24].
The Sox2 and Sox9 transcription factors mark lung bud endoderm as proximal and distal epithelial progenitors respectively (Figure 1). Sox2 regulates the differentiation of proximal progenitors into secretory and ciliated epithelium; whereas, Sox9 directs distal progenitors into alveolar epithelial cells [60,61,62,63,64,65]. During early lung development, fibroblast growth factor-10 (FGF-10), which is highly expressed in the distal mesenchyme and is regulated by Wnt signalling (Figure 1), acts on the distal lung epithelial progenitors to maintain them and prevent them from differentiating into proximal airway epithelial cells by inducing Sox9 and repressing Sox2 expression [66,67,68,69,70]. When the lung epithelium extends into the mesenchyme, more proximally located cells become less exposed to distally sourced FGF-10 and gradually start to differentiate [20,69,70,71,72]. In contrast, studies show that suppression of FGF-10 around the developing airway, as well as during late gestation and postnatal development, facilitates proper maturation of the lung epithelium [73,74,75,76]. Furthermore, ectopic overexpression of FGF-10 at later stages of lung development blocks the alveolar differentiation program via induction of Sox9 expression [66,72,77]. Therefore, FGF-10 is thought to maintain distal epithelial progenitor population helping branching morphogenesis during early lung development in a Sox9 dependent manner [78].
While Sox9+ progenitors are largely under regulation of FGF-10, the Sox2+ progenitors in the lung endoderm are regulated by histone deacetylases 1 (HDAC1) and HDAC2 (Figure 1). HDAC1/2 deficiency leads to a loss of Sox2 expression resulting in suppression of proximal airway development. This is mediated in part by de-repression of BMP-4, which is direct transcriptional target of HDAC1/2. In contrast to development, postnatal loss of HDAC1/2 in airway epithelium does not affect the expression of Sox2 or BMP-4 [79].
Despite high-sequence homology and affinity for the same cognate receptor, FGF-7 and FGF-10 exert distinct effects on the developing lung. FGF-7 predominantly influences airway branching by promoting epithelial cell proliferation and expansion [18,80,81]. FGF-7 acts as differentiation factor in developing lung. Lung organotypic culture experiments show that in absence of any mesenchyme or serum, exogenous application of FGF-7 can induce AECII-like epithelial cell differentiation [81,82]. During postnatal life, alveolarisation is regulated by cooperative interaction of various FGFs with FGFR-3 and FGFR-4 receptors [83]. Double mutation of these receptors results in postnatal pulmonary lethality where alveolarisation is severely disrupted leading to an emphysematous phenotype. This transgenic mouse phenotype also exhibits an abnormal elastin synthesis causing a lethal form of parenchymal defect [83]. Furthermore, inactivation of the epidermal growth factor (EGF) receptors affects AECII cell maturity, with decreased expression of the SP-C protein. In contrast, exogenous EGF administration accelerates alveolar type-II cell differentiation in foetal lungs [84].

3. Endogenous Stem Cell Population of the Respiratory System

Endogenous adult lung stem and progenitor cells are regenerative cell populations important for epithelial cell homeostasis and injury repair. In multiple adult organs, tissue-specific stem cells have been identified as multipotent cells with the capacity for long-term self-renewal and the ability to differentiate into other cell lineages. Tissue-specific stem cells are typically quiescent in normal conditions and proliferate during injury repair [85,86,87]. The unperturbed adult lung is a remarkably quiescent. Therefore, the detection of proliferative postnatal stem or progenitor cells that actively take part in repair and regeneration has been challenging. Numerous studies attempting to identify adult stem or progenitor candidates in the lung have used injury and disease models, primarily in rodents. To date, studies of human and animal lung morphology have identified subsets of lung epithelial cells with self-renewal and differentiation capacity, including basal cells (BC), secretory club cells (formerly known as Clara cells) of the proximal and distal airways, and AECII cells of the alveoli (Figure 2) [6]. All of these cells have the capacity to enter the cell cycle in response to lung injury [88,89,90,91,92,93,94,95]. More recently, advanced lineage tracing techniques have suggested that most types of lung epithelial cells, except for ciliated cells, can proliferate and expand after injury to promote repair [96,97,98,99,100]. Identifying these stem/progenitor cell populations and understanding their healing mechanism is important to harness the endogenous abilities of the lung to regenerate from a therapeutic perspective.

3.1. Trachea and Proximal Airways Stem Cells

The trachea and the principal bronchi (proximal airways) of both humans and mice are lined with a pseudostratified epithelium composed of basal and luminal cells. Luminal airway cells include secretory club cells, ciliated cells and neuroendocrine cells. In the humans and other mammals, the airway epithelium also contains SPDEF and MUC5AC expressing goblet cells. In this region, secretory club cells are marked by expression of secretoglobin family 1a member 1 (Scgb1a1+) [6]. The basal cells, on the other hand, are believed to be the most important stem cells of upper airways, marked by the expression of basal cell-restricted transcription factor p63, keratin (Krt)-5 (Krt5), Krt14, nerve growth factor receptor (NFGR) and cell-surface marker Pdpn (Figure 2A) [101,102,103]. This basal cell population, both in mice and human, have extensive proliferative potential, self-renewal capacity and ability to differentiate into secretory club and ciliated cells [102,104]. The 3D “tracheosphere” culture of FACS-sorted pure population of Krt5+p63+NGFR+ basal cells shows clonal expansion of differentiated cells which are positive for both ciliated and secretory club (Clara) cells [102,103].
Figure 2. Endogenous stem cells in the airways and alveoli with their differentiation in the postnatal lung. (A) The trachea and bronchi of the rodent and human lung are lined with multiple epithelial lineages. Basal cells are located in this region and can generate secretory club and ciliated cell lineages. Notch signalling is crucial for differentiation of basal cells and also suppresses ciliated cell differentiation. HDAC1 and HDAC2 are essential for secretory epithelial regeneration; (B) The bronchiolar lining epithelium of rodents lacks basal cells but contains variant club cells, secretory cells and ciliated cells; (C) Progenitor cell populations and their differentiated progeny in the lung alveolus. The alveolar epithelium consists of alveolar epithelial cell AECI and AECII cells. AECII cells can generate AECI cells during homeostasis and after injury. Generation of alveolar epithelium by other cells, such as BASCs has yet to be supported by lineage tracing. The AECII to AECI differentiation signalling is not clear (marked by “?”), but AECII self-renewal and proliferation occurs via EGFR/KRAS signalling. Curved arrows indicate differentiation and line arrows indicate molecular or physical stimulus.
Figure 2. Endogenous stem cells in the airways and alveoli with their differentiation in the postnatal lung. (A) The trachea and bronchi of the rodent and human lung are lined with multiple epithelial lineages. Basal cells are located in this region and can generate secretory club and ciliated cell lineages. Notch signalling is crucial for differentiation of basal cells and also suppresses ciliated cell differentiation. HDAC1 and HDAC2 are essential for secretory epithelial regeneration; (B) The bronchiolar lining epithelium of rodents lacks basal cells but contains variant club cells, secretory cells and ciliated cells; (C) Progenitor cell populations and their differentiated progeny in the lung alveolus. The alveolar epithelium consists of alveolar epithelial cell AECI and AECII cells. AECII cells can generate AECI cells during homeostasis and after injury. Generation of alveolar epithelium by other cells, such as BASCs has yet to be supported by lineage tracing. The AECII to AECI differentiation signalling is not clear (marked by “?”), but AECII self-renewal and proliferation occurs via EGFR/KRAS signalling. Curved arrows indicate differentiation and line arrows indicate molecular or physical stimulus.
Ijms 17 00128 g002
An H1N1 influenza virus-induced mouse lung injury model recently demonstrated that following a sub-lethal viral infection p63+ basal cells in the bronchiolar epithelium underwent rapid proliferation and migration to damaged alveolar regions. Then, these migrated cells assemble into discrete, Krt5+ pods and express typical alveoli-associated markers. This study suggests that this p63+Krt5+ cell population plays a role as an intermediate in the reconstitution of the alveolar-capillary network eradicated by viral infection [105]. On the other hand, lineage-tracing studies in mice show that in an injury condition Scgb1a1+ secretory club cells can give birth to goblet cells, but goblets cannot be derived from ciliated cells [106]. Therefore, these basal and club cell population are considered as major stem/progenitor cells in the upper airways. The molecular mechanisms for basal cell self-renewal and differentiation are poorly understood. However, mouse tracheobronchial epithelium shows that Notch signalling is greatly enhanced during repair and is required for activation in basal cells promoting their luminal differentiation, primarily toward secretory lineages (Figure 2A). Signalling is also conserved in basal cells of the human airway [107].

3.2. Distal Airway Stem Cells

The distal bronchiolar region of mouse lungs are lined with a single-layered epithelium consisting of secretory club cells (Scgb1a1+ and Scgb3a2+), ciliated cells (Foxj1+ and Tubb4+) and neuroendocrine (NE) epithelial cells (CGRP+) (Figure 2B). In human, an additional population of Krt5+p63+ basal cells are also present in the bronchiolar lining of the airways, but their presence has not been well documented in the rodent lung [108]. Neuroendocrine cells are clustered within the epithelium as neuroendocrine bodies (NEBs) and are marked by expression of calcitonin gene-related peptide (CGRP). Lineage tracing studies show that CGRP+ NE cells are capable of self-renew in uninjured adult lung and can differentiate into secretory club and ciliated cells after naphthalene injury [109].
The unperturbed bronchiolar epithelium is remarkably quiescent. However, depletion of bronchiolar epithelium by naphthalene-induced injury spares a subset of secretory club cells that express Scgb1a1 but not cytochrome p450. These Scgb1a1+p450 club cells, which survive from naphthalene toxicity, are designated as facultative progenitors; and are capable of expanding rapidly to regenerate the damaged airways by differentiating into new secretory club and ciliated cell populations (Figure 2B) [108,110]. Moreover, Scgb1a1+p450 club cell populations also can self-renew and differentiate into ciliated epithelium during normal homeostatic turnover [111]. This particular subset of naphthalene resistant club cells is often called “variant club cells”. Interestingly, these variant club cells are mostly localised surrounding NEBs and at the bronchoalveolar duct junctions (BADJs) [110,112]. Activity of Notch signalling in the NEB microenvironment has recently been identified [113,114]. Parabronchial smooth muscle cells, which line most of the proximal airways, were proposed to help make up a niche capable of activating variant club cells. It has been shown that naphthalene injury stimulates FGF-10 secretion by the parabronchial smooth muscle, which appears to be necessary for regeneration of variant club cells [115].
The degree of injury has differential effects in its modulation of secretory club cell mediated airway regeneration. For instance, mild injury initiates proliferation and expansion of randomly distributed club cells, which in turn differentiate into ciliated epithelium and thus promote repair. On the other hand, severe naphthalene injury results in activation of variant club cells located around NEBs and within the BADJ which then regenerate the injured tissues [116]. A growing body of literature supports the notion that region-specific variant club cells of the NEB and BADJ niches preferentially contribute to bronchiolar repair and regeneration after significant depletion of secretory cells, most likely via Notch and FGF-10 regulation [6].

3.3. Stem Cells of the Alveolar Region

Pulmonary gas exchange occurs in alveolar sacs which are lined by squamous type AECI and cuboidal AECII cells. Squamous AECI cells are terminally differentiated cells that line approximately 95% of the alveolar surface through which gas exchange takes place; whereas, AECII cells secret surfactant proteins that prevent alveolar collapse [117]. Classically, AECII cells are categorised as the progenitors for AECI cells which replenish alveolar lining during normal tissue turnover and during repair after injury (Figure 2C) [118]. Significant numbers of in vivo studies have demonstrated that postnatal injury to alveoli initiates AECII cell proliferation and differentiation into AECI cells in the attempt of alveolar repair [93,94,119,120,121,122,123]. Recent genetic lineage-tracing experiments, combined with in vitro experiments, showed that SP-C positive AECII cells self-renew and differentiate into type I AEC progeny over about a year in normal conditions, however, if many AECII cells are specifically ablated, the individual survivor AECIIs undergo rapid clonal expansion and daughter cell dispersal. In addition, when Individual lineage-labelled AECIIs are placed into 3D culture, those cells give rise to self-renewing “alveolospheres”, containing cells which express both AECII and AECI specific markers [124].
Lineage tracing and clonal analysis have demonstrated that during development, AECI and AECII cells arise directly from a bipotent progenitor cell; whereas, after birth new AECI cells derive from a rare subtype of, self-renewing, long-lived, mature AECII cell that produces slowly expanding clonal foci of alveolar renewal. This stem-cell like AECII function is broadly activated by hyperoxic AECI injury. Furthermore, purified EGFR ligands, such as TGF-α, heparin-binding EGF, and NRG1 stimulate AECII proliferation in vitro. However, none of these EGFR ligands can induce AECII differentiation into AECI cells. On the other hand, oncogenic KRAS has shown to stimulate multifocal clonal AECII cell expansion in vivo [125]. Thus, it has been postulated that alveolar injury induces a signalling pathway transduced by EGFR-KRAS that controls AECII self-renewal, whereas a different signal (as yet unknown) controls AECII differentiation into AECI cell type (Figure 2C) [125].
In the mouse, another population of cells which co-express Scgb1a1 and SP-C markers are found at the BADJ area designated as bronchioalveolar stem cells (BASCs) (Figure 2C). These Scgb1a1+Sp-C+ BASCs can expand in vivo after bleomycin-induced lung injury, and the isolated cells can be clonally expanded in vitro, where they show multipotent differentiation into bronchiolar and alveolar lineages [126]. Lineage-tracing studies demonstrate that during development or after hyperoxic alveolar injury, the BASCs do not contribute in alveolar reconstitution [71]. In contrast, bleomycin-induced lung injury model shows that the BASCs are capable of generating AECIIs, although their contribution is limited, and they do not seem to expand substantially [127,128]. Furthermore, co-expression of Scgb1a1 and SP-C is not unique to BASCs, occasionally some AECIIs found throughout mouse alveolar regions also co-express both Scgb1a1 and SP-C [127]. As Scgb1a1 lineage tracing does not entirely distinguish airway BASCs from rare AECII cells, further confirmation is required before defining BASCs as alveolar stem cells [6].

3.4. Lung Mesenchymal Stem Cells

Mesenchymal stem cells (MSCs) are present in the stromal tissue of many organs, including bone marrow, adipose tissue, muscle, periosteum, dermis, and lung [129]. A multitude of studies have confirmed the presence of MSCs in the lungs of mice [130,131,132,133,134], humans [135,136,137], and sheep [138,139]. Lung-derived MSCs share some common features with conventional bone marrow-derived MSCs and fulfil the International Society of Cellular Therapy criteria for MSCs, including marker profile and differentiation capacity [140]. Lung MSCs are also clonogenic when grown on Matrigel with bFGF or hepatocyte growth factor [141,142]. However, the function of these lung MSCs is not completely understood.
Lung MSCs localised at the distal tip of the branching airway epithelium secrete FGF-10, a critical component of the signalling network that regulates BMP, Wnt, and sonic hedgehog (Shh) pathways necessary for cytodifferentiation in the developing lung [143]. Lung MSCs also support the proliferation and differentiation of airway epithelial stem cells in co-culture [144]. Recently, Hoffman and colleagues showed that intravenous administration of lung MSCs alleviates elastase-induced emphysema and increases survival through a paracrine-mediated anti-inflammatory mechanism [145].

