Next Article in Journal
EPs® 7630 Stimulates Tissue Repair Mechanisms and Modifies Tight Junction Protein Expression in Human Airway Epithelial Cells
Previous Article in Journal
AB Toxins as High-Affinity Ligands for Cell Targeting in Cancer Therapy
Previous Article in Special Issue
The Overlooked Microbiome—Considering Archaea and Eukaryotes Using Multiplex Nanopore-16S-/18S-rDNA-Sequencing: A Technical Report Focusing on Nasopharyngeal Microbiomes
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Stem/Progenitor Cells and Related Therapy in Bronchopulmonary Dysplasia

1
German Center for Lung Research (DZL), Department of Pulmonary and Critical Care Medicine and Infectious Diseases, Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Justus Liebig University Giessen, 35392 Giessen, Germany
2
Department of Pediatrics, Centre for Clinical and Translational Research (CCTR), Helios University Hospital Wuppertal, Witten/Herdecke University, 42283 Wuppertal, Germany
3
Institute for Lung Health (ILH), Member of the German Center for Lung Research (DZL), Justus Liebig University Giessen, 35392 Giessen, Germany
4
Department of Pediatrics, University Medical Center Rostock, University of Rostock, 18057 Rostock, Germany
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2023, 24(13), 11229; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms241311229
Submission received: 24 April 2023 / Revised: 18 June 2023 / Accepted: 4 July 2023 / Published: 7 July 2023

Abstract

:
Bronchopulmonary dysplasia (BPD) is a chronic lung disease commonly seen in preterm infants, and is triggered by infection, mechanical ventilation, and oxygen toxicity. Among other problems, lifelong limitations in lung function and impaired psychomotor development may result. Despite major advances in understanding the disease pathologies, successful interventions are still limited to only a few drug therapies with a restricted therapeutic benefit, and which sometimes have significant side effects. As a more promising therapeutic option, mesenchymal stem cells (MSCs) have been in focus for several years due to their anti-inflammatory effects and their secretion of growth and development promoting factors. Preclinical studies provide evidence in that MSCs have the potential to contribute to the repair of lung injuries. This review provides an overview of MSCs, and other stem/progenitor cells present in the lung, their identifying characteristics, and their differentiation potential, including cytokine/growth factor involvement. Furthermore, animal studies and clinical trials using stem cells or their secretome are reviewed. To bring MSC-based therapeutic options further to clinical use, standardized protocols are needed, and upcoming side effects must be critically evaluated. To fill these gaps of knowledge, the MSCs’ behavior and the effects of their secretome have to be examined in more (pre-) clinical studies, from which only few have been designed to date.

1. Introduction

Preterm birth is the leading cause of infant mortality and long-term morbidity and represents a significant healthcare burden [1,2,3]. Premature newborns may suffer from respiratory distress requiring mechanical ventilation and oxygen support. The prolonged use of oxygen increases its concentration in the lungs (hyperoxia), which can eventually lead to inflammation resulting in abnormal lung development [4,5]. The affected infants are at high risk to develop bronchopulmonary dysplasia (BPD), a multifactorial chronic lung disease characterized by immature lung vasculature, inflammation, restricted lung development, and disordered lung repair [6]. The lower the gestational age and birth weight, the higher the risk to suffer from clinical syndromes which are considered as BPD. To date, the pathologic description of BPD includes disrupted alveolarization and microvasculature development, fibrosis, and cystic emphysema, and is likely to emerge due to disturbed signaling pathways [7].
Alongside a number of postnatal challenges that contribute to the development of BPD, the results of several studies have implicated that adverse conditions during pregnancy like placental vascular disease, intrauterine growth restriction, or chorioamnionitis may already be sufficient to induce or at least promote BPD and its related consequences [8].
Several advances in perinatal and neonatal medicine have improved the survival rates of preterm babies. However, these infants are susceptible to short- or long-term respiratory complications and are at an increased risk of BPD [6]. The current therapy is limited to a few drugs, such as caffeine and vitamin A, or corticosteroids like dexamethasone. However, this pharmacological approach has been controversially discussed, since on the one hand it has been shown to be associated with neurodevelopmental complications [9,10], whereas on the other hand, a low-dose treatment of high-risk infants turned out to have a positive benefit vs danger relation [11,12]. Nevertheless, these drugs failed to address all aspects of the disease, including the impaired alveolarization and the inflammation. Thus, no successful handling is available to prevent the long-lasting consequences of BPD, like pulmonary hypertension, exercise intolerance with sometimes the need for life long oxygen supply, and echocardiographic abnormalities [13]. In this scenario, the need of alternative safe therapies has strongly emerged.
In BPD, inflammation is considered as an initiative characteristic, and infections are deemed as high-risk factors. The levels of proinflammatory cytokines and chemokines, like interleukin (IL)-1b, IL-6, IL-8, tumor necrosis factor (TNF)-α, MCP-1, -2, -3, and transforming growth factor (TGF)-β1 are all increased, while anti-inflammatory molecules, like IL-4, IL-13, and IL-10, growth factors (fibroblast growth factor 10 (FGF-10), and vascular endothelial growth factor A (VEGFA), and platelet-derived growth factor subunit A (PDGF-A) are all decreased, whereas they are required for the physiological development of the lung and its repair during injury [14]. Regarding inflammation, the nuclear factor-kappa B (NF-κB) pathway has been implicated in the pathogenesis of BPD. NF-κB is a transcription factor that plays a critical role in the regulation of the inflammation and cellular stress responses. In BPD, activation of the NF-κB pathway has been observed and linked to the persistent inflammation and oxidative stress that are characteristics of this disease [14].
Stem-cell based therapy has been proven to be promising. Stem/progenitor cells are important in preserving the structure of the lung, and their presence is fundamental for the repair of the damaged tissue in lung diseases.
Therapeutic approaches should therefore address the restoration of the function of the different stem/progenitor cells (as well as their presence) in their compartments, including growth factors modulating the inflammation derived from the damage. Interestingly, mesenchymal stem cells (MSCs) have recently emerged as a good candidate, as they support cells during their development [15,16]. Additionally, MSCs can modulate the immune response with a reduction in inflammation and the levels of pro-inflammatory cytokines, such as IL-6 and TNF-α. MSCs were identified in the tracheal aspirate of preterm newborns, showing phenotypical alterations compared to the MSCs from the control group of healthy babies [17,18]. The expression of proinflammatory cytokines in these cells was found to be higher compared to the control, strongly suggesting a role of MSCs in the pathology of BPD. Replacing these cells with exogenous MSCs could therefore represent a feasible treatment of BPD. This review focus on the mesenchymal and epithelial stem/progenitor cells.

2. Stem/Progenitor Cells in the Lung

In BPD, inflammation, oxidative stress, and mechanical ventilation can lead to abnormal lung growth and a reduced lung function. Studies have demonstrated that the number and function of stem and progenitor cells are altered in the lungs of infants with BPD [19]. Lung stem/progenitor cells belong to the endothelial, mesenchymal, and epithelial lineages [20]. The regeneration and repair of damaged lung tissue occurs by either the proliferation of already differentiated cells, or by the differentiation of stem/progenitor cells. Basal cells and variant club cells (VCCs) are present in the bronchi and in proximity of the neuroepithelial bodies, and they can give rise to club cells (CCs), ciliated cells, and goblet cells (GCs) [21]. In the alveoli, it is possible to identify bronchioalveolar stem cells (BASCs) that can differentiate into CCs, ciliated cells, GCs, and alveolar type 1 and 2 (AT1 and 2) cells, with AT2 cells having the ability to self-renew and give rise to the AT1 cells.
The lung contains several different stem and progenitor cells, including basal cells (BCs), variant cells, GCs, BASCs, distal airway stem cells (DASCs), and AT2 cells (Figure 1 and Table 1 summarize the different localization of the cells and their specific markers). The activation of the stem/progenitor cells is associated with the level of lung injury. A severe injury of the alveolar epithelium leads to the activation of the non-alveolar epithelial type II cells to repair the damage; a mild injury induces the activation of the AT2 cells for the resolution of the damage. All these different types of cells could cover a potential therapeutic role in the treatment of BPD, including the MSCs and their extracellular vesicles (EVs) that have emerged as a promising therapeutic option as will be discussed further below.

2.1. Epithelial Progenitors

2.1.1. Basal Cells (BCs)

BCs are the most primitive cell type in the airways and are capable of self-renewal and differentiation into other cell types, including ciliated, goblet, pulmonary neuroendocrine cells (PNECs), ionocytes, and club cells in steady-state and after acute lung injury to restore lung homeostasis [22,23,24,25]. They are present throughout the respiratory tree and account for about 30% of the total airway epithelial cells in the upper airways, while their number gradually decreases to 6% in the smaller airways, respectively [26,27]. Tumor protein (TP)63 and cytokeratin (KRT)5 are considered as the common markers to identify these BCs, along with podoplanin (PDPN) [28,29]. KRT8 is upregulated in the BCs just before they differentiate into luminal cells, generating an intermediate KRT8+ basal luminal precursor cell under homeostatic conditions [24,25,26]. Intermediate [29,30,31] luminal precursor cells can differentiate into secretory and ciliated cells and PNECs [25,32,33,34,35]. In addition to the KRT5+ progenitors, new KRT17+ basal progenitor cells were identified in the mouse airway epithelium using time-series single-cell transcriptomic analysis combined with in vivo lineage tracing [36]. It has been shown in murine models that basal cells are formed during prenatal development and contribute to the pool of luminal cells [37].
It seems that BCs play a role in regeneration and repair as well as they have been implicated in chronic lung disease. However, little is known about these cells in preterm neonates due to limitations in basal cell isolation techniques. A study has shown that these cells, isolated from the nasopharyngeal aspirates of preterm infants, could differentiate into the airway epithelium, thereby providing the possibility for future therapy in neonatal lung diseases like BPD [38].