4. Signalling Pathways Involved in Postnatal Lung Injury Repair and Regeneration

To understand the molecular mechanisms that regulate the activation, expansion, and differentiation of stem and progenitor cells in response to injury and repair is crucial to aid development of targeted therapeutic strategies to combat lung diseases. The surrounding mesenchyme also plays critical role in promoting lung stem cells during injury repair [146]. Unilateral pneumonectomy (PNX) demonstrated that resection of a single lobe, leaving other lobes, intact, resulted in regrowth of alveoli within the remaining lungs [147,148,149]. This “neoalveolarisation” occurs through activation and proliferation of bronchiolar cells, AECII cells, endothelial cells and PDGFRα+ stromal cells [150,151,152,153,154,155]. The post-PNX progenitor cell activation and alveolar regeneration is believed to be regulated by elastin content, and EGF/FGF mediated signalling pathways [156,157]. Other signalling pathways, including Wnt and Notch, are known to play important roles in stem cell self-renewal and differentiation and to play key roles in lung repair and regeneration.

4.1. Wnt Signalling

Wnt signalling is an essential regulator of early lung endoderm specification and development and has been implicated in regulating regenerative responses [146]. During postnatal lung repair and regeneration canonical Wnt signalling is activated in the epithelium of various lung compartments [158,159,160,161,162]. Naphthalene-induced selective depletion of secretory club cells of airway epithelium activates Wnt signalling. This activation of Wnt signalling through loss of the transcription factor GATA6 leads to expansion of the BASCs population after naphthalene injury [163]. However, canonical Wnt signalling may not be essential for lung regeneration as postnatal deletion of β-catenin does not obliterate secretory cell proliferation [163]. Moreover, loss of β-catenin in postnatal alveolar epithelium leads to increased fibrosis and AEC death [164]. In addition, blocking of Wnt signalling by pharmacologically active molecules can reduce bleomycin-induced lung fibrosis [165]. Furthermore, increased activation of Wnt signalling has been associated with idiopathic pulmonary fibrosis (IPF) lesions [166]. Therefore, precaution should be taken with the manipulation of Wnt signalling in the activation of lung progenitors to promote lung regeneration. Excessive activation of Wnt signalling may lead to fibrosis and other deleterious effects [6].

4.2. Notch Signalling

Notch signalling plays an important role in airway epithelial regeneration. During lung development, Notch signalling is essential for differentiation of the secretory epithelium from proximal lung endoderm [167,168]. In postnatal lung injury, Notch signalling is essential for differentiation of basal cells into secretory cells [107]. Increased Notch activation can expand the secretory lineage at the expense of the ciliated lineage [107,169]. In addition, reactive oxygen species (ROS) activate Notch signalling via Nrf2 activation. The ROS-Notch pathway is important for BC self-renewal through regulation of cell proliferation, which may be critical for maintaining the BC population in the upper airways of rodent and human lungs [170]. This unique ability of Notch to promote a specific secretory cell lineage differentiation may offer a potential therapeutic target to activate lung resident stem cells towards promotion of airway epithelial regeneration after acute lung injury [6].

4.3. Histone Deacetylases (HDACs) Signalling

Histone deacetylases (HDACs) are enzymes that eliminate acetyl groups from histones and modulate gene function. Genetic deletion of HDAC1 and HDAC2 in postnatal secretory club cells in mice led to the induction of tumour suppressor genes Rb1, p21, and p16 after naphthalene injury, which resulted in inhibition of club cell proliferation and permanently impaired epithelial regeneration [79]. This finding indicates that HDAC1 and HDAC2 may play important role in airway epithelial regeneration after injury in postnatal lung. HDAC may also play important role in the regulation of AECII function during development and regeneration in a Hop-dependent manner. Hop is an unusual homeodomain protein expressed in airway epithelium coincident with HDAC2, acting downstream of Nkx2.1 and GATA6 signalling pathways to negatively regulate surfactant protein expression by the AECII cells. Loss of Hop expression in vivo results in defective AECII cell development with increased surfactant production and disrupted alveolar formation. This Hop-mediated regulation of alveolar maturation is found to be HDAC-dependent [171]. Individuals with COPD have a significant decrease in HDAC activity in their lung epithelia with an associated complete loss of HDAC2 expression [172]. In asthma, bronchial epithelium displays reduction of HDAC activity as well [173]. Taken together, this suggests that HDACs have an important role in pulmonary epithelium homeostasis and repair. Therefore, pharmacological interventions targeting HDACs to increase histone acetylation and gene expression could have therapeutic benefits in promoting lung regeneration.

4.4. miRNAs and lncRNAs Regulation

Noncoding micro RNAs (miRNAs) and long noncoding RNAs (lncRNAs) play crucial roles in epithelial branching and differentiation during lung development. Two miRNA clusters, miR17-92 and miR302-367 play important roles in lung epithelial proliferation and differentiation. Both of these clusters are highly expressed during early lung development and then are significantly downregulated by birth [174,175]. Overexpression of these miRNA clusters results in increased lung epithelial proliferation at the expense of differentiation inhibition [174,176]. On the other hand, a recent study identified hundreds of lncRNAs expressed in the developing and postnatal lung [177]. A subset of these lncRNAs is located near to transcription factors Nkx2.1 and Foxf1. One of these lncRNAs, called NANCI, has been found to regulate SP-C and Scgb1a1 expression. Therefore, modulation of noncoding RNA candidates could be another way to activate and pulmonary regeneration process during disease condition as therapeutics [146].

5. Effects of Matrix Components in Lung Regeneration

The focus of this review is primarily to discuss the cellular components and their participation in lung regeneration; however extracellular matrix (ECM) also plays an important role. The ECM components play key roles in lung development, injury repair, regeneration and disease pathogenesis [178]. The major components of the lung ECM are collagen and elastin, interwoven with fibronectin and proteoglycans, that provide plasticity of the lung parenchyma [179]. Other important native lung ECM components are laminins, heparan sulfate proteoglycans, entactin, hyaluronate, chondroitin sulfate, and glycosaminoglycans [180]. In the lung, collagen subtypes I and III provide structural integrity, while collagen IV is predominantly a basement component [180,181]. Fibronectin is important for the adherence of a variety of pulmonary cell types to the ECM, and regulates cellular morphology, motility, and differentiation [182]. A wide range of matrix molecules mediate the fate of epithelial cell differentiation. For instance, AECIIs maintain their phenotype on collagen and laminin-5 substrates, but differentiate towards AECIs when cultured on collagen or laminin-5 alone [183,184]. ECM also significantly impacts stem cell differentiation. It has been reported that laminin-332 and Matrigel coatings led to increased SP-C expression by ESC-derived AECII cells [185]. For more understanding in the effects of ECM in the stem cell niche, physical stimuli and ECM-growth factor interaction in lung regeneration the following ECM-focused reviews are recommended [186,187].

6. Exogenous Stem Cell Interventions in Regenerative Therapies

6.1. Differentiation of ESCs and iPSCs into Pulmonary Epithelium

Pluripotent embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) offer enormous potential for regeneration of injured tissue and repair of disease states. ESCs are isolated from the inner cell mass of preimplantation blastocysts of an early mammalian embryo, and they can be maintained indefinitely in an undifferentiated state with the ability to differentiate into cells of all three embryonic germ layers [188]. On the other hand, iPSCs are classically generated by reprogramming fibroblasts by retroviral transduction of the transcription factors Klf4, Sox2, Oct4, and c-Myc. Once generated, iPSCs show all features of pluripotent ESCs and are capable of differentiation into cells of all three embryonic germ layers [189,190,191]. The possibility of patient-specific iPSCs offers great hope for generating genetically matched patient-specific lung progenitor cells, with the opportunity for patient specific regenerative cell therapy and drug screening as well as in vitro modelling of human diseases [87].
ESCs, in in vitro culture can perform differentiation reminiscent of the early embryo; therefore, efforts have been taken to direct differentiation of ESCs towards lung progenitors by applying signalling transduction that occurs during lung development. Definitive endoderm progenitor cells of the developing foregut give rise to the differentiated tissue of the thyroid, lung, liver, and pancreas. The lung epithelia development progresses through a primordial progenitor stage defined by the onset of Nkx2.1 expression and downregulation of Sox2 [34,40,63]. In the later stage of development, Sox2 expression increases in the area of the future lung trachea, bronchus, and bronchioles where the Nkx2.1-expressing progenitor cells of the embryonic lung giving rise to mature airway epithelium [63]. Sox9 and FoxP2 are expressed in the distal embryonic lung and mark a multipotent embryonic lung progenitor population [192,193].
Initial attempts to generate functional lung epithelium from ESCs generated mixed cell populations with the risk of teratoma formation after transplantation because of the remaining undifferentiated pluripotent stem cells [194]. To overcome this risk, Green et al. [195] found that dual inhibition of TGF-β and BMP signalling after specification of definitive endoderm (directed via Activin treatment) from ESCs highly enriches anterior foregut endoderm for the next steps in differentiation. Recently, definitive endoderm has been derived from mouse ESCs and converted first into foregut endoderm, and then into Nkx2.1+ lung endoderm using precisely-timed TGF-β, BMP-4, FGF-2, and Wnt signalling. Nkx2.1+ lung endoderm was then converted into multipotent embryonic lung progenitor and airway progenitor cells with the formation of “tracheospheres” when subcutaneously transplanted into nude mice [196]. Using the same differentiation technique, Mou et al [196] were able to generate disease-specific lung progenitor cells from human Cystic Fibrosis iPSCs, and these lung progenitors formed respiratory epithelium when subcutaneously engrafted into immunodeficient mice.
Similarly, Longmire et al. [197] purified and directed differentiation of ESCs following initial inhibition of TGF-β and BMP signalling, and then via stimulation of BMP-4 and FGF-2 signalling to form definitive endodermal precursors able to re-cellularise a 3D lung scaffold. An in vitro directed differentiation protocol has also been developed for generating functional cystic fibrosis transmembrane conductance regulator (CFTR)-expressing airway epithelia from human ESCs [198]. Carefully timed treatment by exogenous growth factors (FGF-7, FGF-10, FGF-18 and BMP-4) followed by air-liquid interface culture stimulated the differentiation of hESCs into airway epithelial cells that express functional CFTR [198]. Very recently, Huang and colleagues have derived lung progenitor cells (with ~80% efficiency) from hESC/iPSCs, which were capable of differentiating into airway and alveolar epithelial cells in vitro and in vivo [199]. Through refinement of the differentiation protocol, they first derived definitive endoderm from hESC (and iPSC) by Activin/BMP-4/bFGF treatment. This definitive endoderm was then sequentially exposed to the BMP inhibitor dorsomorphin, TGF-β inhibitor SB431542 and endogenous Wnt blocker IWP2 for shorter period, followed by a longer exposure to CHIR99021 (a small molecule that mimics Wnt signalling), FGF-10, KGF, BMP-4 and retinoic acid for derivation of anterior foregut endoderm. These lung progenitors were then differentiated into variety of pulmonary epithelial/progenitor cells including basal cells, club (Clara) cells, goblet cells, ciliated cells, AECII and AECI in vivo in immunodeficient mice and in vitro culture [199]. Tremendous efforts to better understand the details of ESCs- and iPSCs-directed differentiation in the pulmonary field are ongoing, showing great progress and therapeutic promise.

6.2. MSC-Mediated Regenerative Therapies

MSCs have been recently evaluated as a potential cell-based therapy for lung diseases. Preclinical studies and clinical trials demonstrate that the application of MSC stimulates wound repair and regeneration with efficient amelioration of a number of clinical conditions [200,201,202,203]. The precise mechanism of MSC-mediated wound repair and regeneration is not clear; however, evidence suggests that paracrine mechanisms are likely to be involved. One of the unique properties of MSCs is their site-specific migration and engraftment to injured tissues and differentiation into specific cell types. A variety of experimental animal models suggest active participation in wound repair and tissue regeneration [201,202,203]. On the other hand, some studies demonstrate that MSC-secreted paracrine factors play a vital role for wound repair, most likely through their anti-inflammatory, anti-apoptotic, angiogenic and immunomodulatory properties [204,205,206,207,208,209]. Additional reports suggest that MSC secretory products are capable of stimulating tissue-specific regional progenitor cells propagating tissue regeneration [210,211]. Recent evidence suggests that in addition to releasing soluble anti-inflammatory factors, the MSCs transfer microvesicles containing mitochondria, protein, and microRNAs to other cells [212,213]. Paracrine-modulating factors secreted by MSCs appear to be responsible for injury repair rather than MSC engraftment [213]. MSCs can be readily isolated from a variety of human tissues including bone marrow, adipose tissue, placenta, Umbilical cord and peripheral blood [214]. MSC-based regenerative therapy has been studied in pre-clinical animal models of several incurable and devastating lung diseases, with promising results are outlined below (reviewed in [214]).
Acute respiratory distress syndrome (ARDS) is a common and devastating clinical syndrome of acute lung injury (ALI) caused by various direct and indirect insults including infection, trauma, and major surgery. It can result in respiratory failure and ultimately death [215]. Several animal model studies demonstrate compelling data on the beneficial effects of MSCs in resolving acute lung injuries induced by endotoxin [216,217,218,219], hyperoxia [220], pneumonia [221] and systemic sepsis [206]. In a recent description, endotoxin-induced lung injury in explanted human lungs was ameliorated with the infusion of MSCs [206]. The accumulation of this pre-clinical data offers considerable hope that MSCs could be a potential candidate for the effective therapy of ARDS. Very recently, a Phase I clinical trial has been completed to evaluate the safety of single dose intravenous administration of allogenic bone marrow-derived MSCs in a cohort of nine moderate-to-severe ARDS patients. Single dose MSCs administration was well tolerated without any intervention-associated adverse effect or death [222]. Based on this result, a Phase II clinical trial has been started.
Evaluation of the lipopolysaccharide (LPS)-induced mouse ALI model demonstrates that intravenous or intratracheal administration of MSCs significantly attenuated LPS-induced pulmonary inflammation and alveolar injuries, improved alveolar fluid clearance, and reduced mortality [217]. This improvement of the pulmonary condition occurs in the absence of any significant engraftment of MSCs in the lung, suggesting a paracrine role of MSCs in the alleviation of ALI via suppression of pro-inflammatory cytokines tumour necrosis factor (TNF)-α and upregulation of anti-inflammatory cytokine IL-10 [217]. In support of MSC-paracrine mediated anti-inflammatory effects, Ortiz and colleagues demonstrates that MSCs and/or acellular conditioned media collected from cultured MSCs attenuates acute pulmonary inflammation. This attenuation was via suppression of both IL-1α-dependent T-lymphocyte proliferation and inhibition of TNF-α secretion by activated macrophages via MSC-secreted IL-1 receptor antagonist in vitro and in the bleomycin-induced murine lung injury model [207]. Furthermore, under inflammatory condition MSCs can be stimulated by pro-inflammatory cytokines and endotoxins, such as TNF-α and LPS. Endotoxin-mediated MSC activation occurs via toll-like receptor-4, resulting in increased production of cyclooxygenase-2 and increased prostaglandin-E2 release. MSC-secreted prostaglandin-E2 leads increased macrophage IL-10 secretion and thus can attenuate sepsis and sepsis-associated lung injury [206].
The therapeutic potential of MSCs has also been tested and shows promise in several chronic lung conditions including chronic obstructive pulmonary disease (COPD), idiopathic pulmonary fibrosis (IPF), bronchial asthma and pulmonary hypertension (PH). PH is a progressive disease with ongoing endothelial dysfunction and vascular remodelling. MSC administration has shown therapeutic benefits in murine models of PH [213]. Moreover, genetically engineered MSCs that overexpress endothelial nitric oxide synthase, prostacyclin, or heme oxygenase-1 had even greater reversal effects on PH [223,224].
IPF is a progressive disease currently without any effective therapy [225,226,227]. MSC delivery to the bleomycin injury IPF mouse model has shown low levels of MSC engraftment, but significant improvement in lung injury and fibrosis [228,229,230]. In 2007, Ortiz and colleagues showed that MSCs protects against bleomycin-induced lung injury and reduced fibrosis by blocking pro-inflammatory cytokines such as TNF-α and IL-1 by MSC-associated IL-1 receptor antagonist [207]. The administration of KGF-expressing MSCs in the bleomycin-induced mouse lung fibrosis model has shown reduced fibrosis via suppression of collagen accumulation [228]. KGF has an established role in the repair of alveolar epithelium through stimulation of AECII cell proliferation, migration and spreading [231,232,233]. Ongoing work continues to explore the possible benefits of treatment with MSCs for this lung disease with high morbidity and mortality.
COPD is the fourth leading cause of death worldwide and has been projected to be the third leading cause in 2020 [234]. No curative therapy is available for COPD at this time. COPD is characterised by an ongoing cycle of repeated destruction and repair of bronchilo-alveolar regions with subsequent tissue remodelling and sustained irreversible airway obstruction [235]. Systemic administration of bone marrow-derived MSCs was shown to ameliorate the emphysematous changes in the irradiation and papain-induced experimental mouse COPD models [208]. In this model, transplanted MSCs were localised to the emphysematous lung parenchyma and had differentiated into AECII cells. This was accompanied by reduced alveolar epithelial cell apoptosis, via Bcl-2 expression, and reduced enlargement of alveolar spaces [208]. Autologous intratracheal transplantation of bone marrow MSCs significantly mitigated elastase-induced COPD in the rabbit model [236]. The transplantation of bone marrow stem cells was associated with improved lung function, an attenuation of inflammation, an inhibition of epithelial apoptosis, a decrease in matrix metalloproteinase-2 expression, and the stimulation of alveolar and bronchiolar cell proliferation where engraftment and differentiation of the transplanted stem was negligible [236].
A recent placebo-controlled, randomised, multi-centre clinical trial has examined treatment with systemic MSCs for COPD and showed neither any adverse effects nor any differences in pulmonary function tests or quality of life indicators [237]. This clinical trial showed important safety data, and it provides a basis for further clinical trials to test the efficacy of MSC treatment in respiratory diseases. Extensive work is ongoing to investigate the role of treatment with MSCs in lung diseases in both mouse models of respiratory disease and human clinical trials.