2.1.2. Secretory Cells (SCs): Variant Club Cells (VCCs), Club Cells, and Goblet Cells

CCs express secretory proteins (e.g., CC10) in addition to bronchiolar surfactants and proteases important for the regulation of epithelial integrity, immunity, and host-defense interactions [38,39,40]. A destruction and airway ciliated cell abnormality leading to a defect in mucociliary clearance has been found in premature infants developing BPD [40]. Moreover, ciliated cells of preterm infants showed a decline in ciliary beating, thus making the epithelium more vulnerable to external stimuli and injuries [38,41]. They are replenished by the basal cells, and their differentiation is maintained by Notch signaling [29]. It is possible to recognize two different types of CCs based on their sensitivity to naphthalene. The toxin induces the death of most of the CCs. However, a subpopulation is resistant to the injury, and is characterized by the lack of expression of the cytochrome P450 family member CYP2F2 [41]. These cells are the VCCs. They can reconstitute the injured epithelium through self-renewal and differentiation into the ciliated cells and CCs. VCCs can be considered as epithelial progenitors in small airways after injury.
GCs are secretory cells possessing numerous vesicle-bound mucin granules for mucus production, thus creating a mucous gel layer for the efficient mucociliary clearance and pathogen entrapment. Under healthy conditions, there is an equilibrium present between mucus production and clearance, however, in chronic airway diseases, hyperplasia and metaplasia of GCs can occur [41,42,43]. Moreover, they were found to metabolize xenobiotics via the production of P450 mono-oxygenase [44].
In infants with BPD, a decrease in the number of CCs as well as in the expression of the CC-derived protein secretoglobin (SCGB) 1A1 in the bronchiolar epithelium was reported [45,46,47]. This accelerated CC death might be a result of supplemental oxygen therapy in these infants. CCs are termed as the second stem cells in the airways since they serve as progenitors for ciliated and mucus-secreting cells.
Taken together, this could indicate a decrease in the levels of goblet cells in BPD because of CC hypoplasia, and hence this deficiency in the stem cell pool could lead to a lower host defense and interfere with the normal epithelial turnover and regeneration [48]. It has also been shown that GCs are plastic cells, and they may be able of self-renewal and trans-differentiation into a hybrid ciliated epithelial cell, thereby suggesting the presence of a transitional state [49].

2.1.3. Pulmonary Neuroendocrine Cells (PNECs)

The first cells to be formed within the epithelium are the PNECs. They are present throughout the bronchial tree either as single cells in humans, or in clusters as neuroepithelial bodies in rodents, and their number increases from the bronchi to the terminal bronchioles [50,51,52]. A great number of PNECs is present in the fetal lung, suggesting that they play a role during fetal lung development [53]. Moreover, they are involved in the communication between the nervous and immune systems [53,54,55]. It has been shown that the number of PNECs as well as their peptide contents change in babies with BPD [56,57,58], which was also observed in a rodent model of hyperoxia. Additionally, increased levels of gastrin-releasing peptide (GRP)-, and calcitonin- and serotonin-immunoreactive PNECs have been reported in infants who died from BPD. In contrast, post mortem analysis of pre-term infants with respiratory distress syndrome revealed a decrease in the GRP+ PNECs that was assumingly caused by degranulation [56]. The overactivation and hyperplasia of PNECs have been reported in BPD, indicating a changed environment in BPD induced by chronic hyperoxia [56,59]. Hence, the dysfunction of pulmonary progenitor cells in BPD leads to an impairment in pulmonary growth and to the inability of the immature lung to repair itself [60].

2.1.4. Alveolar Progenitor Cells

The distal alveolar region is lined with squamous type 1 and cuboidal 2 alveolar epithelial cells, also termed alveolar type 1 (AT1) and alveolar type 2 (AT2). AT2s are considered as alveolar progenitors with self-renewal abilities. Their turnover within the normal lung is slow, however, upon lung injury, they were shown to rapidly proliferate and differentiate into AT1 cells for maintaining the alveolar homeostasis [61,62,63,64,65,66]. The differentiation of AT2 into AT1 cells was observed in a study on neonatal rats under hyperoxia, and increased AT2 cell proliferation was observed in human severe type 2 BPD as well as in premature baboons following oxygen support and mechanical ventilation [67,68,69,70]. It is known that infants affected with BPD show a reduced formation of alveoli which continues to persist into adulthood [71,72]. Moreover, a decline in AT1 cells has been observed in infants with BPD, which is accompanied with alveolar simplification and the suspension of alveolarization [63,73].
Distal lung progenitor cells represent a subset of AT2 cells. They are responsible for the physiological turnover and for the repair through their ability to self-renew and differentiate into AT1 cells both in vivo and in vitro. In addition, they have regenerative potential: following pneumonectomy, an increase in AT2 cell numbers has been observed in mice, with a compensatory growth of the remaining lung parenchyma [74,75,76,77]. In the in vivo BPD model, hyperoxia exposure has been shown to deplete the AT2 cells [78]. This damage to the lung may contribute to an impaired or incomplete lung development and a resultant lifelong lung disease. The regenerative potential of AT2 cells therefore makes them interesting as a possible therapeutic intervention. In the in vivo model, primary murine AT2 cells were shown to prevent O2-induced functional and structural lung injury. Furthermore, it was demonstrated that induced human pluripotent stem cells also protected against lung injury in mice [79]. It has shown in past research how the intratracheal injection of progenitors can improve the structure and functions of the lung. Isolated AT2 cells were injected to prevent lung injury in an established oxygen-induced mouse model mimicking BPD. The results confirmed the safety of the administration and the efficacy of this procedure. However, the difficulty of isolating a sufficient number of these cells needs further work. Recently, different subpopulations of AT2 cells in terms of their activation status have been identified, thus increasing the complexity of a possible therapeutic approach with AT2 cells [80,81]
Figure 1. Schematic overview of the different cell types within the respiratory epithelium. Trachea to bronchi: the epithelial surfaces of the trachea and proximal airways are lined by a pseudostratified columnar epithelium consisting of basal, brush, ciliated, goblet, and club cells, along with the less frequent pulmonary neuroendocrine cells (PNECs) and ionocytes. Bronchioles to alveoli: small airways are lined by a simple columnar or cuboidal epithelium that consists of ciliated, club, and a few goblet cells. Alveoli: the alveoli are lined by either squamous alveolar type 1 (AT1) or cuboidal alveolar type 2 (AT2) cell.
Figure 1. Schematic overview of the different cell types within the respiratory epithelium. Trachea to bronchi: the epithelial surfaces of the trachea and proximal airways are lined by a pseudostratified columnar epithelium consisting of basal, brush, ciliated, goblet, and club cells, along with the less frequent pulmonary neuroendocrine cells (PNECs) and ionocytes. Bronchioles to alveoli: small airways are lined by a simple columnar or cuboidal epithelium that consists of ciliated, club, and a few goblet cells. Alveoli: the alveoli are lined by either squamous alveolar type 1 (AT1) or cuboidal alveolar type 2 (AT2) cell.
Ijms 24 11229 g001

2.1.5. Non-Alveolar Progenitors

The proximal airway progenitor cells include the already mentioned basal and secretory cells, while the distal progenitors comprise the VCCs and the non-alveolar epithelial type II derived progenitors, such as DASCs, BASCs, and lineage-negative epithelial progenitors (LNEPs).

2.1.6. BASCs and DASCs

BASCs expressing the cell markers of both AT2 (surfactant protein C (SFTPC)) and secretory cells (secretoglobin 1A1 (SCGB1A1)) differentiate into AT1 and AT2 cells following bleomycin or influenza virus-induced injury, but not after hyperoxia-induced alveolar injury [41,82,83]. AT2 cells are capable of self-renewal and differentiation into AT1 (HOPX+) cells during homeostasis and upon injury [65,67]. In addition, AT2 cells differentiate into SFTPC+ and stem cell antigen (SCA)l+ alveolar progenitor cells, which are capable in differentiating into AT1 cells. In addition to the progenitor AT2 cells, another type of alveolar progenitor cells expressing alpha-6-beta4-integrin but not SFTPC and SCGB1A1 have been identified [84]. These cells can self-renew and differentiate into CCs and AT2 cells. Interestingly, it was found that a small population of AT1 cells expressing HOPX can give rise to AT2 cells following partial pneumonectomy [85]. A new population of stem cells distinct from AT2 and club progenitor cells has been identified, termed DASCs [86,87]. They express TP63/KRT5, can migrate, and give rise to SCGB1A1+/CYP2F2+ secretory club cells, which can self-renew and differentiate into ciliated cells and BCs (SCGB1A1+ and SFTPC+) upon lung injury.