7. Stem Cells in Lung Tissue Engineering

Research in the tissue engineering field is focused on exploration of 3-dimensional tissue culture systems for use in development of functional lung tissue. The ultimate ambition of these studies is to reduce donor-dependent lung transplantation [238,239]. The unique architecture of the lung and its anatomical and physiological complexity presents a major challenge to tissue engineers. Tissue engineered tracheas have been developed using MSCs isolated from various sources before being cultured on biodegradable and biosynthetic scaffolds to generate tracheal cartilage for the repair of congenital tracheal defects in both animal and human clinical trial models [240,241]. Very recently, a group of tissue engineers, stem cell researchers, and clinicians developed a functional human airway by culturing MSC-derived chondrocytes on an acellular tracheal scaffold, which was subsequently transplanted in a female patient who had suffered airway damage from tuberculosis [242]. A de-cellularised 7-cm long segment of human trachea was taken from a 51-year-old white female donor and then the recipient’s bone marrow-derived MSCs were differentiated into chondrocytes and airway epithelial cells were cultured in vitro. The MSC-derived chondrocytes were seeded on the external surface of the de-cellularised trachea and airway epithelial cells seeded on the luminal surface, in an equal ratio, and cultured in an air-liquid interface rotating bioreactor for 96 h. After in vitro preparation, the tracheal construct was transplanted to the left bronchus of the recipient, which improved breathing difficulties without graft rejection [242]. This achievement encourages the development of more complicated parts of the lung, such the alveoli and pulmonary vasculature, and even the entire lung.
More recently, whole-lung tissue engineering has been attempted in animal models [243,244]. These approaches make use of whole-lung de-cellularisation strategies [245] followed by reseeding the de-cellularised lung scaffold with primary airway epithelial cells, vascular endothelial cells [243,244], whole lung cell suspension digests [243], MSCs [246,247], or differentiated ESCs or iPSCs [197,248,249]. After culture within a bioreactor, the organs were re-implanted into syngeneic recipients and demonstrated efficient gas exchange [243,244]. The strategies for de-cellularising whole lungs followed by re-cellularisation with lung progenitors/stem cells have been applied to pigs, nonhuman primates [250], and human [251] lungs [252,253,254]. However, the complexity of human lung structure and difficulties in reseeding cells into all desired compartments of lung have made this task challenging and will take many years to establish and transplant a fully functional tissue-engineered whole-lung for clinical use.

8. Conclusions

Lung disease is a major cause of morbidity and mortality worldwide. Despite being highly quiescent, the lung’s ability to regenerate extensively after injury suggests that this regenerative capability could be promoted and utilised in disease condition where loss of lung tissue occurs, and the lung fails to regenerate. Endogenous stem cells are indispensable during normal tissue turnover and repair or regeneration after injury to restore the function of an organ. This seems to be true for the lung as well. However, understanding of the endogenous stem/progenitor cell population involved and the underlying mechanisms that control the proper regeneration of lung tissue remains broadly elusive. Although several animal injury models provide evidence of compartmental stem/progenitor activation and proliferation, the underlying signalling pathway that initiates this regenerative activity is largely unknown. Evidence suggests that, in many disease conditions, prenatal lung developmental signalling is reactivated. Our understanding in mammalian lung development, in particular the process of alveologenesis, is poorly understood. However, the recent advent of organotypic ex vivo models, such as “tracheosphere” and “alveolosphere” culture offer novel platforms for studying stem/progenitor dynamics and associated signalling pathways that could be involved in lung regeneration. Furthermore, derivation of lung progenitors from ESCs and iPSCs for the repair of lung after injury and disease condition opens another exciting avenue towards development of regenerative therapeutics for lung disease. In this case, however, the pluripotent nature of ESCs or iPSCs could present a potential risk of teratogenic effects, which needs to be rigorously addressed before moving into human clinical trial. Much study, so far, has been done to evaluate MSC-mediated cell therapy in various lung conditions, albeit mostly in animal models. In this case, it is important to note that the bulk of studies suggest the infused MSCs exhibits reparative/healing effects mostly through paracrine or immunomodulatory effects on recipient lung tissue, but not by engraftment. Thus, it is imperative to view MSC therapy as cell-based immunomodulatory therapy rather than as attempts to regenerate or reconstitute lung tissues [6]. Nonetheless, much effort has been taken to date to understand the molecular mechanisms of lung development, disease dynamics and its regenerative process. Much, of course, still remains to be determined. By combining the strength of basic studies along with rational clinical approaches we can fully unravel the power and promise of next-generation regenerative therapeutics in the field of lung disease in the coming years.