2.1.7. Lineage-Negative Epithelial Stem/Progenitors (LNEPs)

A lineage tracing study identified a very small subpopulation of quiescent club-like cells, characterized by a high expression of H2-K1 and a lack of TP63 that have the potential to differentiate into alveolar epithelial cells. These LNEPs are localized in the small distal airways and can contribute to the repair of the distal lung area [87].
Table 1. Summary of the specific markers that enable the precise identification of the epithelial stem/progenitor cell subpopulations present within the lung.
Table 1. Summary of the specific markers that enable the precise identification of the epithelial stem/progenitor cell subpopulations present within the lung.
Cell TypeLocationMarkersDifferentiation PotentialRefs.
Basal cells (BCs)Epithelium in the trachea and bronchiolesTP63, KRT5, KRT14, NGFR, PDPN, KRT8,
N2ICD, and
c-Myb/TP73
Self-renewal; AT1, AT2
basal luminal precursor cell,
neuroendocrine cells, ionocytes
CCs, GCs, and ciliated cells
[23,87,88,89,90,91,92,93,94]
Variant club cells/
Clara cells
Bronchi and bronchioles (epithelium)SCGB1A1 and CYP2F2Self-renewal: GCs and ciliated cells [82,95,96]
Neuroendocrine cells (PNECs)Epithelium, at branching points in the bronchi and bronchiolesGRP, ASCL1, CXCR4, NCAD and ROBOMulticiliated cells and secretory cells [49,55,97,98,99,100]
Broncho-alveolar stem cells (BASCs)Epithelium and distal airway alveolar sacsSCGB1A1 and SFTPCSelf-renewal: AT2
Ciliated cells: GCs and AT1
[101,102,103,104]
AT2Distal airway alveolar sacsSFTPC and LysMSelf-Renewal: AT1,
SCA1+ mesenchymal progenitors,
SFTPC+ AT1 progenitors,
[66,83,105,106,107,108]
Distal airway stem cells (DASCs),
Alveolar progenitor
Distal airways, bronchioles, and alveolar regionsTP63 and KRT5Self-Renewal: AT2[84]
Self-Renewal: [86]
AT2, SCGB1A1+, and CYP2F2+ CCs[86,96]
AT1 HOPX,AT2[84]
Club cells, ciliated cellsTerminal bronchioles and less in the respiratory bronchioles,
trachea, and bronchioles
SCGB1A1, and CYP2F2BCs[86]
Ciliated cells[96]
TubIVa and FOXJ1GCs [109]
Integrin α6β4+ alveolar progenitorTerminal bronchioles, broncho-alveolar junctions, and less in the respiratory bronchiolesSCGB1A1, SPC, ITGα6, and ITGβ4Self-Renewal: AT1
Ciliated cells: AT2 and CCs
[96]
Abbreviations: ASCL-1, achaete-scute homolog 1; AT1/2, alveolar type 1/2 cells; BCs, basal cells; CCs, club cells; c-Myb, transcription factor; CYP2F2, cytochrome P450 family 2 subfamily f member 2; CXCR4, chemokine receptor; FOXJ1, transcription factor of fox family; GCs, goblet cells; GRP, bombesin or gene-related peptide; HOPX, homeodomain-only protein; ITG, integrin; KRT, cytokeratin; LysM, lysozyme M; N2ICD, Notch intracellular domain 2; NCAD, N-cadherin; NGFR, nerve growth factor receptor; PDPN, podoplanin; ROBO, roundabout receptor; SCA-1, stem cell antigen-1; SCGB1A1, secretoglobin 1A1; SFTPC, surfactant protein C; TP, tumor protein; and TubIVa, human tubulin beta class Iva.

3. Role of EVs—Secretome of MSCs

Extracellular vesicles (EVs), enveloped by the nuclear membrane, are 40–100 nm in diameter and are secreted by a variety of cells. Their components form the secretome of the respective cells of origin. EVs carry extracellular messages and mediate cell–cell communication [17,110,111].
The secretome of the MSCs consists of growth factors, cytokines, RNAs, and microRNAs, and differs depending on the tissue of MSC origin [112]. It can contribute to the beneficial effects of the EV on inflammation, repair, and organ development [113]. Their biological properties make MSCs very attractive for therapeutic use in the prevention and treatment of BPD [114]. This has already been assessed in several in vivo models with positive results on different diseases, such as in lung injury, cardiovascular disease, acute kidney injury, fetal hypoxic ischemic brain injury, wound healing, and hypoxic pulmonary hypertension [115,116,117,118,119]. Further studies have also shown that the use of MSC EVs alone does not lead to any disadvantages when compared to the complete MSCs [113]. EVs mimic their parental cells, MCSs, and they represent the major contributors of the paracrine effects of the MSCs. Moreover, EVs can be bioengineered to enhance their migratory properties [120].
Therefore, the use of MSC-derived EVs is a promising new therapeutic modality for BPD as it is a cell-free procedure, and thus can circumvent the concerns associated with viable MSC treatment [121]. However, before considering EVs as a therapeutic approach, additional preclinical studies are required to enlighten the effects on BPD and its underlying mechanisms, as well as to standardize the vesicle’s isolation and delivery.

4. Growth Factors and Inflammation in BPD

Risk factors for developing BPD include infections and injuries of the lung leading to inflammatory reactions. A major characteristic of BPD is the persistent inflammation contributing to impaired alveolarization and disturbing the cell–cell communication, as well as the correct response to the proliferation/differentiation signals. It is known that the dysregulation of several growths factors, like the vascular endothelial growth factor (VEGF), insulin-like growth factor 1 (IGF-1), PDGF, TGF-β1, and FGF10, plays an important role in the pathogenesis of BPD by affecting the inflammatory response. Several studies have shown an imbalance between the pro- and anti-inflammatory cytokines, which can predict the onset of BPD. For example, the concentration of pro-inflammatory cytokines has been shown to be increased in the amniotic fluid of preterm infants who develop BPD. Furthermore, cytokines are produced and secreted by several types of cells, are involved in cell–cell communications, and aid in modulating the answer to tissue damage.

5. Fibroblast Growth Factor 10 (FGF10)

FGF10 is a key growth factor for organ morphogenesis, including the lung [122]. During homeostasis in adult lungs, its expression is restricted to the mesenchymal niches and lipofibroblasts (LIFs), which are located close to the AT2 cells in the alveoli. LIFs, a fibroblast subpopulation, are emerging as a new possible source of FGF10 [123]. LIFs are characterized by the presence of lipid droplets, which are the main but not the only source of lipids to produce the surfactant lipoproteins in the AT2 cells [124]. Little is known about these cells, and even if they seem to be important in de novo alveologenesis during regeneration after lung injury [125], further studies are needed to define their role and relation with FGF10. In case of injury, induced by either bleomycin or influenza [126], FGF10 expression stimulates epithelial proliferation to promote the regeneration of the lung. FGF10 signaling is dysregulated in several lung diseases, including BPD [127].
In preterm infants with a severe or fatal course of BPD, a reduction in FGF10 levels has been observed. It has been published that in a mouse model of BPD (hyperoxia exposure) decreased FGF10 levels led to the premature death of the animals associated with profound alveolar defects and impaired AT2 cell differentiation, along with a decreased surfactant production [128]. Moreover, the administration of exogenous FGF10 to pups exposed to hyperoxia induced de novo alveologenesis. Furthermore, it has been published on how MSCs from the distal airways (LR-MSCs) of FGF10-pretreated rats can be easily isolated due to the mobilization effect of the FGF10 administration. LR-MSCs were injected into rat lungs with lipopolysaccharide (LPS)-induced acute injury, proving their protective role [129]. In these MSCs, FGF10 was additionally demonstrated to play a role in proliferation, mobilization, and organ-specific protective effects in acute lung injury [129]. Another in vivo study demonstrated that the administration of recombinant FGF10 induced de novo alveologenesis in a BPD mouse model [130].
Interestingly, it has been shown that FGF10 released from the mesenchyme correlates with an increased expression of VEGF in the epithelium [131].

6. Vascular Endothelial Growth Factor (VEGF)

VEGF regulates angiogenesis under both physiological and pathological conditions. The expression of VEGF is regulated by cytokines, such as IGF-1, but is also influenced by hyperoxia. This occurs via the mitogen-activated protein kinase (MAPK) and AKT signaling pathways, and thus impacts endothelial repair, regeneration, and vascularization. VEGF is secreted by MSCs and influences the differentiation of endothelial progenitor cells (EPCs). MSCs, can in turn differentiate into endothelial cells under VEGF treatment in vitro [132]. Interestingly, in vitro and in vivo hyperoxia experiments have shown that transplanted MSCs secrete VEGF, suggesting that this growth factor plays a role in protection against oxygen injury [133].

7. NF-κB

In the inflammation status observed in BPD and other lung injuries, the mesenchymal as well as the epithelial compartment is impacted by the cytokine storm, along with a dysregulation in several signaling pathways, like these of FGF10 and VEGF. In vivo models revealed that an increase in NF-κB is associated with lung injury [134]. Furthermore, it has been observed that hyperoxia, mechanical ventilation, oxidative stress, and endotoxins can lead to NF-κB activation [135,136,137]. One of several described anti-inflammatory modes of action of NF-κB is its ability to modulate macrophage polarization [138]. However, NF-κB can be either a pro-inflammatory or an anti-inflammatory factor based on the respective situation [139,140]. Many downstream target genes of NF-κB are important for innate and adaptive immune responses, and the activation of NF-κB has been described in various inflammatory diseases. In addition to these inflammatory processes, NF-κB regulates proliferation, differentiation, and angiogenesis [141,142]. As NF-κB mediates angiogenesis through the regulation of cytokines and growth factors, the inhibition of NF-κB has been described to block FGF-induced angiogenesis [142]. NF-κB regulates proliferation and apoptosis by controlling the transcription of several anti-apoptotic genes [143]. In an in vivo mouse model, NF-κB was shown to be constitutively active in newborn mice at the beginning of the alveolar stage, whereas it is only weakly expressed in the adult lungs [144]. The inhibition of the NF-κB pathway impairs alveolarization in neonatal mice through the inhibition of proliferation and the upregulation of apoptosis. Studies have indicated that the activation of NF-κB during the early stages of lung development can have a negative impact on development. For example, in lung explants at the late canalicular/early saccular stage, it has been shown that treatment with LPSs impedes lung branching [145]. This was thought to be due to NF-κB activation by LPSs in lung macrophages. Consequently, the depletion of macrophages or the inactivation of NF-κB could rescue lung branching [144], and NF-κB activation in macrophages resulted in an impaired branching [146]. NF-κB has been suspected to either act constitutively or to be induced otherwise. It has also been assumed that it acts differently at various stages of lung development. In preterm infants with BPD, an increased activity of NF-κB in tracheal lavage fluid could be detected [147]. Nevertheless, other studies have shown that a polymorphism in the NF-ΚB IA gene and a resulting increased activation of NF-κB reduces the risk of moderate-to-severe BPD [148,149]. Therefore, it has been suggested that NF-κB may play a protective role in late lung development [148].
MSCs and NF-κB are closely linked. MSCs isolated from preterm infants exhibit alterations in the PDGF-A, β-catenin, and TGF-β1 pathways, which are responsible for the MSCs in developing a myofibroblastic phenotype [150]. NF-κB can drive the proliferation of the MSCs, with the accumulation of the NF-κBp65 complex in the nucleus being the central regulatory mechanism. This is accompanied with a reduction in α-smooth muscle actin (α-SMA) expression, which is typical of the myofibroblastic phenotype [151]. However, we need to keep in mind that NF-κB pathway alterations, both activation and suppression, can lead to a disrupted lung development, emphasizing the intricate interplay between the NF-κB and other pathways in the cell. Due to the described impact on the MSCs and more generally on the inflammation, further analysis is needed to clarify the mechanisms underlying the interaction between NF-κB and the MSCs in order to balance the therapeutic approach to reduce the inflammatory process and its subsequent damages. Considering that NF-κB can exhibit both positive and negative impact on lung development, the time window along with the mechanisms of action on MSCs are crucial points [152].

8. Use of Stem Cells for the Treatment of BPD

Protocols for stem cell isolation and its subsequent in vivo administration are just barely developed far enough to be transferred from animal models to initial human clinical trials. A number of such studies have already addressed the safety and dose-responses with promising results, meaning that the next steps can focus more and more on therapeutic applications.
However, successful interventions still require a better understanding of these molecular processes, especially during differentiation of the cell types involved. This includes unique markers to improve cell-specific isolation, as well as a deeper knowledge of stem/progenitor cell physiology.
The mesenchymal compartment plays a fundamental role in supporting the proliferation and maintenance of the stem/progenitor niches in the lung (Figure 2A). Since MSCs give rise to different mesenchymal populations, like LIFs and myofibroblasts (MYFs), depending on the status of the tissues, the disturbance of their physiological function can cause an impaired lung development (Figure 2B). Thus, MSCs appear to be good candidates for stem cell therapy, and also because they are immune-privileged cells that do not induce host responses or cell rejection. Moreover, they can produce key molecules, such as FGF10 and PDGF [153,154], which influence the proliferation and differentiation of the stem/progenitor cells. As another advantage, they can be easily isolated from a wide range of tissues, like the bone marrow, peripheral blood, placenta, umbilical cord, and Wharton’s jelly [16,155]. The umbilical cord and Wharton’s jelly-isolated MSCs display more potent anti-inflammatory and immunomodulatory properties. Additional studies have focused more on the MSCs from the bone marrow and adipose tissues [15]. The safety profile of MSCs has been well established, and it is promising in that not only the MSC administration shows an improvement in the inflammation of the lung and its repair (e.g., alveolarization), but also of their EVs, suggesting a paracrine effect [112,113,114].
A recent study showed that a population of repair-supportive mesenchymal cells (RSMCs) rising from the GLI1+ cells is able to activate the club cell progenitors [156]. These RSMCs express high levels of FGF10 and enhanced WNT signaling. GLI1+ cells are usually located in the parabronchial, perivascular, and alveolar regions, and provide the FGF10-producing RSMCs in the lung. The comparison between the RSMCs and the resident MSCs (rMSCs) by scRNA-seq suggested the possibility that rMSCs could originate from the adjacent alveolar region surrounding the bronchi. Further investigations are needed to clarify whether this hypothesis is correct and transferable to human medicine [156].
To date, many animal studies have been published, highlighting a practical application of MSCs in the therapy of BPD, while only few clinical trials are being conducted. Nevertheless, from the preliminary results, one can be optimistic that within the next few years, we should be able to collect enough data to set up an effective therapeutic approach for BPD. In the next sections, we will focus on the most promising studies.

9. Animal Studies

Numerous studies in animal models provide evidence of the beneficial use of stem cell therapy in BPD, with MSCs being the most extensively used cell type [157,158,159]. The benefits of MSC therapy include reduced pulmonary fibrosis and edema, restoration of lung function, and attenuation of lung inflammation [160,161,162,163]. Considering the perturbation of stem cells in BPD, the repopulation, repair, and regeneration of the lung with healthy stem/progenitor cells would be an ideal therapeutic approach. Several studies have shown that intratracheal, intravenous, and intraperitoneal administrations of bone-marrow MSCs (BMSCs) have prevented alveolar arrest and attenuated lung inflammation in a neonatal lung injury model [164]. Additionally, this treatment has stimulated other stem/progenitor cells, termed BASCs, to aid in lung repair [164,165]. A recent study on BMSC administration in a mouse model of BPD demonstrated the ability of the cells to migrate from the site of the i.p. injection to the injured lung, resulting in an improvement in the pulmonary architecture, a reduction in pulmonary fibrosis, and an increase in the survival rate [166]. Mice were exposed to hyperoxia for a period of two weeks and were then treated with an intravenous dose of MSCs-conditioned media (MSC-CM). Subsequently, a reversal of hyperoxia-induced parenchymal fibrosis, partially reversed alveolar injury, and fully reversed pulmonary hypertension was observed, thus showing that MSCs-secreted factors can support the reversal of symptoms associated with hyperoxia-induced BPD [167]. In accordance with these findings, a similar study used intratracheal injections of umbilical cord-derived perivascular cells and cord blood-derived-MSCs in rat pups exposed to hyperoxia [155]. The administration of both cell types resulted in an improved alveolar architecture, thus partially restoring the lung function. Comparable results were obtained when using MSC- or PC-conditioned medium, suggesting that the therapeutic outcome observed was due to a paracrine effect [155].
Another cell type to achieve lung repair could be human amnion epithelial cells (hAECs). Administration of these cells in an animal model of LPS-induced injury in fetal sheep ameliorated pulmonary inflammation [168].

10. Clinical Trials

As previously described, in vivo hyperoxia studies in rodents demonstrated that the administration of MSCs resulted in an improvement in the lung structure of hyperoxia-damaged lungs [81]. This effect was even more pronounced when MSC-conditioned medium was administered [165,167], suggesting that MSCs or their secreted components ameliorate the imbalance of the inflammatory response and cytokines in oxygen-damaged lungs. These promising results led to the use of MSCs in clinical trials.
It has been described that in the tracheal aspirates of preterm infants who later developed severe BPD, TGF-β1 activity is significantly increased [18]. The MSCs of preterm infants with BPD express TGF-β1 and possess stronger β-catenin signaling compared to MSCs from infants who do not develop BPD [169]. Studies have shown that TGF-β1 and the nuclear translocation of β-catenin control the MYF differentiation [154,169]. In in vivo models, hyperoxia has been shown to damage the function of lung-resident stem cells with mesenchymal, endothelial, and epithelial differentiation potential [81]. These studies indicate that damage to MSCs in the lung may promote BPD [85].
In recent years, stem cells have been increasingly used in clinical trials not only relating to BPD. Currently, the results of three completed phase I clinical trials in preterm infants with BPD have been published, and a number of clinical trials (phase I and II) are either open for recruiting, ongoing, or have recently been completed. They are focusing on the benefit and safety of stem cell transplantation in BPD patients (NCT02443961; NCT03378063; NCT04003857; NCT03873506; NCT03631420; NCT03645525; and NCT03774537, respectively).
A phase I dose-escalation study examined the safety and feasibility of the intratracheal administration of MSCs in nine preterm infants with a mean birth weight of 793 ± 127 g and mechanical ventilation support being at high risk of developing BPD [170]. Three patients were given a low dose (1 × 107 cells/kg), and six patients were given a high dose (2 × 107 cells/kg), respectively. In tracheal aspirates at day 7 after MSC administration, a decrease in the pro-inflammatory cytokines TNF-α, IL6, IL8, and matrix metalloproteinase 9 levels was found, going in line with a lower severity of respiratory disease [170].
In a second phase I study, MSCs from the umbilical cord were administered to twelve preterm infants with a birth weight of <1000 g and a gestational age <28 weeks, respectively. While intratracheal treatment was well tolerated, ten infants developed severe BPD, and two encountered a mild BPD, respectively. In addition, three cases of sepsis were recorded [158]. In this study, premature infants were less mature, and had more severe respiratory disease compared to the first study, which may explain the poorer outcome.
In the third phase I study, the safety of late allogeneic amniotic cell transplantation was investigated in infants with severe BPD who either required invasive ventilation or non-invasive respiratory support with oxygen fractions of 0.3–0.5 [171]. In total, six premature infants were given a dose of 1 × 106 cells/kg, and after a slight adaptation of the procedure after the first patient, five infants tolerated the infusion without experiencing side effects. The treatment did not lower the need for respiratory support in any of the patients within the first week. However, FiO2 requirements decreased in three infants [171]. Although no treatment-associated adverse events were observed up to 24 months of age, the significance remained limited due to the severe BPD of the infants and the persistent problems of somatic growth, and cardiorespiratory and psychomotor function [172].
The subsequent phase I dose-escalation study has already begun, in which the administered cell number was increased up to 10 × 106 cells/kg per infusion and up to 3 × 107 cells/kg in total, respectively [173].
These first three phase I trials have primarily focused on the safety aspects of MSC administration and did not consider the overall benefit since the application and dosage or extraction of the MSCs must first be clarified for a successful MSC therapy. It has been shown that MSC treatment has no side effects in preterm infants and does not contribute to deterioration. However, the long-term effects of MSCs administration for more than two years has not yet been investigated.
Taken together, these clinical trials indicate that the safe administration of MSCs and any benefit from this treatment are likely to depend on several factors. The dose and timing of MSC administration and repeated treatments appear to be important in this regard, each alone and in combination. However, these factors have not yet been studied in detail.