Acknowledgments

The authors wish to acknowledge the North Staffordshire Medical Institute and the Guy Hilton Asthma Trust for their generous support.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Beers, M.F.; Morrisey, E.E. The three R’s of lung health and disease: Repair, remodeling, and regeneration. J. Clin. Investig. 2011, 121, 2065–2073. [Google Scholar] [CrossRef] [PubMed]
  2. Wansleeben, C.; Barkauskas, C.E.; Rock, J.R.; Hogan, B.L. Stem cells of the adult lung: Their development and role in homeostasis, regeneration, and disease. Wiley Interdiscip. Rev. Dev. Biol. 2013, 2, 131–148. [Google Scholar] [CrossRef] [PubMed]
  3. Pillai, S.G.; Ge, D.; Zhu, G.; Kong, X.; Shianna, K.V.; Need, A.C.; Feng, S.; Hersh, C.P.; Bakke, P.; Gulsvik, A.; et al. ICGN Investigators. A genome-wide association study in chronic obstructive pulmonary disease (COPD): Identification of two major susceptibility loci. PLoS Genet. 2009, 5, e1000421. [Google Scholar] [CrossRef] [PubMed]
  4. Wang, B.; Zhou, H.; Yang, J.; Xiao, J.; Liang, B.; Li, D.; Zhou, H.; Zeng, Q.; Fang, C.; Rao, Z.; et al. Association of HHIP polymorphisms with COPD and COPD-related phenotypes in a Chinese Han population. Gene 2013, 531, 101–105. [Google Scholar] [CrossRef] [PubMed]
  5. Wilk, J.B.; Chen, T.H.; Gottlieb, D.J.; Walter, R.E.; Nagle, M.W.; Brandler, B.J.; Myers, R.H.; Borecki, I.B.; Silverman, E.K.; Weiss, S.T.; et al. A genome-wide association study of pulmonary function measures in the Framingham heart study. PLoS Genet. 2009, 5, e1000429. [Google Scholar] [CrossRef] [PubMed]
  6. Kotton, D.N.; Morrisey, E.E. Lung regeneration: Mechanisms, applications and emerging stem cell populations. Nat. Med. 2014, 20, 822–832. [Google Scholar] [CrossRef] [PubMed]
  7. Boyden, E.A. Development and growth of the airways. In Development of the Lung; Hodson, W.A., Ed.; Marcel Dekker: New York, NY, USA, 1977; pp. 3–35. [Google Scholar]
  8. Ten Have-Opbroek, A.A. Lung development in the mouse embryo. Exp. Lung Res. 1991, 17, 111–130. [Google Scholar] [CrossRef] [PubMed]
  9. Loosli, C.G.; Potter, E.L. Pre- and postnatal development of the respiratory portion of the human lung. Am. Rev. Respir. Dis. 1959, 80, 5–23. [Google Scholar] [PubMed]
  10. Bucher, U.; Reid, L.M. Development of intrasegmental bronchial tree: The pattern of branching and development of cartilage at various stages of intra-uterine life. Thorax 1961, 16, 207–218. [Google Scholar] [CrossRef] [PubMed]
  11. Boyden, E.A. Development of human lung. In Brenneman’s Practice of Paediatrics; Kelley, V., Ed.; Harper & Row: Hagerstown, MD, USA, 1980; pp. 1–17. [Google Scholar]
  12. Langston, C.; Kida, K.; Reed, M.; Thurlbeck, W.M. Human lung growth in late gestation and in the neonate. Am. Rev. Respir. Dis. 1984, 129, 607–613. [Google Scholar] [PubMed]
  13. Warburton, D.; El-Hashash, A.; Carraro, G.; Tiozzo, C.; Sala, F.; Rogers, O.; de Langhe, S.; Kemp, P.J.; Riccardi, D.; Torday, J.; et al. Lung organogenesis. Curr. Top. Dev. Biol. 2010, 90, 73–158. [Google Scholar] [PubMed]
  14. Alescio, T.; Cassini, A. Induction in vitro of tracheal buds by pulmonary mesenchyme grafted on tracheal epithelium. J. Exp. Zool. 1962, 150, 83–94. [Google Scholar] [CrossRef] [PubMed]
  15. Spooner, B.S.; Wessells, N.K. Mammalian lung development: Interactions in primordium formation and bronchial morphogenesis. J. Exp. Zool. 1970, 175, 445–454. [Google Scholar] [CrossRef] [PubMed]
  16. Wessells, N.K. Mammalian lung development: Interactions in formation and morphogenesis of tracheal buds. J. Exp. Zool. 1970, 175, 455–466. [Google Scholar] [CrossRef] [PubMed]
  17. Hilfer, S.R.; Rayner, R.M.; Brown, J.W. Mesenchymal control of branching pattern in the fetal mouse lung. Tissue Cell 1985, 17, 523–538. [Google Scholar] [CrossRef]
  18. Nogawa, H.; Ito, T. Branching morphogenesis of embryonic mouse lung epithelium in mesenchyme-free culture. Development 1995, 121, 1015–1022. [Google Scholar] [PubMed]
  19. Deterding, R.R.; Shannon, J.M. Proliferation and differentiation of fetal rat pulmonary epithelium in the absence of mesenchyme. J. Clin. Investig. 1995, 95, 2963–2972. [Google Scholar] [CrossRef] [PubMed]
  20. Bellusci, S.; Grindley, J.; Emoto, H.; Itoh, N.; Hogan, B.L. Fibroblast growth factor 10 (FGF10) and branching morphogenesis in the embryonic mouse lung. Development 1997, 124, 4867–4878. [Google Scholar] [PubMed]
  21. Masters, J.R.W. Epithelial-mesenchymal interaction during lung development: The effect of mesenchymal mass. Dev. Biol. 1976, 51, 98–108. [Google Scholar] [CrossRef]
  22. Shannon, J.M. Induction of alveolar type II cell differentiation in fetal tracheal epithelium by grafted distal lung mesenchyme. Dev. Biol. 1994, 166, 600–614. [Google Scholar] [CrossRef] [PubMed]
  23. Shannon, J.M.; Nielson, L.D.; Gebb, S.A.; Randell, S.H. Mesenchyme specifies epithelial differentiation in reciprocal recombinants of embryonic lung and trachea. Dev. Dyn. 1998, 212, 482–494. [Google Scholar] [CrossRef]
  24. Deutsch, G.H.; Pinar, H. Prenatal lung development. In Chronic Obstructive Lung Diseases; Voelkel, N.F., MacNee, W., Eds.; BC Decker Inc.: London, UK, 2002; Chapter 2; pp. 1–14. [Google Scholar]
  25. Ayers, M.M.; Jeffery, P.K. Proliferation and differentiation in mammalian airway epithelium. Eur. Respir. J. 1988, 1, 58–80. [Google Scholar] [PubMed]
  26. Weaver, T.E.; Whitsett, J.A. Function and regulation of expression of pulmonary surfactant-associated proteins. Biochem. J. 1991, 273, 249–264. [Google Scholar] [CrossRef] [PubMed]
  27. Wert, S.; Glasser, S.; Korfhagen, T.; Whitsett, J.A. Transcriptional elements from the human SP-C gene direct expression in the primordial respiratory epithelium of transgenic mice. Dev. Biol. 1993, 156, 426–443. [Google Scholar] [CrossRef] [PubMed]
  28. Singh, G.; Singh, J.; Katyal, S.L.; Brown, W.E.; Kramps, J.A.; Paradis, I.L.; Dauber, J.H.; Macpherson, T.A.; Squeglia, N. Identification, cellular localization, isolation, and characterization of human Clara cell-specific 10 KD protein. J. Histochem. Cytochem. 1988, 36, 73–80. [Google Scholar] [CrossRef] [PubMed]
  29. Hackett, B.P.; Shimizu, N.; Gitlin, J.D. Clara cell secretory protein gene expression in bronchiolar epithelium. Am. J. Physiol. 1992, 262, L399–L404. [Google Scholar] [PubMed]
  30. Wuenschell, C.W.; Sunday, M.E.; Singh, G.; Minoo, P.; Slavkin, H.C.; Warburton, D. Embryonic mouse lung progenitor cells co-express immunohistochemical markers of diverse mature cell lineages. J. Histochem. Cytochem. 1996, 44, 113–123. [Google Scholar] [CrossRef] [PubMed]
  31. Guazzi, S.; Lonigro, R.; Pintonello, L.; Boncinelli, E.; di Lauro, R.; Mavilio, F. The thyroid transcription factor-1 gene is a candidate target for regulation by Hox proteins. EMBO J. 1994, 13, 3339–3347. [Google Scholar] [PubMed]
  32. Minoo, P.; Hamdan, H.; Bu, D.; Warburton, D.; Stepanik, P.; deLemos, R. TTF-1 regulates lung epithelial morphogenesis. Dev. Biol. 1995, 172, 694–698. [Google Scholar] [CrossRef] [PubMed]
  33. Kimura, S.; Yoshinobu, H.; Pineau, T.; Fernandez-Salguero, P.; Fox, C.H.; Ward, J.M.; Gonzalez, F.J. The T/EBP null mouse: Thyroid-specific enhancer-binding protein is essential for the organogenesis of the thyroid, lung, ventral forebrain, and pituitary. Genes Dev. 1996, 10, 60–69. [Google Scholar] [CrossRef] [PubMed]
  34. Minoo, P.; Su, G.; Drum, H.; Bringas, P.; Kimura, S. Defects in tracheoesophageal and lung morphogenesis in Nkx2.1(-/-) mouse embryos. Dev. Biol. 1999, 209, 60–71. [Google Scholar] [CrossRef] [PubMed]
  35. Bohinski, R.J.; DiLauro, R.; Whitsett, J.A. The lung-specific surfactant protein B gene promoter is a target for thyroid transcription factor 1 and hepatocyte nuclear factor 3, indicating common factors for organ-specific gene expression along the foregut axis. Mol. Cell. Biol. 1994, 14, 5671–5681. [Google Scholar] [CrossRef] [PubMed]
  36. Bruno, M.D.; Bohinski, R.J.; Huelsman, K.M.; Whitsett, J.A.; Korfhagen, T.R. Lung cell specific expression of the murine surfactant protein A (SP-A) gene is mediated by interactions between the SPA promoter and thyroid transcription factor-1. J. Biol. Chem. 1995, 270, 6531–6536. [Google Scholar] [PubMed]
  37. Yan, C.; Sever, Z.; Whitsett, J.A. Upstream enhancer activity in the surfactant protein B gene is mediated by thyroid transcription factor 1. J. Biol. Chem. 1995, 270, 24852–24857. [Google Scholar] [CrossRef] [PubMed]
  38. Kelly, S.E.; Bachurski, C.J.; Burhans, M.S.; Glasser, S.W. Transcription of the lung-specific surfactant protein C gene is mediated by thyroid transcription factor 1. J. Biol. Chem. 1996, 271, 6881–6888. [Google Scholar] [PubMed]
  39. Zhang, L.; Whitsett, J.A.; Stripp, B.R. Regulation of Clara cell secretory protein gene transcription by thyroid transcription factor-1. Biochim. Biophys. Acta 1997, 1350, 359–367. [Google Scholar] [CrossRef]
  40. Lazzaro, D.; Price, M.; DeFelice, M.; DiLauro, R. The transcription factor TTF-1 is expressed at the onset of thyroid and lung morphogenesis and in restricted regions of the foetal brain. Development 1991, 113, 1093–1104. [Google Scholar] [PubMed]
  41. Ikeda, K.; Clark, J.C.; Shaw-White, J.R.; Stahlman, M.T.; Boutell, C.J.; Whitsett, J.A. Gene structure and expression of human thyroid transcription factor-1 in respiratory epithelial cells. J. Biol. Chem. 1995, 270, 8108–8114. [Google Scholar] [CrossRef] [PubMed]
  42. Kimura, S.; Ward, J.M.; Minoo, P. Thyroid-specific enhancerbinding protein/thyroid transcription factor 1 is not required for the initial specification of the thyroid and lung primordia. Biochimie 1999, 81, 321–327. [Google Scholar] [CrossRef]
  43. Lai, E.; Clark, K.L.; Burley, S.K.; Darnell, J.E., Jr. Hepatocyte nuclear factor 3/forkhead or “winged helix” proteins: A family of transcription factors of diverse biologic function. Proc. Natl. Acad. Sci. USA 1993, 90, 10421–10423. [Google Scholar] [CrossRef] [PubMed]
  44. Ikeda, K.; Shaw-White, J.R.; Wert, S.E.; Whitsett, J.A. Hepatocyte nuclear factor 3 activates transcription of thyroid transcription factor 1 in respiratory epithelial cells. Mol. Cell. Biol. 1996, 16, 3626–3636. [Google Scholar] [CrossRef] [PubMed]
  45. Shaw-White, J.R.; Bruno, M.D.; Whitsett, J.A. GATA-6 activates transcription of thyroid transcription factor-1. J. Biol. Chem. 1999, 274, 2658–2664. [Google Scholar] [CrossRef] [PubMed]
  46. Monaghan, A.P.; Kaestner, K.H.; Grau, E.; Schutz, G. Postimplantation expression patterns indicate a role for the mouse forkhead/HNF-3 α, β and γ genes in determination of the definitive endoderm, chordamesoderm and neuroectoderm. Development 1993, 119, 563–578. [Google Scholar]
  47. Ang, S.-L.; Wierda, A.; Wong, D.; Stevens, K.A.; Cascio, S.; Rossant, J.; Zaret, K.S. The formation and maintenance of the definitive endoderm lineage in the mouse: Involvement of HNF-3/forkhead proteins. Development 1993, 119, 1301–1315. [Google Scholar] [PubMed]
  48. Kuo, C.T.; Morrisey, E.E.; Anandappa, R.; Sigrist, K.; Lu, M.M.; Parmacek, M.S.; Soudais, C.; Leiden, J.M. GATA4 transcription factor is required for ventral morphogenesis and heart tube formation. Genes Dev. 1997, 11, 1048–1060. [Google Scholar] [CrossRef] [PubMed]
  49. Alcorn, J.L.; Gao, E.; Chen, Q.; Smith, M.E.; Gerard, R.D.; Mendelson, C.R. Genomic elements involved in transcriptional regulation of the surfactant protein-A gene. Mol. Endocrinol. 1993, 7, 1072–1085. [Google Scholar] [PubMed]
  50. Bingle, C.D.; Hackett, B.P.; Moxley, M.; Longmore, W.; Gitlin, J.D. Role of HNF-3 α and HNF-3 β in Clara cell secretory protein gene expression in bronchiolar epithelium. Biochem. J. 1995, 308, 197–202. [Google Scholar] [CrossRef] [PubMed]
  51. Ang, S.-L.; Rossant, J. HNF-3B is essential for node and notochord formation in mouse development. Cell 1994, 78, 561–74. [Google Scholar] [CrossRef]
  52. Bogue, C.W.; Lou, L.J.; Vasavada, H.; Wilson, C.M.; Jacobs, H.C. Expression of Hoxb genes in the developing mouse foregut and lung. Am. J. Respir. Cell Mol. Biol. 1996, 15, 163–171. [Google Scholar] [CrossRef] [PubMed]
  53. Kappen, C. Hox genes in the lung. Am. J. Respir. Cell Mol. Biol. 1996, 15, 156–162. [Google Scholar] [CrossRef] [PubMed]
  54. Volpe, M.V.; Martin, A.; Vosatka, R.J.; Mazzoni, C.L.; Nielsen, H.C. Hoxb-5 expression in the developing mouse lung suggests a role in branching morphogenesis and epithelial cell fate. Histochem. Cell Biol. 1997, 108, 495–504. [Google Scholar] [CrossRef] [PubMed]
  55. Bogue, C.W.; Gross, I.; Vasavada, H.; Dynia, D.W.; Wilson, C.M.; Jacobs, H.C. Identification of Hox genes in newborn lung and effects of gestational age and retinoic acid on their expression. Am. J. Physiol. 1994, 266, L448–L454. [Google Scholar] [PubMed]
  56. Chinoy, M.R.; Volpe, M.V.; Cilley, R.E.; Zgleszewski, S.E.; Vosatka, R.J.; Martin, A.; Nielsen, H.C.; Krummel, T.M. Growth factors and dexamethasone regulate Hoxb5 protein in cultured murine fetal lungs. Am. J. Physiol. 1998, 274, L610–L620. [Google Scholar] [PubMed]