11. Summary and Conclusions

The technical advance in the cell analysis leads to a better understanding of cell diversity, thereby improving the knowledge on the stem/progenitor cells present in the different regions of the lung. Alveolar progenitors and epithelial progenitors from the amniotic fluid are not yet well characterized to be translated in clinical research. Recent findings highlighted the MSCs as a possible therapeutic option to improve the damage occurred in the lung in case of injury, like virus-induced, hyperoxia-induced, or mechanical-induced injuries. MSCs seem to act via paracrine mechanisms, supporting the central role of their EVs. EVs contain a variety of mRNA, lipid, and proteins, and its content could be the key to the beneficial effects observed in the experiments. The administration of both the MSCs and their eVs showed positive outcomes, with an improved respiratory function and lung structure. However, there is still not enough knowledge in terms of the cell preparation, the number of cells, the time points, the type of administration, and the long-term studies on their safety. MSCs from the umbilical cord represent the best therapeutic option to be translated for clinical usage. One of the major obstacles is related to the expansion of the MSCs in vitro, as it is barely possible to keep track of the changes in the cells leading to an altered behavior. For this reason, the use of EVs or MSC-conditioned media could be an alternative. So far, the application of both is limited to animals.
Taken together, stem/progenitor cell-based therapies are very promising to counteract the damage in the injured lung. MSCs are the most extensively studied type of cells for this therapeutic application, and the preliminary studies are very promising. However, first, it is necessary to fill the gap of knowledge in the understanding of the MSC’s behavior, as well as to optimize/standardize the isolation protocol and their administration for future clinical trials.

Author Contributions

Conceptualization, C.-M.C. and S.B.; writing—original draft preparation, M.M., M.Y.G., N.E.-M. and V.O.; writing—review and editing, C.-M.C., M.M., N.E.-M., V.O. and S.B.; visualization, M.M., M.Y.G. and N.E.-M.; supervision, C.-M.C. and S.B. All authors have read and agreed to the published version of the manuscript.

Funding

We would like to thank Helios University Hospital Wuppertal and Witten/Herdecke University for their support.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Liu, L.; Oza, S.; Hogan, D.; Perin, J.; Rudan, I.; Lawn, J.E.; Cousens, S.; Mathers, C.; Black, R.E. Global, Regional, and National Causes of Child Mortality in 2000–13, with Projections to Inform Post-2015 Priorities: An Updated Systematic Analysis. Lancet 2015, 385, 430–440. [Google Scholar] [CrossRef]
  2. Liu, L.; Johnson, H.L.; Cousens, S.; Perin, J.; Scott, S.; Lawn, J.E.; Rudan, I.; Campbell, H.; Cibulskis, R.; Li, M.; et al. Global, Regional, and National Causes of Child Mortality: An Updated Systematic Analysis for 2010 with Time Trends since 2000. Lancet 2012, 379, 2151–2161. [Google Scholar] [CrossRef] [PubMed]
  3. Botet, F.; Figueras-Aloy, J.; Miracle-Echegoyen, X.; Rodriguez-Miguelez, J.M.; Salvia-Roiges, M.D.; Carbonell-Estrany, X. Trends in Survival among Extremely-Low-Birth-Weight Infants (Less than 1000 g) without Significant Bronchopulmonary Dysplasia. BMC Pediatr. 2012, 12, 63. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Bhandari, V.; Elias, J.A. Cytokines in Tolerance to Hyperoxia-Induced Injury in the Developing and Adult Lung. Free Radic. Biol. Med. 2006, 41, 4–18. [Google Scholar] [CrossRef] [PubMed]
  5. Bhandari, V.; Choo-Wing, R.; Lee, C.G.; Zhu, Z.; Nedrelow, J.H.; Chupp, G.L.; Zhang, X.; Matthay, M.A.; Ware, L.B.; Homer, R.J.; et al. Hyperoxia Causes Angiopoietin 2–Mediated Acute Lung Injury and Necrotic Cell Death. Nat. Med. 2006, 12, 1286–1293. [Google Scholar] [CrossRef] [Green Version]
  6. Smith, L.K.; Hindori-Mohangoo, A.D.; Delnord, M.; Durox, M.; Szamotulska, K.; Macfarlane, A.; Alexander, S.; Barros, H.; Gissler, M.; Blondel, B.; et al. Quantifying the Burden of Stillbirths before 28 Weeks of Completed Gestational Age in High-Income Countries: A Population-Based Study of 19 European Countries. Lancet 2018, 392, 1639–1646. [Google Scholar] [CrossRef]
  7. Thébaud, B.; Goss, K.N.; Laughon, M.; Whitsett, J.A.; Abman, S.H.; Steinhorn, R.H.; Aschner, J.L.; Davis, P.G.; McGrath-Morrow, S.A.; Soll, R.F.; et al. Bronchopulmonary Dysplasia. Nat. Rev. Dis. Prim. 2019, 5, 78. [Google Scholar] [CrossRef]
  8. Mestan, K.K.; Leibel, S.L.; Sajti, E.; Pham, B.; Hietalati, S.; Laurent, L.; Parast, M. Leveraging the Placenta to Advance Neonatal Care. Front. Pediatr. 2023, 11, 1174174. [Google Scholar] [CrossRef]
  9. Oluwole, I.; Tan, J.B.C.; DeSouza, S.; Hutchinson, M.; Leigh, R.M.; Cha, M.; Rodriguez, A.; Hou, G.; Rao, S.S.; Narang, A.; et al. The Association between Bronchopulmonary Dysplasia Grade and Risks of Adverse Neurodevelopmental Outcomes among Preterm Infants Born at Less than 30 Weeks of Gestation. J. Matern. Fetal Neonatal Med. 2023, 36, 2167074. [Google Scholar] [CrossRef]
  10. Yeh, T.F.; Lin, Y.J.; Lin, H.C.; Huang, C.C.; Hsieh, W.S.; Lin, C.H.; Tsai, C.H. Outcomes at School Age after Postnatal Dexamethasone Therapy for Lung Disease of Prematurity. N. Engl. J. Med. 2004, 350, 1304–1313. [Google Scholar] [CrossRef]
  11. Jensen, E.A.; Wiener, L.E.; Rysavy, M.A.; Dysart, K.C.; Gantz, M.G.; Eichenwald, E.C.; Greenberg, R.G.; Harmon, H.M.; Laughon, M.M.; Watterberg, K.L.; et al. Assessment of Corticosteroid Therapy and Death or Disability According to Pretreatment Risk of Death or Bronchopulmonary Dysplasia in Extremely Preterm Infants. JAMA Netw. Open 2023, 6, e2312277. [Google Scholar] [CrossRef] [PubMed]
  12. Hillman, N.H.; Jobe, A.H. Preterm Lung and Brain Responses to Mechanical Ventilation and Corticosteroids. J. Perinatol. 2023. [Google Scholar] [CrossRef] [PubMed]
  13. Homan, T.D.; Nayak, R.P. Short-and Long-Term Complications of Bronchopulmonary Dysplasia. Respir. Care 2021, 66, 1618–1629. [Google Scholar] [CrossRef]
  14. Shahzad, T.; Radajewski, S.; Chao, C.M.; Bellusci, S.; Ehrhardt, H. Pathogenesis of Bronchopulmonary Dysplasia: When Inflammation Meets Organ Development. Mol. Cell. Pediatr. 2016, 3, 23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Behnke, J.; Kremer, S.; Shahzad, T.; Chao, C.M.; Bottcher-Friebertshauser, E.; Morty, R.E.; Bellusci, S.; Ehrhardt, H. MSC Based Therapies-New Perspectives for the Injured Lung. J. Clin. Med. 2020, 9, 682. [Google Scholar] [CrossRef] [Green Version]
  16. Goetz, M.J.; Kremer, S.; Behnke, J.; Staude, B.; Shahzad, T.; Holzfurtner, L.; Chao, C.M.; Morty, R.E.; Bellusci, S.; Ehrhardt, H. MSC Based Therapies to Prevent or Treat BPD-A Narrative Review on Advances and Ongoing Challenges. Int. J. Mol. Sci. 2021, 22, 1138. [Google Scholar] [CrossRef]
  17. Jiang, W.; Xu, J. Immune Modulation by Mesenchymal Stem Cells. Cell Prolif. 2020, 53, e12712. [Google Scholar] [CrossRef] [Green Version]
  18. Popova, A.P.; Bozyk, P.D.; Bentley, J.K.; Linn, M.J.; Goldsmith, A.M.; Schumacher, R.E.; Weiner, G.M.; Filbrun, A.G.; Hershenson, M.B. Isolation of Tracheal Aspirate Mesenchymal Stromal Cells Predicts Bronchopulmonary Dysplasia. Pediatrics 2010, 126, e1127–e1133. [Google Scholar] [CrossRef] [Green Version]
  19. Lignelli, E.; Palumbo, F.; Myti, D.; Morty, R.E. Recent Advances in Our Understanding of the Mechanisms of Lung Alveolarization and Bronchopulmonary Dysplasia. Am. J. Physiol.-Lung Cell. Mol. Physiol. 2019, 317, L832–L887. [Google Scholar] [CrossRef]
  20. Alysandratos, K.-D.; Herriges, M.J.; Kotton, D.N. Epithelial Stem and Progenitor Cells in Lung Repair and Regeneration. Annu. Rev. Physiol. 2021, 83, 529–550. [Google Scholar] [CrossRef]
  21. Rock, J.R.; Randell, S.H.; Hogan, B.L.M. Airway Basal Stem Cells: A Perspective on Their Roles in Epithelial Homeostasis and Remodeling. Dis. Models Mech. 2010, 3, 545–556. [Google Scholar] [CrossRef] [Green Version]
  22. Hong, K.U.; Reynolds, S.D.; Watkins, S.; Fuchs, E.; Stripp, B.R. Basal Cells Are a Multipotent Progenitor Capable of Renewing the Bronchial Epithelium. Am. J. Pathol. 2004, 164, 577–588. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. 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] [Green Version]
  24. 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]
  25. Montoro, D.T.; Haber, A.L.; Biton, M.; Vinarsky, V.; Lin, B.; Birket, S.E.; Yuan, F.; Chen, S.; Leung, H.M.; Villoria, J.; et al. A Revised Airway Epithelial Hierarchy Includes CFTR-Expressing Ionocytes. Nature 2018, 560, 319–324. [Google Scholar] [CrossRef]
  26. Nakajima, M.; Kawanami, O.; Jin, E.; Ghazizadeh, M.; Honda, M.; Asano, G.; Horiba, K.; Ferrans, V.J. Immunohistochemical and Ultrastructural Studies of Basal Cells, Clara Cells and Bronchiolar Cuboidal Cells in Normal Human Airways. Pathol. Int. 1998, 48, 944–953. [Google Scholar] [CrossRef]
  27. Boers, J.E.; Ambergen, A.W.; Thunnissen, F.B. Number and Proliferation of Clara Cells in Normal Human Airway Epithelium. Am. J. Respir. Crit. Care Med. 1999, 159, 1585–1591. [Google Scholar] [CrossRef] [Green Version]
  28. Tata, P.R.; Mou, H.; Pardo-Saganta, A.; Zhao, R.; Prabhu, M.; Law, B.M.; Vinarsky, V.; Cho, J.L.; Breton, S.; Sahay, A.; et al. Dedifferentiation of Committed Epithelial Cells into Stem Cells in Vivo. Nature 2013, 503, 218–223. [Google Scholar] [CrossRef] [Green Version]
  29. 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] [Green Version]
  30. Watson, J.K.; Rulands, S.; Wilkinson, A.C.; Wuidart, A.; Ousset, M.; Van Keymeulen, A.; Göttgens, B.; Blanpain, C.; Simons, B.D.; Rawlins, E.L. Clonal Dynamics Reveal Two Distinct Populations of Basal Cells in Slow-Turnover Airway Epithelium. Cell Rep. 2015, 12, 90–101. [Google Scholar] [CrossRef] [Green Version]
  31. Teixeira, V.H.; Nadarajan, P.; Graham, T.A.; Pipinikas, C.P.; Brown, J.M.; Falzon, M.; Nye, E.; Poulsom, R.; Lawrence, D.; Wright, N.A.; et al. Stochastic Homeostasis in Human Airway Epithelium Is Achieved by Neutral Competition of Basal Cell Progenitors. eLife 2013, 2, e00966. [Google Scholar] [CrossRef]
  32. Plasschaert, L.W.; Zilionis, R.; Choo-Wing, R.; Savova, V.; Knehr, J.; Roma, G.; Klein, A.M.; Jaffe, A.B. A Single-Cell Atlas of the Airway Epithelium Reveals the CFTR-Rich Pulmonary Ionocyte. Nature 2018, 560, 377–381. [Google Scholar] [CrossRef] [PubMed]
  33. Carraro, G.; Mulay, A.; Yao, C.; Mizuno, T.; Konda, B.; Petrov, M.; Lafkas, D.; Arron, J.R.; Hogaboam, C.M.; Chen, P.; et al. Single-Cell Reconstruction of Human Basal Cell Diversity in Normal and Idiopathic Pulmonary Fibrosis Lungs. Am. J. Respir. Crit. Care Med. 2020, 202, 1540–1550. [Google Scholar] [CrossRef] [PubMed]
  34. Zaragosi, L.E.; Deprez, M.; Barbry, P. Using Single-Cell RNA Sequencing to Unravel Cell Lineage Relationships in the Respiratory Tract. Biochem. Soc. Trans. 2020, 48, 327–336. [Google Scholar] [CrossRef] [PubMed]
  35. Ruiz Garcia, S.; Deprez, M.; Lebrigand, K.; Cavard, A.; Paquet, A.; Arguel, M.J.; Magnone, V.; Truchi, M.; Caballero, I.; Leroy, S.; et al. Novel Dynamics of Human Mucociliary Differentiation Revealed by Single-Cell RNA Sequencing of Nasal Epithelial Cultures. Development 2019, 146, 177428. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Jaeger, B.; Schupp, J.C.; Plappert, L.; Terwolbeck, O.; Artysh, N.; Kayser, G.; Engelhard, P.; Adams, T.S.; Zweigerdt, R.; Kempf, H.; et al. Airway Basal Cells Show a Dedifferentiated KRT17highPhenotype and Promote Fibrosis in Idiopathic Pulmonary Fibrosis. Nat. Commun. 2022, 13, 5637. [Google Scholar] [CrossRef] [PubMed]
  37. Yang, Y.; Riccio, P.; Schotsaert, M.; Mori, M.; Lu, J.; Lee, D.K.; Garcia-Sastre, A.; Xu, J.; Cardoso, W. V Spatial-Temporal Lineage Restrictions of Embryonic P63(+) Progenitors Establish Distinct Stem Cell Pools in Adult Airways. Dev. Cell 2018, 44, 752–761.e4. [Google Scholar] [CrossRef] [Green Version]