  57. Cardoso, W.V.; Williams, M.C.; Mitsialis, S.A.; Joyce-Brady, M.; Rishi, A.K.; Brody, J.S. Retinoic acid induces changes in the pattern of airway branching and alters epithelial cell differentiation in the developing lung in vitro. Am. J. Respir. Cell Mol. Biol. 1995, 12, 464–476. [Google Scholar] [CrossRef] [PubMed]
  58. Bellusci, S.; Henderson, R.; Winnier, G.; Oikawa, T.; Hogan, B.L. Evidence from normal expression and targeted misexpression that bone morphogenetic protein-4 (BMP-4) plays a role in mouse embryonic lung morphogenesis. Development 1996, 122, 1693–1702. [Google Scholar] [PubMed]
  59. Weaver, M.; Yingling, Y.M.; Dunn, N.R.; Bellusci, S.; Hogan, B.L. BMP signaling regulates proximal-distal differentiation of endoderm in mouse lung development. Development 1999, 126, 4005–4015. [Google Scholar] [PubMed]
  60. Metzger, R.J.; Klein, O.D.; Martin, G.R.; Krasnow, M.A. The branching programme of mouse lung development. Nature 2008, 453, 745–750. [Google Scholar] [CrossRef] [PubMed]
  61. Tompkins, D.H.; Besnard, V.; Lange, A.W.; Keiser, A.R.; Wert, S.E.; Bruno, M.D.; Whitsett, J.A. Sox2 activates cell proliferation and differentiation in the respiratory epithelium. Am. J. Respir. Cell Mol. Biol. 2011, 45, 101–110. [Google Scholar] [CrossRef] [PubMed]
  62. Tompkins, D.H.; Besnard, V.; Lange, A.W.; Wert, S.E.; Keiser, A.R.; Smith, A.N.; Lang, R.; Whitsett, J.A. Sox2 is required for maintenance and differentiation of bronchiolar Clara, ciliated, and goblet cells. PLoS ONE 2009, 4, e8248. [Google Scholar] [CrossRef] [PubMed]
  63. Que, J.; Luo, X.; Schwartz, R.J.; Hogan, B.L. Multiple roles for Sox2 in the developing and adult mouse trachea. Development 2009, 136, 1899–1907. [Google Scholar] [CrossRef] [PubMed]
  64. Que, J.; Okubo, T.; Goldenring, J.R.; Nam, K.T.; Kurotan, I.R.; Morrisey, E.E.; Taranova, O.; Pevny, L.H.; Hogan, B.L. Multiple dose-dependent roles for Sox2 in the patterning and differentiation of anterior foregut endoderm. Development 2007, 134, 2521–2531. [Google Scholar] [CrossRef] [PubMed]
  65. Rockich, B.E.; Hrycaj, S.M.; Shih, H.P.; Nagy, M.S.; Ferguson, M.A.; Kopp, J.L.; Sander, M.; Wellik, D.M.; Spence, J.R. Sox9 plays multiple roles in the lung epithelium during branching morphogenesis. Proc. Natl. Acad. Sci. USA 2013, 110, E4456–E4464. [Google Scholar] [CrossRef] [PubMed]
  66. Chen, F.; Cao, Y.; Qian, J.; Shao, F.; Niederreither, K.; Cardoso, W.V. A retinoic acid-dependent network in the foregut controls formation of the mouse lung primordium. J. Clin. Investig. 2010, 120, 2040–2048. [Google Scholar] [CrossRef] [PubMed]
  67. De Langhe, S.P.; Carraro, G.; Tefft, D.; Li, C.; Xu, X.; Chai, Y.; Minoo, P.; Hajihosseini, M.K.; Drouin, J.; Kaartinen, V.; et al. Formation and differentiation of multiple mesenchymal lineages during lung development is regulated by β-catenin signaling. PLoS ONE 2008, 3, e1516. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Goss, A.M.; Tian, Y.; Cheng, L.; Yang, J.; Zhou, D.; Cohen, E.D.; Morrisey, E.E. Wnt2 signaling is necessary and sufficient to activate the airway smooth muscle program in the lung by regulating myocardin/Mrtf-B and Fgf10 expression. Dev. Biol. 2011, 356, 541–552. [Google Scholar] [CrossRef] [PubMed]
  69. Nyeng, P.; Norgaard, G.A.; Kobberup, S.; Jensen, J. FGF10 maintains distal lung bud epithelium and excessive signaling leads to progenitor state arrest, distalization, and goblet cell metaplasia. BMC Dev. Biol. 2008, 8. [Google Scholar] [CrossRef] [PubMed]
  70. Ramasamy, S.K.; Mailleux, A.A.; Gupte, V.V.; Mata, F.; Sala, F.G.; Veltmaat, J.M.; del Moral, P.M.; de Langhe, S.; Parsa, S.; Kelly, L.K.; et al. Fgf10 dosage is critical for the amplification of epithelial cell progenitors and for the formation of multiple mesenchymal lineages during lung development. Dev. Biol. 2007, 307, 237–247. [Google Scholar] [CrossRef] [PubMed]
  71. Rawlins, E.L.; Clark, C.P.; Xue, Y.; Hogan, B.L. The Id2+ distal tip lung epithelium contains individual multipotent embryonic progenitor cells. Development 2009, 136, 3741–3745. [Google Scholar] [CrossRef] [PubMed]
  72. Volckaert, T.; Campbell, A.; Dill, E.; Li, C.; Minoo, P.; de Langhe, S. Localized Fgf10 expression is not required for lung branching morphogenesis but prevents differentiation of epithelial progenitors. Development 2013, 140, 3731–3742. [Google Scholar] [CrossRef] [PubMed]
  73. Izvolsky, K.I.; Shoykhet, D.; Yang, Y.; Yu, Q.; Nugent, M.A.; Cardoso, W.V. Heparan sulfate-FGF10 interactions during lung morphogenesis. Dev. Biol. 2003, 258, 185–200. [Google Scholar] [CrossRef]
  74. Izvolsky, K.I.; Zhong, L.; Wei, L.; Yu, Q.; Nugent, M.A.; Cardoso, W.V. Heparan sulfates expressed in the distal lung are required for Fgf10 binding to the epithelium and for airway branching. Am. J. Physiol. Lung Cell. Mol. Physiol. 2003, 285, L838–L846. [Google Scholar] [CrossRef] [PubMed]
  75. Sedita, J.; Izvolsky, K.; Cardoso, W.V. Differential expression of heparan sulfate 6-O-sulfotransferase isoforms in the mouse embryo suggests distinctive roles during organogenesis. Dev. Dyn. 2004, 231, 782–794. [Google Scholar] [CrossRef] [PubMed]
  76. McKeehan, W.L.; Wang, F.; Kan, M. The heparan sulfate-fibroblast growth factor family: Diversity of structure and function. Prog. Nucleic Acid Res. Mol. Biol. 1998, 59, 135–176. [Google Scholar] [PubMed]
  77. Chang, D.R.; Alanis, D.M.; Miller, R.K.; Ji, H.; Akiyama, H.; McCrea, P.D.; Chen, J. Lung epithelial branching program antagonizes alveolar differentiation. Proc. Natl. Acad. Sci. USA 2013, 110, 18042–18051. [Google Scholar] [CrossRef] [PubMed]
  78. Volckaert, T.; de Langhe, S. Lung epithelial stem cells and their niches: Fgf10 takes center stage. Fibrogenesis Tissue Repair. 2014, 8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Wang, Y.; Tian, Y.; Morley, M.P.; Lu, M.M.; Demayo, F.J.; Olson, E.N.; Morrisey, E.E. Development and regeneration of Sox2+ endoderm progenitors are regulated by a Hdac1/2-Bmp4/Rb1 regulatory pathway. Dev. Cell 2013, 24, 345–358. [Google Scholar] [CrossRef] [PubMed]
  80. Shiratori, M.; Oshika, E.; Ung, L.P.; Singh, G.; Shinozuka, H.; Warburton, D.; Michalopoulos, G.; Katyal, S.L. Keratinocyte growth factor and lung morphogenesis. Am. J. Respir. Cell Mol. Biol. 1996, 15, 328–338. [Google Scholar] [CrossRef] [PubMed]
  81. Cardoso, W.V.; Itoh, A.; Nogawa, H.; Mason, I.; Brody, J.S. FGF-1 and FGF-7 induce distinct patterns of growth and differentiation in embryonic lung epithelium. Dev. Dyn. 1997, 8, 398–405. [Google Scholar] [CrossRef]
  82. Shannon, J.M.; Gebb, S.A.; Nielson, L.D. Induction of alveolar type II cell differentiation in embryonic tracheal epithelium in mesenchyme-free culture. Development 1999, 126, 1675–1688. [Google Scholar] [PubMed]
  83. Weinstein, M.; Xu, X.; Ohyama, K.; Deng, C.-X. FGFR-3 and FGFR-4 function cooperatively to direct alveogenesis in the murine lung. Development 1998, 125, 3615–3623. [Google Scholar] [PubMed]
  84. Plopper, C.G.; st George, J.A.; Read, L.C.; Nishio, S.J.; Weir, A.J.; Edwards, L.; Tarantal, A.F.; Pinkerton, K.E.; Merritt, T.A.; Whitsett, J.A.; et al. Acceleration of type II cell differentiation in fetal rhesus monkey lung by administration of EGF. Am. J. Physiol. 1992, 262, L313–L321. [Google Scholar] [PubMed]
  85. Kolios, G.; Moodley, Y. Introduction to stem cells and regenerative medicine. Respiration 2013, 85, 3–10. [Google Scholar] [CrossRef] [PubMed]
  86. Potten, C.; Loeffler, M. Stem cells: Attributes, cycles, spirals, pitfalls and uncertainties. Lessons for and from the crypt. Development 1990, 110, 1001–1020. [Google Scholar] [PubMed]
  87. Leeman, K.T.; Fillmore, C.M.; Kim, C.F. Lung stem and progenitor cells in tissue homeostasis and disease. Curr. Top. Dev. Biol. 2014, 107, 207–233. [Google Scholar] [PubMed]
  88. Adamson, I.Y.; Bowden, D.H. The type 2 cell as progenitor of alveolar epithelial regeneration. A cytodynamic study in mice after exposure to oxygen. Lab. Investig. 1974, 30, 35–42. [Google Scholar] [PubMed]
  89. Adamson, I.Y.; Bowden, D.H. Origin of ciliated alveolar epithelial cells in bleomycin-induced lung injury. Am. J. Pathol. 1977, 87, 569–580. [Google Scholar] [PubMed]
  90. Adamson, I.Y.; Bowden, D.H. Bleomycin-induced injury and metaplasia of alveolar type 2 cells. Relationship of cellular responses to drug presence in the lung. Am. J. Pathol. 1979, 96, 531–544. [Google Scholar] [PubMed]
  91. Bowden, D.H.; Adamson, I.Y.; Wyatt, J.P. Reaction of the lung cells to a high concentration of oxygen. Arch. Pathol. 1968, 86, 671–675. [Google Scholar] [PubMed]
  92. Evans, M.J.; Cabral, L.C.; Stephens, R.J.; Freeman, G. Acute kinetic response and renewal of the alveolar epithelium following injury by nitrogen dioxide. Chest 1974, 65, 62S–65S. [Google Scholar] [CrossRef] [PubMed]
  93. Evans, M.J.; Cabral, L.J.; Stephens, R.J.; Freeman, G. Renewal of alveolar epithelium in the rat following exposure to NO2. Am. J. Pathol. 1973, 70, 175–198. [Google Scholar] [PubMed]
  94. Evans, M.J.; Cabral, L.J.; Stephens, R.J.; Freeman, G. Transformation of alveolar type 2 cells to type 1 cells following exposure to NO2. Exp. Mol. Pathol. 1975, 22, 142–150. [Google Scholar] [CrossRef]
  95. Evans, M.J.; Dekker, N.P.; Cabral-Anderson, L.J.; Freeman, G. Quantitation of damage to the alveolar epithelium by means of type 2 cell proliferation. Am. Rev. Respir. Dis. 1978, 118, 787–790. [Google Scholar] [CrossRef] [PubMed]
  96. Rawlins, E.L.; Hogan, B.L. Epithelial stem cells of the lung: Privileged few or opportunities for many? Development 2006, 133, 2455–2465. [Google Scholar] [CrossRef] [PubMed]
  97. Rawlins, E.L.; Ostrowski, L.E.; Randell, S.H.; Hogan, B.L. Lung development and repair: Contribution of the ciliated lineage. Proc. Natl. Acad. Sci. USA 2007, 104, 410–417. [Google Scholar] [CrossRef] [PubMed]
  98. Rawlins, E.L.; Hogan, B.L. Ciliated epithelial cell lifespan in the mouse trachea and lung. Am. J. Physiol. Lung Cell. Mol. Physiol. 2008, 295, L231–L234. [Google Scholar] [CrossRef] [PubMed]
  99. Rawlins, E.L.; Okubo, T.; Que, J.; Xue, Y.; Clark, C.; Luo, X.; Hogan, B.L. Epithelial stem/progenitor cells in lung postnatal growth, maintenance, and repair. Cold Spring Harb. Symp. Quant. Biol. 2008, 73, 291–295. [Google Scholar] [CrossRef] [PubMed]
  100. Borthwick, D.W.; Shahbazian, M.; Krantz, Q.T.; Dorin, J.R.; Randell, S.H. Evidence for stem-cell niches in the tracheal epithelium. Am. J. Respir. Cell Mol. Biol. 2001, 24, 662–670. [Google Scholar] [CrossRef] [PubMed]
  101. Rock, J.; Randell, S.; Hogan, B. Airway basal stem cells: A perspective on their roles in epithelial homeostasis and remodeling. Dis. Models Mech. 2010, 3, 545–556. [Google Scholar] [CrossRef] [PubMed]
  102. Rock, J.R.; Onaitis, M.W.; Rawlins, E.L.; Lu, Y.; Clark, C.P.; Xue, Y.; Randell, S.H.; Hogan, B.L. Basal cells as stem cells of the mouse trachea and human airway epithelium. Proc. Natl. Acad. Sci. USA 2009, 106, 12771–12775. [Google Scholar] [CrossRef] [PubMed]
  103. Cole, B.B.; Smith, R.W.; Jenkins, K.M.; Graham, B.B.; Reynolds, P.R.; Reynolds, S.D. Tracheal basal cells: A facultative progenitor cell pool. Am. J. Pathol. 2010, 177, 362–376. [Google Scholar] [CrossRef] [PubMed]
  104. Hong, K.U.; Reynolds, S.D.; Watkins, S.; Fuchs, E.; Stripp, B.R. In vivo differentiation potential of tracheal basal cells: Evidence for multipotent and unipotent subpopulations. Am. J. Physiol. Lung Cell. Mol. Physiol. 2004, 286, L643–L649. [Google Scholar] [CrossRef] [PubMed]
  105. Kumar, P.A.; Hu, Y.; Yamamoto, Y.; Hoe, N.B.; Wei, T.S.; Mu, D.; Hoe, N.B.; Wei, T.S.; Mu, D.; Sun, Y.; et al. Distal airway stem cells yield alveoli in vitro and during lung regeneration following H1N1 influenza infection. Cell 2011, 147, 525–538. [Google Scholar] [CrossRef] [PubMed]
  106. Pardo-Saganta, A.; Law, B.M.; Gonzalez-Celeiro, M.; Vinarsky, V.; Rajagopal, J. Ciliated cells of pseudostratified airway epithelium do not become mucous cells after ovalbumin challenge. Am. J. Respir. Cell Mol. Biol. 2013, 48, 364–373. [Google Scholar] [CrossRef] [PubMed]
  107. Rock, J.R.; Gao, X.; Xue, Y.; Randell, S.H.; Kong, Y.Y.; Hogan, B.L. Notch-dependent differentiation of adult airway basal stem cells. Cell Stem Cell 2011, 8, 639–648. [Google Scholar] [CrossRef] [PubMed]
  108. Reynolds, S.D.; Reynolds, P.R.; Pryhuber, G.S.; Finder, J.D.; Stripp, B.R. Secretoglobins SCGB3A1 and SCGB3A2 define secretory cell subsets in mouse and human airways. Am. J. Respir. Crit. Care Med. 2002, 166, 1498–1509. [Google Scholar] [CrossRef] [PubMed]
  109. Song, H.; Yao, E.; Lin, C.; Gacayan, R.; Chen, M.-H.; Chuang, P.-T. Functional characterization of pulmonary neuroendocrine cells in lung development, injury, and tumorigenesis. Proc. Natl. Acad. Sci. USA 2012, 109, 17531–17536. [Google Scholar] [CrossRef] [PubMed]
  110. Giangreco, A.; Reynolds, S.D.; Stripp, B.R. Terminal bronchioles harbor a unique airway stem cell population that localizes to the bronchoalveolar duct junction. Am. J. Pathol. 2002, 161, 173–182. [Google Scholar] [CrossRef]