  38. Shui, J.E.; Wang, W.; Liu, H.; Stepanova, A.; Liao, G.; Qian, J.; Ai, X.; Ten, V.; Lu, J.; Cardoso, W.V. Prematurity Alters the Progenitor Cell Program of the Upper Respiratory Tract of Neonates. Sci. Rep. 2021, 11, 10799. [Google Scholar] [CrossRef]
  39. Wang, H.; Liu, Y.; Liu, Z. Clara Cell 10-KD Protein in Inflammatory Upper Airway Diseases. Curr. Opin. Allergy Clin. Immunol. 2013, 13, 25–30. [Google Scholar] [CrossRef]
  40. Lee, R.M.; O’Brodovich, H. Airway Epithelial Damage in Premature Infants with Respiratory Failure. Am. Rev. Respir. Dis. 1988, 137, 450–457. [Google Scholar] [CrossRef]
  41. 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] [Green Version]
  42. Whitsett, J.A. Airway Epithelial Differentiation and Mucociliary Clearance. Ann. Am. Thorac. Soc. 2018, 15, S143–S148. [Google Scholar] [CrossRef] [PubMed]
  43. Lumsden, A.B.; McLean, A.; Lamb, D. Goblet and Clara Cells of Human Distal Airways: Evidence for Smoking Induced Changes in Their Numbers. Thorax 1984, 39, 844–849. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Vieira Braga, F.A.; Kar, G.; Berg, M.; Carpaij, O.A.; Polanski, K.; Simon, L.M.; Brouwer, S.; Gomes, T.; Hesse, L.; Jiang, J.; et al. A Cellular Census of Human Lungs Identifies Novel Cell States in Health and in Asthma. Nat. Med. 2019, 25, 1153–1163. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. De Water, R.; Willems, L.N.; Van Muijen, G.N.; Franken, C.; Fransen, J.A.; Dijkman, J.H.; Kramps, J.A. Ultrastructural Localization of Bronchial Antileukoprotease in Central and Peripheral Human Airways by a Gold-Labeling Technique Using Monoclonal Antibodies. Am. Rev. Respir. Dis. 1986, 133, 882–890. [Google Scholar] [PubMed]
  46. Barth, P.J.; Wolf, M.; Ramaswamy, A. Distribution and Number of Clara Cells in the Normal and Disturbed Development of the Human Fetal Lung. Pediatr. Pathol. 1994, 14, 637–651. [Google Scholar] [CrossRef] [PubMed]
  47. Thomas, W.; Seidenspinner, S.; Kawczyńska-Leda, N.; Chmielnicka-Kopaczyk, M.; Marx, A.; Wirbelauer, J.; Szymankiewicz, M.; Speer, C.P. Clara Cell Secretory Protein in Tracheobronchial Aspirates and Umbilical Cord Serum of Extremely Premature Infants with Systemic Inflammation. Neonatology 2010, 97, 228–234. [Google Scholar] [CrossRef]
  48. Ramsay, P.L.; DeMayo, F.J.; Hegemier, S.E.; Wearden, M.E.; Smith, C.V.; Welty, S.E. Clara Cell Secretory Protein Oxidation and Expression in Premature Infants Who Develop Bronchopulmonary Dysplasia. Am. J. Respir. Crit. Care Med. 2001, 164, 155–161. [Google Scholar] [CrossRef]
  49. Schilders, K.A.; Eenjes, E.; van Riet, S.; Poot, A.A.; Stamatialis, D.; Truckenmuller, R.; Hiemstra, P.S.; Rottier, R.J. Regeneration of the Lung: Lung Stem Cells and the Development of Lung Mimicking Devices. Respir. Res. 2016, 17, 44. [Google Scholar] [CrossRef] [Green Version]
  50. Evans, M.J.; Plopper, C.G. The Role of Basal Cells in Adhesion of Columnar Epithelium to Airway Basement Membrane. Am. Rev. Respir. Dis. 1988, 138, 481–483. [Google Scholar] [CrossRef] [PubMed]
  51. Hoyt, R.F., Jr.; Feldman, H.; Sorokin, S.P. Neuroepithelial Bodies (NEB) and Solitary Endocrine Cells in the Hamster Lung. Exp. Lung Res. 1982, 3, 299–311. [Google Scholar] [CrossRef] [PubMed]
  52. Kuo, C.S.; Krasnow, M.A. Formation of a Neurosensory Organ by Epithelial Cell Slithering. Cell 2015, 163, 394–405. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Weichselbaum, M.; Sparrow, M.P.; Hamilton, E.J.; Thompson, P.J.; Knight, D.A. A Confocal Microscopic Study of Solitary Pulmonary Neuroendocrine Cells in Human Airway Epithelium. Respir. Res. 2005, 6, 115. [Google Scholar] [CrossRef] [Green Version]
  54. Cutz, E.; Pan, J.; Yeger, H.; Domnik, N.J.; Fisher, J.T. Recent Advances and Contraversies on the Role of Pulmonary Neuroepithelial Bodies as Airway Sensors. Semin. Cell Dev. Biol. 2013, 24, 40–50. [Google Scholar] [CrossRef]
  55. Lee, R.J.; Kofonow, J.M.; Rosen, P.L.; Siebert, A.P.; Chen, B.; Doghramji, L.; Xiong, G.; Adappa, N.D.; Palmer, J.N.; Kennedy, D.W.; et al. Bitter and Sweet Taste Receptors Regulate Human Upper Respiratory Innate Immunity. J. Clin. Investig. 2014, 124, 1393–1405. [Google Scholar] [CrossRef]
  56. Branchfield, K.; Nantie, L.; Verheyden, J.M.; Sui, P.; Wienhold, M.D.; Sun, X. Pulmonary Neuroendocrine Cells Function as Airway Sensors to Control Lung Immune Response. Science 2016, 351, 707–710. [Google Scholar] [CrossRef] [Green Version]
  57. Johnson, D.E.; Lock, J.E.; Elde, R.P.; Thompson, T.R. Pulmonary Neuroendocrine Cells in Hyaline Membrane Disease and Bronchopulmonary Dysplasia. Pediatr. Res. 1982, 16, 446–454. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Johnson, D.E.; Kulik, T.J.; Lock, J.E.; Elde, R.P.; Thompson, T.R. Bombesin-, Calcitonin-, and Serotonin-Immunoreactive Pulmonary Neuroendocrine Cells in Acute and Chronic Neonatal Lung Disease. Pediatr. Pulmonol. 1985, 1, S13–S20. [Google Scholar]
  59. Cullen, A.; Van Marter, L.J.; Allred, E.N.; Moore, M.; Parad, R.B.; Sunday, M.E. Urine Bombesin-like Peptide Elevation Precedes Clinical Evidence of Bronchopulmonary Dysplasia. Am. J. Respir. Crit. Care Med. 2002, 165, 1093–1097. [Google Scholar] [CrossRef]
  60. Cutz, E.; Yeger, H.; Pan, J. Pulmonary Neuroendocrine Cell System in Pediatric Lung Disease-Recent Advances. Pediatr. Dev. Pathol. 2007, 10, 419–435. [Google Scholar] [CrossRef]
  61. Fujinaga, H.; Baker, C.D.; Ryan, S.L.; Markham, N.E.; Seedorf, G.J.; Balasubramaniam, V.; Abman, S.H. Hyperoxia Disrupts Vascular Endothelial Growth Factor-Nitric Oxide Signaling and Decreases Growth of Endothelial Colony-Forming Cells from Preterm Infants. Am. J. Physiol. Lung Cell. Mol. Physiol. 2009, 297, L1160–L1169. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. 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] [PubMed]
  63. Fehrenbach, H. Alveolar Epithelial Type II Cell: Defender of the Alveolus Revisited. Respir. Res. 2001, 2, 33–46. [Google Scholar] [CrossRef] [PubMed]
  64. Beers, M.F.; Moodley, Y. When Is an Alveolar Type 2 Cell an Alveolar Type 2 Cell? A Conundrum for Lung Stem Cell Biology and Regenerative Medicine. Am. J. Respir. Cell Mol. Biol. 2017, 57, 18–27. [Google Scholar] [CrossRef] [PubMed]
  65. 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]
  66. Nabhan, A.N.; Brownfield, D.G.; Harbury, P.B.; Krasnow, M.A.; Desai, T.J. Single-Cell Wnt Signaling Niches Maintain Stemness of Alveolar Type 2 Cells. Science 2018, 359, 1118–1123. [Google Scholar] [CrossRef] [Green Version]
  67. 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] [Green Version]
  68. Uhal, B.D. Cell Cycle Kinetics in the Alveolar Epithelium. Am. J. Physiol. 1997, 272, L1031–L1045. [Google Scholar] [CrossRef]
  69. Raman, T.; O’Connor, T.P.; Hackett, N.R.; Wang, W.; Harvey, B.G.; Attiyeh, M.A.; Dang, D.T.; Teater, M.; Crystal, R.G. Quality Control in Microarray Assessment of Gene Expression in Human Airway Epithelium. BMC Genom. 2009, 10, 493. [Google Scholar] [CrossRef] [Green Version]
  70. Maniscalco, W.M.; Watkins, R.H.; O’Reilly, M.A.; Shea, C.P. Increased Epithelial Cell Proliferation in Very Premature Baboons with Chronic Lung Disease. Am. J. Physiol. Lung. Cell. Mol. Physiol. 2002, 283, L991–L1001. [Google Scholar] [CrossRef] [Green Version]
  71. Abman, S.H.; Collaco, J.M.; Shepherd, E.G.; Keszler, M.; Cuevas-Guaman, M.; Welty, S.E.; Truog, W.E.; McGrath-Morrow, S.A.; Moore, P.E.; Rhein, L.M.; et al. Interdisciplinary Care of Children with Severe Bronchopulmonary Dysplasia. J. Pediatr. 2017, 181, 12–28.e1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Caskey, S.; Gough, A.; Rowan, S.; Gillespie, S.; Clarke, J.; Riley, M.; Megarry, J.; Nicholls, P.; Patterson, C.; Halliday, H.L.; et al. Structural and Functional Lung Impairment in Adult Survivors of Bronchopulmonary Dysplasia. Ann. Am. Thorac. Soc. 2016, 13, 1262–1270. [Google Scholar] [CrossRef] [Green Version]
  73. Thebaud, B.; Abman, S.H. Bronchopulmonary Dysplasia: Where Have All the Vessels Gone? Roles of Angiogenic Growth Factors in Chronic Lung Disease. Am. J. Respir. Crit. Care Med. 2007, 175, 978–985. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Angusamy, S.; Mansour, T.; Abdulmageed, M.; Han, R.; Schutte, B.C.; LaPres, J.; Harkema, J.R.; Omar, S.A. Altered Thymocyte and T Cell Development in Neonatal Mice with Hyperoxia-Induced Lung Injury. J. Perinat. Med. 2018, 46, 441–449. [Google Scholar] [CrossRef] [PubMed]
  75. Liu, S. Mouse Pneumonectomy Model of Compensatory Lung Growth. J. Vis. Exp. 2014, 94, e52294. [Google Scholar] [CrossRef] [Green Version]
  76. 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] [Green Version]
  77. Lechner, A.J.; Driver, I.H.; Lee, J.; Conroy, C.M.; Nagle, A.; Locksley, R.M.; Rock, J.R. Recruited Monocytes and Type 2 Immunity Promote Lung Regeneration Following Pneumonectomy. Cell Stem Cell 2017, 21, 120–134.e7. [Google Scholar] [CrossRef] [Green Version]
  78. Ahmadvand, N.; Khosravi, F.; Lingampally, A.; Wasnick, R.; Vazquez-Armendariz, I.; Carraro, G.; Heiner, M.; Rivetti, S.; Lv, Y.; Wilhelm, J.; et al. Identification of a Novel Subset of Alveolar Type 2 Cells Enriched in PD-L1 and Expanded Following Pneumonectomy. Eur. Respir. J. 2021, 58, 2004168. [Google Scholar] [CrossRef]
  79. Kalymbetova, T.V.; Selvakumar, B.; Rodríguez-Castillo, J.A.; Gunjak, M.; Malainou, C.; Heindl, M.R.; Moiseenko, A.; Chao, C.M.; Vadász, I.; Mayer, K.; et al. Resident Alveolar Macrophages Are Master Regulators of Arrested Alveolarization in Experimental Bronchopulmonary Dysplasia. J. Pathol. 2018, 245, 153–159. [Google Scholar] [CrossRef]
  80. Shafa, M.; Ionescu, L.I.; Vadivel, A.; Collins, J.J.P.; Xu, L.; Zhong, S.; Kang, M.; de Caen, G.; Daneshmand, M.; Shi, J.; et al. Human Induced Pluripotent Stem Cell–Derived Lung Progenitor and Alveolar Epithelial Cells Attenuate Hyperoxia-Induced Lung Injury. Cytotherapy 2018, 20, 108–125. [Google Scholar] [CrossRef]
  81. Choi, J.; Park, J.-E.; Tsagkogeorga, G.; Yanagita, M.; Koo, B.-K.; Han, N.; Lee, J.-H. Inflammatory Signals Induce AT2 Cell-Derived Damage-Associated Transient Progenitors That Mediate Alveolar Regeneration. Cell Stem Cell 2020, 27, 366–382.e7. [Google Scholar] [CrossRef] [PubMed]
  82. Lee, J.H.; Bhang, D.H.; Beede, A.; Huang, T.L.; Stripp, B.R.; Bloch, K.D.; Wagers, A.J.; Tseng, Y.H.; Ryeom, S.; Kim, C.F. Lung Stem Cell Differentiation in Mice Directed by Endothelial Cells via a BMP4-NFATc1-Thrombospondin-1 Axis. Cell 2014, 156, 440–455. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. 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] [Green Version]
  84. Chapman, H.A.; Li, X.; Alexander, J.P.; Brumwell, A.; Lorizio, W.; Tan, K.; Sonnenberg, A.; Wei, Y.; Vu, T.H. Integrin Alpha6beta4 Identifies an Adult Distal Lung Epithelial Population with Regenerative Potential in Mice. J. Clin. Investig. 2011, 121, 2855–2862. [Google Scholar] [CrossRef] [Green Version]
  85. Jain, R.; Barkauskas, C.E.; Takeda, N.; Bowie, E.J.; Aghajanian, H.; Wang, Q.; Padmanabhan, A.; Manderfield, L.J.; Gupta, M.; Li, D.; et al. Plasticity of Hopx(+) Type I Alveolar Cells to Regenerate Type II Cells in the Lung. Nat. Commun. 2015, 6, 6727. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Zuo, W.; Zhang, T.; Wu, D.Z.; Guan, S.P.; Liew, A.A.; Yamamoto, Y.; Wang, X.; Lim, S.J.; Vincent, M.; Lessard, M.; et al. P63(+)Krt5(+) Distal Airway Stem Cells Are Essential for Lung Regeneration. Nature 2015, 517, 616–620. [Google Scholar] [CrossRef]
  87. Vaughan, A.E.; Brumwell, A.N.; Xi, Y.; Gotts, J.E.; Brownfield, D.G.; Treutlein, B.; Tan, K.; Tan, V.; Liu, F.C.; Looney, M.R.; et al. Lineage-Negative Progenitors Mobilize to Regenerate Lung Epithelium after Major Injury. Nature 2015, 517, 621–625. [Google Scholar] [CrossRef] [Green Version]
  88. 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] [Green Version]
  89. Zhao, R.; Fallon, T.R.; Saladi, S.V.; Pardo-Saganta, A.; Villoria, J.; Mou, H.; Vinarsky, V.; Gonzalez-Celeiro, M.; Nunna, N.; Hariri, L.P.; et al. Yap Tunes Airway Epithelial Size and Architecture by Regulating the Identity, Maintenance, and Self-Renewal of Stem Cells. Dev. Cell 2014, 30, 151–165. [Google Scholar] [CrossRef] [Green Version]
  90. Mori, M.; Mahoney, J.E.; Stupnikov, M.R.; Paez-Cortez, J.R.; Szymaniak, A.D.; Varelas, X.; Herrick, D.B.; Schwob, J.; Zhang, H.; Cardoso, W. V Notch3-Jagged Signaling Controls the Pool of Undifferentiated Airway Progenitors. Development 2015, 142, 258–267. [Google Scholar] [CrossRef] [Green Version]
  91. Gao, X.; Bali, A.S.; Randell, S.H.; Hogan, B.L. GRHL2 Coordinates Regeneration of a Polarized Mucociliary Epithelium from Basal Stem Cells. J. Cell Biol. 2015, 211, 669–682. [Google Scholar] [CrossRef] [PubMed]
  92. 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] [Green Version]
  93. Shan, L.; Aster, J.C.; Sklar, J.; Sunday, M.E. Notch-1 Regulates Pulmonary Neuroendocrine Cell Differentiation in Cell Lines and in Transgenic Mice. Am. J. Physiol. Lung Cell. Mol. Physiol. 2007, 292, L500–L509. [Google Scholar] [CrossRef]
  94. Ito, T.; Udaka, N.; Yazawa, T.; Okudela, K.; Hayashi, H.; Sudo, T.; Guillemot, F.; Kageyama, R.; Kitamura, H. Basic Helix-Loop-Helix Transcription Factors Regulate the Neuroendocrine Differentiation of Fetal Mouse Pulmonary Epithelium. Development 2000, 127, 3913–3921. [Google Scholar] [CrossRef] [PubMed]
  95. Kumar, P.A.; Hu, Y.; Yamamoto, Y.; Hoe, N.B.; Wei, T.S.; Mu, D.; Sun, Y.; Joo, L.S.; Dagher, R.; Zielonka, E.M.; 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] [Green Version]
  96. 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]
  97. Linnoila, R.I. Functional Facets of the Pulmonary Neuroendocrine System. Lab. Investig. 2006, 86, 425–444. [Google Scholar] [CrossRef] [Green Version]
  98. 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] [Green Version]
  99. Noguchi, M.; Sumiyama, K.; Morimoto, M. Directed Migration of Pulmonary Neuroendocrine Cells toward Airway Branches Organizes the Stereotypic Location of Neuroepithelial Bodies. Cell Rep. 2015, 13, 2679–2686. [Google Scholar] [CrossRef] [Green Version]
  100. Yao, E.; Lin, C.; Wu, Q.; Zhang, K.; Song, H.; Chuang, P.T. Notch Signaling Controls Transdifferentiation of Pulmonary Neuroendocrine Cells in Response to Lung Injury. Stem Cells 2018, 36, 377–391. [Google Scholar] [CrossRef] [Green Version]
  101. Borges, M.; Linnoila, R.I.; Van De Velde, H.J.K.; Chen, H.; Nelkin, B.D.; Mabry, M.; Baylin, S.B.; Ball, D.W. An Achaete-Scute Homologue Essential for Neuroendocrine Differentiation in the Lung. Nature 1997, 386, 852–855. [Google Scholar] [CrossRef] [PubMed]
  102. Bender 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] [Green Version]
  103. 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] [Green Version]
  104. Parekh, K.R.; Nawroth, J.; Pai, A.; Busch, S.M.; Senger, C.N.; Ryan, A.L. Stem Cells and Lung Regeneration. Am. J. Physiol. Cell Physiol. 2020, 319, C675–C693. [Google Scholar] [CrossRef] [PubMed]
  105. Lynch, T.J.; Liu, X.; Wei, J.; Engelhardt, J.F. Stem Cell Niches in the Lung. In Lung Stem Cells in the Epithelium and Vasculature; Firth, A., Yuan, J.X.J., Eds.; Springer International Publishing: Cham, Switzeland, 2015; pp. 35–58. ISBN 978-3-319-16232-4. [Google Scholar]
  106. Kapere Ochieng, J.; Schilders, K.; Kool, H.; Buscop-van Kempen, M.; Boerema-De Munck, A.; Grosveld, F.; Wijnen, R.; Tibboel, D.; Rottier, R.J. Differentiated Type II Pneumocytes Can Be Reprogrammed by Ectopic Sox2 Expression. PLoS ONE 2014, 9, e107248. [Google Scholar] [CrossRef]
  107. Liu, Y.; Kumar, V.S.; Zhang, W.; Rehman, J.; Malik, A.B. Activation of Type II Cells into Regenerative Stem Cell Antigen-1(+) Cells during Alveolar Repair. Am. J. Respir. Cell Mol. Biol. 2015, 53, 113–124. [Google Scholar] [CrossRef] [Green Version]
  108. 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] [Green Version]
  109. Raiser, D.M.; Kim, C.F. Commentary: Sca-1 and Cells of the Lung: A Matter of Different Sorts. Stem Cells 2009, 27, 606–611. [Google Scholar] [CrossRef] [Green Version]
  110. Willis, G.R.; Fernandez-Gonzalez, A.; Anastas, J.; Vitali, S.H.; Liu, X.; Ericsson, M.; Kwong, A.; Mitsialis, S.A.; Kourembanas, S. Mesenchymal Stromal Cell Exosomes Ameliorate Experimental Bronchopulmonary Dysplasia and Restore Lung Function through Macrophage Immunomodulation. Am. J. Respir. Crit. Care Med. 2018, 197, 104–116. [Google Scholar] [CrossRef]
  111. Willis, G.R.; Reis, M.; Gheinani, A.H.; Fernandez-Gonzalez, A.; Taglauer, E.S.; Yeung, V.; Liu, X.; Ericsson, M.; Haas, E.; Mitsialis, S.A.; et al. Extracellular Vesicles Protect the Neonatal Lung from Hyperoxic Injury through the Epigenetic and Transcriptomic Reprogramming of Myeloid Cells. Am. J. Respir. Crit. Care Med. 2021, 204, 1418–1432. [Google Scholar] [CrossRef]
  112. Rani, S.; Ryan, A.E.; Griffin, M.D.; Ritter, T. Mesenchymal Stem Cell-Derived Extracellular Vesicles: Toward Cell-Free Therapeutic Applications. Mol. Ther. 2015, 23, 812–823. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Worthington, E.N.; Hagood, J.S. Therapeutic Use of Extracellular Vesicles for Acute and Chronic Lung Disease. Int. J. Mol. Sci. 2020, 21, 2318. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Gowen, A.; Shahjin, F.; Chand, S.; Odegaard, K.E.; Yelamanchili, S. V Mesenchymal Stem Cell-Derived Extracellular Vesicles: Challenges in Clinical Applications. Front. Cell Dev. Biol. 2020, 8, 149. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. He, X.; Dong, Z.; Cao, Y.; Wang, H.; Liu, S.; Liao, L.; Jin, Y.; Yuan, L.; Li, B. MSC-Derived Exosome Promotes M2 Polarization and Enhances Cutaneous Wound Healing. Stem Cells Int. 2019, 2019, 7132708. [Google Scholar] [CrossRef] [Green Version]
  116. Aguiar Koga, B.A.; Fernandes, L.A.; Fratini, P.; Sogayar, M.C.; Carreira, A.C.O. Role of MSC-Derived Small Extracellular Vesicles in Tissue Repair and Regeneration. Front. Cell Dev. Biol. 2022, 10, 1047094. [Google Scholar] [CrossRef] [PubMed]
  117. Karami Fath, M.; Moayedi Banan, Z.; Barati, R.; Mohammadrezakhani, O.; Ghaderi, A.; Hatami, A.; Ghiabi, S.; Zeidi, N.; Asgari, K.; Payandeh, Z.; et al. Recent Advancements to Engineer Mesenchymal Stem Cells and Their Extracellular Vesicles for Targeting and Destroying Tumors. Prog. Biophys. Mol. Biol. 2023, 178, 1–16. [Google Scholar] [CrossRef]
  118. Lu, Y.; Wang, L.; Zhang, M.; Chen, Z. Mesenchymal Stem Cell-Derived Small Extracellular Vesicles: A Novel Approach for Kidney Disease Treatment. Int. J. Nanomed. 2022, 17, 3603–3618. [Google Scholar] [CrossRef]
  119. Wu, Y.; Zhang, Z.; Li, J.; Zhong, H.; Yuan, R.; Deng, Z.; Wu, X. Mechanism of Adipose-Derived Mesenchymal Stem Cell-Derived Extracellular Vesicles Carrying MiR-21-5p in Hyperoxia-Induced Lung Injury. Stem Cell Rev. Rep. 2022, 18, 1007–1024. [Google Scholar] [CrossRef]
  120. Xi, Y.; Ju, R.; Wang, Y. Mesenchymal Stem Cell-Derived Extracellular Vesicles for the Treatment of Bronchopulmonary Dysplasia. Front. Pediatr. 2022, 10, 852034. [Google Scholar] [CrossRef]
  121. Ai, D.; Shen, J.; Sun, J.; Zhu, Z.; Gao, R.; Du, Y.; Yuan, L.; Chen, C.; Zhou, J. Mesenchymal Stem Cell-Derived Extracellular Vesicles Suppress Hyperoxia-Induced Transdifferentiation of Rat Alveolar Type 2 Epithelial Cells. Stem Cells Dev. 2022, 31, 53–66. [Google Scholar] [CrossRef]
  122. El Agha, E.; Bellusci, S. Walking along the Fibroblast Growth