  111. Rawlins, E.L.; Okubo, T.; Xue, Y.; Brass, D.M.; Auten, R.L.; Hasegawa, H.; Wang, F.; Hogan, B.L. The role of Scgb1a1+ Clara cells in the long-term maintenance and repair of lung airway, but not alveolar, epithelium. Cell Stem Cell 2009, 4, 525–534. [Google Scholar] [CrossRef] [PubMed]
  112. Hong, K.U.; Reynolds, S.D.; Giangreco, A.; Hurley, C.M.; Stripp, B.R. Clara cell secretory protein-expressing cells of the airway neuroepithelial body microenvironment include a label-retaining subset and are critical for epithelial renewal after progenitor cell depletion. Am. J. Respir. Cell Mol. Biol. 2001, 24, 671–681. [Google Scholar] [CrossRef] [PubMed]
  113. Guha, A.; Vasconcelos, M.; Cai, Y.; Yoneda, M.; Hinds, A.; Qian, J.; Li, G.; Dickel, L.; Johnson, J.E.; Kimura, S.; et al. Neuroepithelial body microenvironment is a niche for a distinct subset of Clara-like precursors in the developing airways. Proc. Natl. Acad. Sci. USA 2012, 109, 12592–12597. [Google Scholar] [CrossRef] [PubMed]
  114. Morimoto, M.; Nishinakamura, R.; Saga, Y.; Kopan, R. Different assemblies of Notch receptors coordinate the distribution of the major bronchial Clara, ciliated and neuroendocrine cells. Development 2012, 139, 4365–4373. [Google Scholar] [CrossRef] [PubMed]
  115. Volckaert, T.; Dill, E.; Campbell, A.; Tiozzo, C.; Majka, S.; Bellusci, S.; de Langhe, S.P. Parabronchial smooth muscle constitutes an airway epithelial stem cell niche in the mouse lung after injury. J. Clin. Investig. 2011, 121, 4409–4419. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Giangreco, A.; Arwert, E.N.; Rosewell, I.R.; Snyder, J.; Watt, F.M.; Stripp, B.R. Stem cells are dispensable for lung homeostasis but restore airways after injury. Proc. Natl. Acad. Sci. USA 2009, 106, 9286–9291. [Google Scholar] [CrossRef] [PubMed]
  117. Bertalanffy, F.D.; Leblond, C.P. Structure of respiratory tissue. Lancet 1955, 266, 1365–1368. [Google Scholar] [CrossRef]
  118. Mason, R.J.; Williams, M.C. Type II alveolar cell. Defender of the alveolus. Am. Rev. Respir. Dis. 1977, 115, 81–91. [Google Scholar] [PubMed]
  119. Kauffman, S.L. Alterations in cell proliferation in mouse lung following urethane exposure. 3. Effects of chronic exposure on type 2 alveolar epithelial cell. Am. J. Pathol. 1972, 68, 317–326. [Google Scholar] [PubMed]
  120. Kauffman, S.L. Kinetics of alveolar epithelial hyperplasia in lungs of mice exposed to urethane. I. quantitative analysis of cell populations. Lab. Investig. 1974, 30, 170–175. [Google Scholar] [PubMed]
  121. Kalina, M.; Riklis, S.; Blau, H. Pulmonary epithelial cell proliferation in primary culture of alveolar type II cells. Exp. Lung Res. 1993, 19, 153–175. [Google Scholar] [CrossRef] [PubMed]
  122. Witschi, H. Proliferation of type II alveolar cells: A review of common responses in toxic lung injury. Toxicology 1976, 5, 267–277. [Google Scholar] [CrossRef]
  123. Bitterman, P.B.; Polunovsky, V.A.; Ingbar, D.H. Repair after acute lung injury. Chest 1994, 105, 118S–121S. [Google Scholar] [CrossRef] [PubMed]
  124. Barkauskas, C.E.; Cronce, M.J.; Rackley, C.R.; Bowie, E.J.; Keene, D.R.; Stripp, B.R.; Randell, S.H.; Noble, P.W.; Hogan, B.L. Type 2 alveolar cells are stem cells in adult lung. J. Clin. Investig. 2013, 123, 3025–3036. [Google Scholar] [CrossRef] [PubMed]
  125. Desai, T.J.; Brownfield, D.G.; Krasnow, M.A. Alveolar progenitor and stem cells in lung development, renewal and cancer. Nature 2014, 507, 190–194. [Google Scholar] [CrossRef] [PubMed]
  126. Kim, C.F.; Jackson, E.L.; Woolfenden, A.E.; Lawrence, S.; Babar, I.; Vogel, S.; Crowley, D.; Bronson, R.T.; Jacks, T. Identification of bronchioalveolar stem cells in normal lung and lung cancer. Cell 2005, 121, 823–835. [Google Scholar] [CrossRef] [PubMed]
  127. Rock, J.R.; Barkauskas, C.E.; Cronce, M.J.; Xue, Y.; Harris, J.R.; Liang, J.; Noble, P.W.; Hogan, B.L. Multiple stromal populations contribute to pulmonary fibrosis without evidence for epithelial to mesenchymal transition. Proc. Natl. Acad. Sci. USA 2011, 108, E1475–E1483. [Google Scholar] [CrossRef] [PubMed]
  128. Zheng, D.; Limmon, G.V.; Yin, L.; Leung, N.H.; Yu, H.; Chow, V.T.; Chen, J. Regeneration of alveolar type I and II cells from Scgb1a1-expressing cells following severe pulmonary damage induced by bleomycin and influenza. PLoS ONE 2012, 7, e48451. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Da Silva Meirelles, L.; Chagastelles, P.C.; Nardi, N.B. Mesenchymal stem cells reside in virtually all post-natal organs and tissues. J. Cell Sci. 2006, 119, 2204–2213. [Google Scholar] [CrossRef] [PubMed]
  130. McQualter, J.L.; Brouard, N.; Williams, B.N.; Baird, B.N.; Sims-Lucas, S.; Yuen, K.; Nilsson, S.K.; Simmons, P.J.; Bertoncello, I. Endogenous fibroblastic progenitor cells in the adult mouse lung are highly enriched in the SCA-1 positive cell fraction. Stem Cells 2009, 27, 623–633. [Google Scholar] [CrossRef] [PubMed]
  131. Summer, R.; Fitzsimmons, K.; Dwyer, D.; Murphy, J.; Fine, A. Isolation of an adult mouse lung mesenchymal progenitor cell population. Am. J. Respir. Cell Mol. Biol. 2007, 37, 152–159. [Google Scholar] [CrossRef] [PubMed]
  132. Hegab, A.E.; Kubo, H.; Fujino, N.; Suzuki, T.; He, M.; Kato, H.; Yamaya, M. Isolation and characterization of murine multipotent lung stem cells. Stem Cells Dev. 2010, 19, 523–536. [Google Scholar] [CrossRef] [PubMed]
  133. Reynolds, S.D.; Shen, H.; Reynolds, P.R.; Betsuyaku, T.; Pilewski, J.M.; Gambelli, F.; di Giuseppe, M.; Ortiz, L.A.; Stripp, B.R. Molecular and functional properties of lung SP cells. Am. J. Physiol. Lung Cell. Mol. Physiol. 2007, 292, L972–L983. [Google Scholar] [CrossRef] [PubMed]
  134. Martin, J.; Helm, K.; Ruegg, P.; Varella-Garcia, M.; Burnham, E.; Majka, S. Adult lung side population cells have mesenchymal stem cell potential. Cytotherapy 2008, 10, 140–151. [Google Scholar] [CrossRef] [PubMed]
  135. Jarvinen, L.; Badri, L.; Wettlaufer, S.; Ohtsuka, T.; Standiford, T.J.; Toews, G.B.; Pinsky, D.J.; Peters-Golden, M.; Lama, V.N. Lung resident mesenchymal stem cells isolated from human lung allografts inhibit T cell proliferation via a soluble mediator. J. Immunol. 2008, 181, 4389–4396. [Google Scholar] [CrossRef] [PubMed]
  136. Lama, V.N.; Smith, L.; Badri, L.; Flint, A.; Andrei, A.C.; Murray, S.; Wang, Z.; Liao, H.; Toews, G.B.; Krebsbach, P.H.; et al. Evidence for tissueresident mesenchymal stem cells in human adult lung from studies of transplanted allografts. J. Clin. Investig. 2007, 117, 989–996. [Google Scholar] [CrossRef] [PubMed]
  137. Hennrick, K.T.; Keeton, A.G.; Nanua, S.; Kijek, T.G.; Goldsmith, A.M.; Sajjan, U.S.; Bentley, J.K.; Lama, V.N.; Moore, B.B.; Schumacher, R.E.; et al. Lung cells from neonates show a mesenchymal stem cell phenotype. Am. J. Respir. Crit. Care Med. 2007, 175, 1158–1164. [Google Scholar] [CrossRef] [PubMed]
  138. Ingenito, E.P.; Sen, E.; Tsai, L.W. Design and testing of biological scaffolds for delivering reparative cells to target sites in the lung. J. Tissue Eng. Regen. Med. 2010, 4, 259–272. [Google Scholar] [CrossRef] [PubMed]
  139. Ingenito, E.P.; Tsai, L.; Murthy, S. Autologous lung-derived mesenchymal stem cell transplantation in experimental emphysema. Cell Transplant. 2011, 21, 175–189. [Google Scholar] [CrossRef] [PubMed]
  140. Dominici, M.; le Blanc, K.; Mueller, I.; Slaper-Cortenbach, I.; Marini, F.; Krause, D.; Deans, R.; Keating, A.; Prockop, D.; Horwitz, E. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 2006, 8, 315–317. [Google Scholar] [CrossRef] [PubMed]
  141. Karoubi, G.; Cortes-Dericks, L.; Breyer, I.; Schmid, R.; Dutly, A. Identification of mesenchymal stromal cells in human lung parenchyma capable of differentiating into aquaporin 5-expressing cells. Lab. Investig. 2009, 89, 1100–1114. [Google Scholar] [CrossRef] [PubMed]
  142. Hegab, A.; Ha, V.; Gilbert, J.; Zhang, K.X.; Malkoski, S.P.; Chon, A.T.; Darmawan, D.O.; Bisht, B.; Ooi, A.T.; Pellegrini, M.; et al. Novel stem/progenitor cell population from murine tracheal submucosal gland ducts with multipotent regenerative potential. Stem Cells 2011, 29, 1283–1293. [Google Scholar] [CrossRef] [PubMed]
  143. Morrisey, E.; Hogan, B. Preparing for the first breath: Genetic and cellular mechanisms in lung development. Dev. Cell 2010, 18, 8–23. [Google Scholar] [CrossRef] [PubMed]
  144. McQualter, J.; Yuen, K.; Williams, B.; Bertoncello, I. Evidence of an epithelial stem/progenitor cell hierarchy in the adult mouse lung. Proc. Natl. Acad. Sci. USA 2010, 107, 1414–1419. [Google Scholar] [CrossRef] [PubMed]
  145. Hoffman, A.; Paxson, J.; Mazan, M.; Davis, A.M.; Tyagi, S.; Murthy, S.; Ingenito, E.P. Lung-derived mesenchymal stromal cell post-transplantation survival, persistence, paracrine expression, and repair of elastase injured lung. Stem Cells Dev. 2011, 20, 1779–1792. [Google Scholar] [CrossRef] [PubMed]
  146. Hogan, B.L.; Barkauskas, C.E.; Chapman, H.A.; Epstein, J.A.; Jain, R.; Hsia, C.C.; Niklason, L.; Calle, E.; Le, A.; Randell, S.H.; et al. Repair and regeneration of the respiratory system: Complexity, plasticity, and mechanisms of lung stem cell function. Cell Stem Cell 2014, 15, 123–138. [Google Scholar] [CrossRef] [PubMed]
  147. Dane, D.M.; Yilmaz, C.; Estrera, A.S.; Hsia, C.C. Separating in vivo mechanical stimuli for postpneumonectomy compensation: Physiological assessment. J. Appl. Physiol. 2013, 114, 99–106. [Google Scholar] [CrossRef] [PubMed]
  148. Ravikumar, P.; Yilmaz, C.; Bellotto, D.J.; Dane, D.M.; Estrera, A.S.; Hsia, C.C. Separating in vivo mechanical stimuli for postpneumonectomy compensation: Imaging and ultrastructural assessment. J. Appl. Physiol. 2013, 114, 961–970. [Google Scholar] [CrossRef] [PubMed]
  149. Ravikumar, P.; Yilmaz, C.; Dane, D.M.; Bellotto, D.J.; Estrera, A.S.; Hsia, C.C. Defining a stimuli-response relationship in compensatory lung growth following major resection. J. Appl. Physiol. 2014, 116, 816–824. [Google Scholar] [CrossRef] [PubMed]
  150. Chen, L.; Acciani, T.; le Cras, T.; Lutzko, C.; Perl, A.K. Dynamic regulation of platelet-derived growth factor receptor a expression in alveolar fibroblasts during realveolarization. Am. J. Respir. Cell Mol. Biol. 2012, 47, 517–527. [Google Scholar] [CrossRef] [PubMed]
  151. Eisenhauer, P.; Earle, B.; Loi, R.; Sueblinvong, V.; Goodwin, M.; Allen, G.B.; Lundblad, L.; Mazan, M.R.; Hoffman, A.M.; Weiss, D.J. Endogenous distal airway progenitor cells, lung mechanics, and disproportionate lobar growth following long-term postpneumonectomy in mice. Stem Cells 2013, 31, 1330–1339. [Google Scholar] [CrossRef] [PubMed]
  152. Hoffman, A.M.; Shifren, A.; Mazan, M.R.; Gruntman, A.M.; Lascola, K.M.; Nolen-Walston, R.D.; Kim, C.F.; Tsai, L.; Pierce, R.A.; Mecham, R.P.; et al. Matrix modulation of compensatory lung regrowth and progenitor cell proliferation in mice. Am. J. Physiol. Lung Cell. Mol. Physiol. 2010, 298, L158–L168. [Google Scholar] [CrossRef] [PubMed]
  153. Hsia, C.C. Signals and mechanisms of compensatory lung growth. J. Appl. Physiol. 2004, 97, 1992–1998. [Google Scholar] [CrossRef] [PubMed]
  154. Thane, K.; Ingenito, E.P.; Hoffman, A.M. Lung regeneration and translational implications of the postpneumonectomy model. Transl. Res. 2014, 163, 363–376. [Google Scholar] [CrossRef] [PubMed]
  155. Voswinckel, R.; Motejl, V.; Fehrenbach, A.; Wegmann, M.; Mehling, T.; Fehrenbach, H.; Seeger, W. Characterisation of post-pneumonectomy lung growth in adult mice. Eur. Respir. J. 2004, 24, 524–532. [Google Scholar] [CrossRef] [PubMed]
  156. Kaza, A.K.; Laubach, V.E.; Kern, J.A.; Long, S.M.; Fiser, S.M.; Tepper, J.A.; Nguyen, R.P.; Shockey, K.S.; Tribble, C.G.; Kron, I.L. Epidermal growth factor augments postpneumonectomy lung growth. J. Thorac. Cardiovasc. Surg. 2000, 120, 916–921. [Google Scholar] [CrossRef] [PubMed]
  157. Kaza, A.K.; Kron, I.L.; Leuwerke, S.M.; Tribble, C.G.; Laubach, V.E. Keratinocyte growth factor enhances post-pneumonectomy lung growth by alveolar proliferation. Circulation 2002, 106, I120–I124. [Google Scholar] [PubMed]
  158. Al Alam, D.; Green, M.; Tabatabai Irani, R.; Parsa, S.; Danopoulos, S.; Sala, F.G.; Branch, J.; El Agha, E.; Tiozzo, C.; Voswinckel, R.; et al. Contrasting expression of canonical Wnt signaling reporters TOPGAL, BATGAL and Axin2(LacZ) during murine lung development and repair. PLoS ONE 2011, 6, e23139. [Google Scholar] [CrossRef] [PubMed]
  159. Aumiller, V.; Balsara, N.; Wilhelm, J.; Günther, A.; Königshoff, M. WNT/β-catenin signaling induces IL-1B expression by alveolar epithelial cells in pulmonary fibrosis. Am. J. Respir. Cell Mol. Biol. 2013, 49, 96–104. [Google Scholar] [CrossRef] [PubMed]
  160. Flozak, A.S.; Lam, A.P.; Russell, S.; Jain, M.; Peled, O.N.; Sheppard, K.A.; Beri, R.; Mutlu, G.M.; Budinger, G.R.; Gottardi, C.J. β-Catenin/T-cell factor signaling is activated during lung injury and promotes the survival and migration of alveolar epithelial cells. J. Biol. Chem. 2010, 285, 3157–3167. [Google Scholar] [CrossRef] [PubMed]
  161. Hashimoto, S.; Chen, H.; Que, J.; Brockway, B.L.; Drake, J.A.; Snyder, J.C.; Randell, S.H.; Stripp, B.R. β-Catenin-SOX2 signaling regulates the fate of developing airway epithelium. J. Cell Sci. 2012, 125, 932–942. [Google Scholar] [CrossRef] [PubMed]
  162. Zhang, Y.; Goss, A.M.; Cohen, E.D.; Kadzik, R.; Lepore, J.J.; Muthukumaraswamy, K.; Yang, J.; DeMayo, F.J.; Whitsett, J.A.; Parmacek, M.S.; et al. A GATA6-Wnt pathway required for epithelial stem cell development and airway regeneration. Nat. Genet. 2008, 40, 862–870. [Google Scholar] [CrossRef] [PubMed]
  163. Zemke, A.C.; Teisanu, R.M.; Giangreco, A.; Drake, J.A.; Brockway, B.L.; Reynolds, S.D.; Stripp, B.R. β-Catenin is not necessary for maintenance or repair of the bronchiolar epithelium. Am. J. Respir. Cell Mol. Biol. 2009, 41, 535–543. [Google Scholar] [CrossRef] [PubMed]
  164. Tanjore, H.; Degryse, A.L.; Crossno, P.F.; Xu, X.C.; McConaha, M.E.; Jones, B.R.; Polosukhin, V.V.; Bryant, A.J.; Cheng, D.S.; Newcomb, D.C.; et al. β-Catenin in the alveolar epithelium protects from lung fibrosis after intratracheal bleomycin. Am. J. Respir. Crit. Care Med. 2013, 187, 630–639. [Google Scholar] [CrossRef] [PubMed]
  165. Henderson, W.R., Jr.; Chi, E.Y.; Ye, X.; Tien, Y.T.; Zhou, B.; Borok, Z.; Knight, D.A.; Kahn, M. Inhibition of Wnt/β-catenin/CREB binding protein (CBP) signaling reverses pulmonary fibrosis. Proc. Natl. Acad. Sci. USA 2010, 107, 14309–14314. [Google Scholar] [CrossRef] [PubMed]
  166. Chilosi, M.; Poletti, V.; Zamo, A.; Lestani, M.; Montagna, L.; Piccoli, P.; Pedron, S.; Bertaso, M.; Scarpa, A.; Murer, B.; et al. Aberrant Wnt/β-catenin pathway activation in idiopathic pulmonary fibrosis. Am. J. Pathol. 2003, 162, 1495–1502. [Google Scholar] [CrossRef]
  167. Morimoto, M.; Liu, Z.; Cheng, H.T.; Winters, N.; Bader, D.; Kopan, R. Canonical Notch signaling in the developing lung is required for determination of arterial smooth muscle cells and selection of Clara vs. ciliated cell fate. J. Cell Sci. 2010, 123, 213–224. [Google Scholar] [CrossRef] [PubMed]
  168. Tsao, P.N.; Vasconcelos, M.; Izvolsky, K.I.; Qian, J.; Lu, J.; Cardoso, W.V. Notch signaling controls the balance of ciliated and secretory cell fates in developing airways. Development 2009, 136, 2297–2307. [Google Scholar] [CrossRef] [PubMed]
  169. Xing, Y.; Li, A.; Borok, Z.; Li, C.; Minoo, P. NOTCH1 is required for regeneration of Clara cells during repair of airway injury. Stem Cells 2012, 30, 946–955. [Google Scholar] [CrossRef] [PubMed]
  170. Paul, M.K.; Bisht, B.; Darmawan, D.O.; Chiou, R.; Ha, V.L.; Wallace, W.D.; Chon, A.T.; Hegab, A.E.; Grogan, T.; Elashoff, D.A.; et al. Dynamic changes in intracellular ROS levels regulate airway basal stem cell homeostasis through Nrf2-dependent Notch signaling. Cell Stem Cell 2014, 15, 199–214. [Google Scholar] [CrossRef] [PubMed]
  171. Yin, Z.; Gonzales, L.; Kolla, V.; Rath, N.; Zhang, Y.; Lu, M.M.; Kimura, S.; Ballard, P.L.; Beers, M.F.; Epstein, J.A.; et al. Hop functions downstream of Nkx2.1 and GATA6 to mediate HDAC-dependent negative regulation of pulmonary gene expression. Am. J. Physiol. Lung Cell. Mol. Physiol. 2006, 291, L191–L199. [Google Scholar] [CrossRef] [PubMed]
  172. Ito, K.; Ito, M.; Elliott, W.M.; Cosio, B.; Caramori, G.; Kon, O.M.; Barczyk, A.; Hayashi, S.; Adcock, I.M.; Hogg, J.C.; et al. Decreased histone deacetylase activity in chronic obstructive pulmonary disease. N. Engl. J. Med. 2005, 352, 1967–1976. [Google Scholar] [CrossRef] [PubMed]
  173. Gunawardhana, L.P.; Gibson, P.G.; Simpson, J.L.; Powell, H.; Baines, K.J. Activity and expression of histone acetylases and deacetylases in inflammatory phenotypes of asthma. Clin. Exp. Allergy 2014, 44, 47–57. [Google Scholar] [CrossRef] [PubMed]
  174. Tian, Y.; Zhang, Y.; Hurd, L.; Hannenhalli, S.; Liu, F.; Lu, M.M.; Morrisey, E.E. Regulation of lung endoderm progenitor cell behavior by miR302/367. Development 2011, 138, 1235–1245. [Google Scholar] [CrossRef] [PubMed]
  175. Ventura, A.; Young, A.G.; Winslow, M.M.; Lintault, L.; Meissner, A.; Erkeland, S.J.; Newman, J.; Bronson, R.T.; Crowley, D.; Stone, J.R.; et al. Targeted deletion reveals essential and overlapping functions of the miR-17 through 92 family of miRNA clusters. Cell 2008, 132, 875–886. [Google Scholar] [CrossRef] [PubMed]
  176. Lu, Y.; Thomson, J.M.; Wong, H.Y.; Hammond, S.M.; Hogan, B.L. Transgenic over-expression of the microRNA miR-17–92 cluster promotes proliferation and inhibits differentiation of lung epithelial progenitor cells. Dev. Biol. 2007, 310, 442–453. [Google Scholar] [CrossRef] [PubMed]
  177. Herriges, M.J.; Swarr, D.T.; Morley, M.P.; Rathi, K.S.; Peng, T.; Stewart, K.M.; Morrisey, E.E. Long noncoding RNAs are spatially correlated with transcription factors and regulate lung development. Genes Dev. 2014, 28, 1363–1379. [Google Scholar] [CrossRef] [PubMed]
  178. Davidson, J.M. Biochemistry and turnover of lung interstitium. Eur. Respir. J. 1990, 3, 1048–1063. [Google Scholar] [PubMed]
  179. Lane, S.; Rippon, H.J.; Bishop, A.E. Stem cells in lung repair and regeneration. Regen. Med. 2007, 2, 407–415. [Google Scholar] [CrossRef] [PubMed]
  180. Dunsmore, S.E.; Rannels, D.E. Extracellular matrix biology in the lung. Am. J. Physiol. 1996, 270, L3–L27. [Google Scholar] [PubMed]
  181. Suki, B.; Ito, S.; Stamenovic, D.; Lutchen, K.R.; Ingenito, E.P. Biomechanics of the lung parenchyma: Critical roles of collagen and mechanical forces. J. Appl. Physiol. 2005, 98, 1892–1899. [Google Scholar] [CrossRef] [PubMed]
  182. Kreis, T.; Vale, R. Guidebook to the Extracellular Matrix, Anchor, and Adhesion Proteins, 2nd ed.; Oxford University Press: Oxford, UK, 1999; p. 422. [Google Scholar]
  183. Olsen, C.O.; Isakson, B.E.; Seedorf, G.J.; Lubman, R.L.; Boitano, S. Extracellular matrix-driven alveolar epithelial cell differentiation in vitro. Exp. Lung Res. 2005, 31, 461–482. [Google Scholar] [CrossRef] [PubMed]
  184. Isakson, B.E.; Lubman, R.L.; Seedorf, G.J.; Boitano, S. Modulation of pulmonary alveolar type II cell phenotype and communication by extracellular matrix and KGF. Am. J. Physiol. Cell Physiol. 2001, 281, C1291–C1299. [Google Scholar] [PubMed]
  185. Lin, Y.M.; Zhang, A.; Rippon, H.J.; Bismarck, A.; Bishop, A.E. Tissue engineering of lung: The effect of extracellular matrix on the differentiation of embryonic stem cells to pneumocytes. Tissue Eng. Part A 2010, 16, 1515–1526. [Google Scholar] [CrossRef] [PubMed]
  186. Petersena, T.H.; Calle, E.A.; Niklason, L.E. Strategies for lung regeneration. Mater. Today 2011, 14, 196–201. [Google Scholar] [CrossRef]
  187. Daley, W.P.; Peters, S.B.; Larsen, M. Extracellular matrix dynamics in development and regenerative medicine. J. Cell Sci. 2008, 121, 255–264. [Google Scholar] [CrossRef] [PubMed]
  188. Odorico, J.; Kaufman, D.; Thomson, J. Multilineage differentiation from human embryonic stem cell lines. Stem Cells 2001, 19, 193–204. [Google Scholar] [CrossRef] [PubMed]
  189. Takahashi, K.; Tanabe, K.; Ohnuki, M.; Narita, M.; Ichisaka, T.; Tomoda, K.; Yamanaka, S. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 2007, 131, 861–872. [Google Scholar] [PubMed]
  190. Takahashi, K.; Yamanaka, S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 2006, 126, 663–676. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  191. Yu, J.; Vodyanik, M.; Smuga-Otto, K.; Antosiewicz-Bourget, J.; Frane, J.L.; Tian, S.; Nie, J.; Jonsdottir, G.A.; Ruotti, V.; Stewart, R.; et al. Induced pluripotent stem cell lines derived from human somatic cells. Science 2007, 318, 1917–1920. [Google Scholar] [CrossRef] [PubMed]
  192. Perl, A.; Kist, R.; Shan, Z.; Scherer, G.; Whitsett, J. Normal lung development and function after Sox9 inactivation in the respiratory epithelium. Genesis 2005, 41, 23–32. [Google Scholar] [CrossRef] [PubMed]
  193. Shu, W.; Lu, M.; Zhang, Y.; Tucker, P.; Zhou, D.; Morrisey, E. Foxp2 and Foxp1 cooperatively regulate lung and esophagus development. Development 2007, 134, 1991–2000. [Google Scholar] [CrossRef] [PubMed]
  194. Van Haute, L.; de Block, G.; Liebaers, I.; Sermon, K.; DeRycke, M. Generation of lung epithelial-like tissue from human embryonic stem cells. Respir. Res. 2009, 5. [Google Scholar] [CrossRef] [PubMed]
  195. Green, M.; Chen, A.; Nostro, M.; d’Souza, S.; Lemischka, I.; Gouon-Evans, V.; Keller, G.; Snoeck, H.W. Generation of anterior foregut endoderm from human embryonic and induced pluripotent stem cells. Nat. Biotechnol. 2011, 29, 267–272. [Google Scholar] [CrossRef] [PubMed]
  196. Mou, H.; Zhao, R.; Sherwood, R.; Ahfeldt, T.; Lapey, A.; Wain, J.; Sicilian, L.; Izvolsky, K.; Musunuru, K.; Cowan, C.; et al. Generation of multipotent lung and airway progenitors from mouse ESCs and patient-specific cystic fibrosis iPSCs. Cell Stem Cell 2012, 10, 385–397. [Google Scholar] [CrossRef] [PubMed]
  197. Longmire, T.A.; Ikonomou, L.; Hawkins, F.; Christodoulou, C.; Cao, Y.; Jean, J.C.; Kwok, L.W.; Mou, H.; Rajagopal, J.; Shen, S.S.; et al. Efficient derivation of purified lung and thyroid progenitors from embryonic stem cells. Cell Stem Cell 2012, 10, 398–411. [Google Scholar] [CrossRef] [PubMed]
  198. Wong, A.; Bear, C.; Chin, S.; Pasceri, P.; Thompson, T.O.; Huan, L.J.; Ratjen, F.; Ellis, J.; Rossant, J. Directed differentiation of human pluripotent stem cells into mature airway epithelia expressing functional CFTR protein. Nat. Biotechnol. 2012, 9, 876–882. [Google Scholar] [CrossRef] [PubMed]
  199. Huang, S.X.; Islam, M.N.; O’Neill, J.; Hu, Z.; Yang, Y.G.; Chen, Y.W.; Mumau, M.; Green, M.D.; Vunjak-Novakovic, G.; Bhattacharya, J.; et al. Efficient generation of lung and airway epithelial cells from human pluripotent stem cells. Nat. Biotechnol. 2014, 32, 84–91. [Google Scholar] [CrossRef] [PubMed]
  200. Berry, M.F.; Engler, A.J.; Woo, Y.J.; Pirolli, T.J.; Bish, L.T.; Jayasankar, V.; Morine, K.J.; Gardner, T.J.; Discher, D.E.; Sweeney, H.L. Mesenchymal stem cell injection after myocardial infarction improves myocardial compliance. Am. J. Physiol. Heart Circ. Physiol. 2006, 290, 196–203. [Google Scholar] [CrossRef] [PubMed]
  201. Kotton, D.N.; Ma, B.Y.; Cardoso, W.V.; Sanderson, E.A.; Summer, R.S.; Williams, M.C.; Fine, A. Bone marrow-derived cells as progenitors of lung alveolar epithelium. Development 2001, 128, 5181–5188. [Google Scholar] [PubMed]
  202. Morigi, M.; Imberti, B.; Zoja, C.; Corna, D.; Tomasoni, S.; Abbate, M.; Rottoli, D.; Angioletti, S.; Benigni, A.; Perico, N.; et al. Mesenchymal stem cells are renotropic, helping to repair the kidney and improve function in acute renal failure. J. Am. Soc. Nephrol. 2004, 15, 1794–1804. [Google Scholar] [CrossRef] [PubMed]
  203. Rojas, M.; Xu, J.; Woods, C.R.; Mora, A.L.; Spears, W.; Roman, J.; Brigham, K.L. Bone marrow-derived mesenchymal stem cells in repair of the injured lung. Am. J. Respir. Cell Mol. Biol. 2005, 33, 145–152. [Google Scholar] [CrossRef] [PubMed]
  204. Baber, S.R.; Deng, W.; Master, R.G.; Bunnell, B.A.; Taylor, B.K.; Murthy, S.N.; Hyman, A.L.; Kadowitz, P.J. Intratracheal mesenchymal stem cell administration attenuates monocrotaline-induced pulmonary hypertension and endothelial dysfunction. Am. J. Physiol. Heart Circ. Physiol. 2007, 292, 120–128. [Google Scholar]
  205. Burdon, T.J.; Paul, A.; Noiseux, N.; Prakash, S.; Shum-Tim, D. Bone marrow stem cell derived paracrine factors for regenerative medicine: Current perspectives and therapeutic potential. Bone Marrow Res. 2011, 2011, 207326. [Google Scholar] [CrossRef] [PubMed]
  206. Nemeth, K.; Leelahavanichkul, A.; Yuen, P.S.; Mayer, B.; Parmelee, A.; Doi, K.; Robey, P.G.; Leelahavanichkul, K.; Koller, B.H.; Brown, J.M.; et al. Bone marrow stromal cells attenuate sepsis via prostaglandin E(2)-dependent reprogramming of host macrophages to increase their interleukin-10 production. Nat. Med. 2009, 15, 42–49. [Google Scholar] [CrossRef] [PubMed]
  207. Ortiz, L.A.; Dutreil, M.; Fattman, C.; Pandey, A.C.; Torres, G.; Go, K.; Phinney, D.G. Interleukin 1 receptor antagonist mediates the anti-inflammatory and anti-fibrotic effect of mesenchymal stem cells during lung injury. Proc. Natl. Acad. Sci. USA 2007, 104, 11002–11007. [Google Scholar] [CrossRef] [PubMed]
  208. Zhen, G.; Liu, H.; Gu, N.; Zhang, H.; Xu, Y.; Zhang, Z. Mesenchymal stem cells transplantation protects against rat pulmonary emphysema. Front. Biosci. 2008, 13, 3415–3422. [Google Scholar] [CrossRef] [PubMed]
  209. Akram, K.M.; Samad, S.; Spiteri, M.A.; Forsyth, N.R. Mesenchymal stem cells promote alveolar epithelial cell wound repair in vitro through distinct migratory and paracrine mechanisms. Respir. Res. 2013, 14. [Google Scholar] [CrossRef] [PubMed]
  210. Gnecchi, M.; Zhang, Z.; Ni, A.; Dzau, V.J. Paracrine mechanisms in adult stem cell signalling and therapy. Circ. Res. 2008, 103, 1204–1219. [Google Scholar] [CrossRef] [PubMed]
  211. Suzuki, G.; Iyer, V.; Lee, T.C.; Canty, J.M., Jr. Autologous mesenchymal stem cells mobilize cKit+ and CD133+ bone marrow progenitor cells and improve regional function in hibernating myocardium. Circ. Res. 2011, 109, 1044–1054. [Google Scholar] [CrossRef] [PubMed]
  212. Islam, M.; Das, S.; Emin, M.; Wei, M.; Sun, L.; Westphalen, K.; Rowlands, D.J.; Quadri, S.K.; Bhattacharya, S.; Bhattacharya, J. Mitochondrial transfer from bone marrow derived stromal cells to pulmonary alveoli protects against acute lung injury. Nat. Med. 2012, 18, 759–765. [Google Scholar] [CrossRef] [PubMed]
  213. Lee, C.; Mitsialis, S.; Aslam, M.; Vitali, S.; Vergadi, E.; Konstantinou, G.; Sdrimas, K.; Fernandez-Gonzalez, A.; Kourembanas, S. Exosomes mediate the cytoprotective action of mesenchymal stromal cells on hypoxia-induced pulmonary hypertension. Circulation 2012, 126, 2601–2611. [Google Scholar] [CrossRef] [PubMed]
  214. Akram, K.M.; Samad, S.; Spiteri, M.; Forsyth, N.R. Mesenchymal stem cell therapy and lung diseases. Adv. Biochem. Eng. Biotechnol. 2013, 130, 105–129. [Google Scholar] [PubMed]
  215. Ware, L.B.; Matthay, M.A. The acute respiratory distress syndrome. N. Engl. J. Med. 2000, 342, 1334–1349. [Google Scholar] [CrossRef] [PubMed]
  216. Danchuk, S.; Joni, H.; Hossain, F.F.; Sorge, R.F.; Ramsey, A.; Ryan, W.; Sullivan, D.E. Human multipotent stromal cells attenuate lipopolysaccharide-induced acute lung injury in mice via secretion of tumor necrosis factor-α-induced protein 6. Stem Cell Res. Ther. 2010, 2, 27–42. [Google Scholar]
  217. Gupta, N.; Su, X.; Popov, B.; Lee, J.W.; Serikov, V.; Matthay, M.A. Intrapulmonary delivery of bone marrow-derived mesenchymal stem cells improves survival and attenuates endotoxin-induced acute lung injury in mice. J. Immunol. 2007, 179, 855–1863. [Google Scholar] [CrossRef]
  218. Lee, J.W.; Fang, X.; Gupta, N.; Serikov, V.; Matthay, M.A. Allogeneic human mesenchymal stem cells for treatment of E. coli endotoxin-induced acute lung injury in the ex vivo perfused human lung. Proc. Natl. Acad. Sci. USA 2009, 106, 16357–16362. [Google Scholar] [CrossRef] [PubMed]
  219. Mei, S.H.J.; McCarter, S.D.; Deng, Y.; Parker, C.H.; Liles, W.C.; Stewart, D.J. Prevention of LPS-induced acute lung injury in mice by mesenchymal stem cells overexpressing angiopoietin 1. PLoS Med. 2007, 4, e269. [Google Scholar] [CrossRef] [PubMed]
  220. Chang, Y.S.; Oh, W.; Choi, S.J.; Sung, D.K.; Kim, S.Y.; Choi, E.Y.; Kang, S.; Jin, H.J.; Yang, Y.S.; Park, W.S. Human umbilical cord blood-derived mesenchymal stem cells attenuate hyperoxia-induced lung injury in neonatal rats. Cell Transplant. 2009, 18, 869–886. [Google Scholar] [CrossRef] [PubMed]
  221. Krasnodembskaya, A.; Song, Y.; Fang, X.; Gupta, N.; Serikov, V.; Lee, J.W.; Matthay, M.A. Antibacterial effect of human mesenchymal stem cells is mediated in part from secretion of the antimicrobial peptide LL-37. Stem Cells 2010, 28, 2229–2238. [Google Scholar] [CrossRef] [PubMed]
  222. Wilson, J.G.; Liu, K.D.; Zhuo, H.; Caballero, L.; McMillan, M.; Fang, X.; Cosgrove, K.; Vojnik, R.; Calfee, C.S.; Lee, J.W.; et al. Mesenchymal stem (stromal) cells for treatment of ARDS: A phase 1 clinical trial. Lancet Respir. Med. 2015, 3, 24–32. [Google Scholar] [CrossRef]
  223. Kanki-Horimoto, S.; Horimoto, H.; Mieno, S.; Kishida, K.; Watanabe, F.; Furuya, E.; Katsumata, T. Implantation of mesenchymal stem cells overexpressing endothelial nitric oxide synthase improves right ventricular impairments caused by pulmonary hypertension. Circulation 2006, 114, I181–I185. [Google Scholar] [CrossRef] [PubMed]
  224. Liang, O.; Mitsialis, S.; Chang, M.; Vergadi, E.; Lee, C.; Aslam, M.; Fernandez-Gonzalez, A.; Liu, X.; Baveja, R.; Kourembanas, S. Mesenchymal stromal cells expressing heme oxygenase-1 reverse pulmonary hypertension. Stem Cells 2011, 29, 99–107. [Google Scholar] [CrossRef] [PubMed]
  225. Lomas, N.J.; Watts, K.L.; Akram, K.M.; Forsyth, N.R.; Spiteri, M.A. Idiopathic pulmonary fibrosis: Immunohistochemical analysis provides fresh insights into lung tissue remodelling with implications for novel prognostic markers. Int. J. Clin. Exp. Pathol. 2012, 5, 58–71. [Google Scholar] [PubMed]
  226. Akram, K.M.; Lomas, N.J.; Spiteri, M.A.; Forsyth, N.R. Club cells inhibit alveolar epithelial wound repair via TRAIL-dependent apoptosis. Eur. Respir. J. 2013, 41, 683–694. [Google Scholar] [CrossRef] [PubMed]
  227. Akram, K.M.; Lomas, N.J.; Forsyth, N.R.; Spiteri, M.A. Alveolar epithelial cells in idiopathic pulmonary fibrosis display upregulation of TRAIL, DR4 and DR5 expression with simultaneous preferential over-expression of pro-apoptotic marker p53. Int. J. Clin. Exp. Pathol. 2014, 7, 552–564. [Google Scholar] [PubMed]
  228. Aguilar, S.; Scotton, C.; McNutty, K.; Nye, E.; Stamp, G.; Laurent, G.; Bonnet, D.; Janes, S.M. Bone marrow stem cells expressing keratinocyte growth factor via an inducible lentivirus protects against bleomycin induced pulmonary fibrosis. PLoS ONE 2009, 4, e8013. [Google Scholar] [CrossRef] [PubMed]
  229. Kumamoto, M.; Nishiwaki, T.; Matsuo, N.; Kimura, H.; Matsushima, K. Minimally cultured bone marrow mesenchymal stem cells ameliorate fibrotic lung injury. Eur. Respir. J. 2009, 34, 740–748. [Google Scholar] [CrossRef] [PubMed]
  230. Ortiz, L.; Gambelli, F.; McBride, C.; Gaupp, D.; Baddoo, M.; Kaminski, N.; Phinney, D.G. Mesenchymal stem cell engraftment in lung is enhanced in response to bleomycin exposure and ameliorates its fibrotic effects. Proc. Natl. Acad. Sci. USA 2003, 100, 8407–8411. [Google Scholar] [CrossRef] [PubMed]
  231. Guo, J.; Yi, E.S.; Havill, A.M.; Sarosi, I.; Whitcomb, L.; Yin, S.; Middleton, S.C.; Piguet, P.; Ulich, T.R. Intravenous keratinocyte growth factor protects against experimental pulmonary injury. Am. J. Physiol. 1998, 275, 800–805. [Google Scholar]
  232. Panos, R.J.; Rubin, J.S.; Csaky, K.G.; Aaronson, S.A.; Mason, R.J. Keratinocyte growth factor and hepatocyte growth factor/scatter factor are heparin-binding growth factors for alveolar type II cells in fibroblast-conditioned medium. J. Clin. Investig. 1993, 92, 969–977. [Google Scholar] [CrossRef] [PubMed]
  233. Yano, T.; Mason, R.J.; Pan, T.; Deterding, R.R.; Nielsen, L.D.; Shannon, J.M. KGF regulates pulmonary epithelial proliferation and surfactant protein gene expression in adult rat lung. Am. J. Physiol. Lung Cell. Mol. Physiol. 2000, 279, 1146–1158. [Google Scholar]
  234. Chapman, H.A.; Allen, C.L.; Stone, O.L. Abnormalities in pathways of alveolar fibrin turnover among patients with interstitial lung disease. Am. Rev. Respir. Dis. 1986, 133, 437–443. [Google Scholar] [PubMed]
  235. Abreu, S.C.; Antunes, M.A.; Pelosi, P.; Morales, M.M.; Rocco, P.R. Mechanisms of cellular therapy in respiratory diseases. Intensive Care Med. 2011, 37, 1421–1431. [Google Scholar] [CrossRef] [PubMed]
  236. Yuhgetsu, H.; Ohno, Y.; Funaguchi, N.; Asai, T.; Sawada, M.; Takemura, G.; Minatoguchi, S.; Fujiwara, H.; Fujiwara, T. Beneficial effects of autologous bone marrow mononuclear cell transplantation against elastase-induced emphysema in rabbits. Exp. Lung Res. 2006, 32, 413–426. [Google Scholar] [CrossRef] [PubMed]
  237. Weiss, D.; Casaburi, R.; Flannery, R.; Leroux-Williams, M.; Tashkin, D. A placebo-controlled, randomized trial of mesenchymal stem cells in COPD. Chest 2013, 143, 1590–1598. [Google Scholar] [CrossRef] [PubMed]
  238. Nichols, J.E.; Cortiella, J. Engineering of a complex organ: Progress toward development of a tissue-engineered lung. Proc. Am. Thorac. Soc. 2008, 5, 723–730. [Google Scholar] [CrossRef] [PubMed]
  239. Tomei, A.A.; Boschetti, F.; Gervaso, F.; Swartz, M.A. 3D collagen cultures under welldefined dynamic strain: A novel strain device with a porous elastomeric support. Biotechnol. Bioeng. 2009, 103, 217–225. [Google Scholar] [CrossRef] [PubMed]
  240. Omori, K.; Tada, Y.; Suzuki, T.; Nomoto, Y.; Matsuzuka, T.; Kobayashi, K.; Nakamura, T.; Kanemaru, S.; Yamashita, M.; Asato, R. Clinical application of in situ tissue engineering using a scaffolding technique for reconstruction of the larynx and trachea. Ann. Otol. Rhinol. Laryngol. 2008, 117, 673–678. [Google Scholar] [CrossRef] [PubMed]
  241. Omori, K.; Nakamura, T.; Kanemaru, S.; Magrufov, A.; Yamashita, M.; Shimizu, Y. In situ tissue engineering of the cricoid and trachea in a canine model. Ann. Otol. Rhinol. Laryngol. 2008, 117, 609–613. [Google Scholar] [CrossRef] [PubMed]
  242. Macchiarini, P.; Jungebluth, P.; Go, T.; Asnaghi, M.A.; Rees, L.E.; Cogan, T.A.; Dodson, A.; Martorell, J.; Bellini, S.; Parnigotto, P.P.; et al. Clinical transplantation of a tissue-engineered airway. Lancet 2008, 372, 2023–2030. [Google Scholar] [CrossRef]
  243. Ott, H.C.; Clippinger, B.; Conrad, C.; Schuetz, C.; Pomerantseva, I.; Ikonomou, L.; Kotton, D.; Vacanti, J.P. Regeneration and orthotopic transplantation of a bioartificial lung. Nat. Med. 2010, 16, 927–933. [Google Scholar] [CrossRef] [PubMed]
  244. Petersen, T.H.; Calle, E.A.; Zhao, L.; Lee, E.J.; Gui, L.; Raredon, M.B.; Gavrilov, K.; Yi, T.; Zhuang, Z.W.; Breuer, C.; et al. Tissue-engineered lungs for in vivo implantation. Science 2010, 329, 538–541. [Google Scholar] [CrossRef] [PubMed]
  245. Price, A.P.; England, K.A.; Matson, A.M.; Blazar, B.R.; Panoskaltsis-Mortari, A. Development of a decellularized lung bioreactor system for bioengineering the lung: The matrix reloaded. Tissue Eng. Part A 2010, 16, 2581–2591. [Google Scholar] [CrossRef] [PubMed]
  246. Daly, A.B.; Wallis, J.M.; Borg, Z.D.; Bonvillain, R.W.; Deng, B.; Ballif, B.A.; Jaworski, D.M.; Allen, G.B.; Weiss, D.J. Initial binding and recellularization of decellularized mouse lung scaffolds with bone marrow–derived mesenchymal stromal cells. Tissue Eng. Part A 2012, 18, 1–16. [Google Scholar] [CrossRef] [PubMed]
  247. Wallis, J.M.; Borg, Z.D.; Daly, A.B.; Deng, B.; Ballif, B.A.; Allen, G.B.; Jaworski, D.M.; Weiss, D.J. Comparative assessment of detergent-based protocols for mouse lung de-cellularization and re-cellularization. Tissue Eng. Part C Methods 2012, 18, 420–432. [Google Scholar] [CrossRef] [PubMed]
  248. Cortiella, J.; Niles, J.; Cantu, A.; Brettler, A.; Pham, A.; Vargas, G.; Winston, S.; Wang, J.; Walls, S.; Nichols, J.E. Influence of acellular natural lung matrix on murine embryonic stem cell differentiation and tissue formation. Tissue Eng. Part A 2010, 16, 2565–2580. [Google Scholar] [CrossRef] [PubMed]
  249. Ghaedi, M.; Calle, E.A.; Mendez, J.J.; Gard, A.L.; Balestrini, J.; Booth, A.; Bove, P.F.; Gui, L.; White, E.S.; Niklason, L.E. Human IPS cell-derived alveolar epithelium repopulates lung extracellular matrix. J. Clin. Investig. 2013, 123, 4950–4962. [Google Scholar] [CrossRef] [PubMed]
  250. Bonvillain, R.W.; Scarritt, M.E.; Pashos, N.C.; Mayeux, J.P.; Meshberger, C.L.; Betancourt, A.M.; Sullivan, D.E.; Bunnell, B.A. Nonhuman primate lung decellularization and recellularization using a specialized large-organ bioreactor. J. Vis. Exp. 2013, 82. [Google Scholar] [CrossRef] [PubMed]
  251. Booth, A.J.; Hadley, R.; Cornett, A.M.; Dreffs, A.A.; Matthes, S.A.; Tsui, J.L.; Weiss, K.; Horowitz, J.C.; Fiore, V.F.; Barker, T.H.; et al. Acellular normal and fibrotic human lung matrices as a culture system for in vitro investigation. Am. J. Respir. Crit. Care Med. 2012, 186, 866–876. [Google Scholar] [CrossRef] [PubMed]
  252. Gilpin, S.E.; Guyette, J.P.; Gonzalez, G.; Ren, X.; Asara, J.M.; Mathisen, D.J.; Vacanti, J.P.; Ott, H.C. Perfusion decellularization of human and porcine lungs: Bringing the matrix to clinical scale. J. Heart Lung Transplant. 2013, 33, 298–308. [Google Scholar] [CrossRef] [PubMed]
  253. Nichols, J.E.E.; Niles, J.; Riddle, M.; Vargas, G.; Schilagard, T.; Ma, L.; Edward, K.; La Francesca, S.; Sakamoto, J.; Vega, S.; et al. Production and assessment of decellularized pig and human lung scaffolds. Tissue Eng. Part A 2013, 19, 2045–2062. [Google Scholar] [CrossRef] [PubMed]
  254. Wagner, D.E.; Bonenfant, N.R.; Sokocevic, D.; Desarno, M.J.; Borg, Z.D.; Parsons, C.S.; Brooks, E.M.; Platz, J.J.; Khalpey, Z.I.; Hoganson, D.M.; et al. Three-dimensional scaffolds of acellular human and porcine lungs for high throughput studies of lung disease and regeneration. Biomaterials 2014, 35, 2664–2679. [Google Scholar] [CrossRef] [PubMed]

Share and Cite

MDPI and ACS Style

Akram, K.M.; Patel, N.; Spiteri, M.A.; Forsyth, N.R. Lung Regeneration: Endogenous and Exogenous Stem Cell Mediated Therapeutic Approaches. Int. J. Mol. Sci. 2016, 17, 128. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms17010128

AMA Style

Akram KM, Patel N, Spiteri MA, Forsyth NR. Lung Regeneration: Endogenous and Exogenous Stem Cell Mediated Therapeutic Approaches. International Journal of Molecular Sciences. 2016; 17(1):128. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms17010128

Chicago/Turabian Style

Akram, Khondoker M., Neil Patel, Monica A. Spiteri, and Nicholas R. Forsyth. 2016. "Lung Regeneration: Endogenous and Exogenous Stem Cell Mediated Therapeutic Approaches" International Journal of Molecular Sciences 17, no. 1: 128. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms17010128

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop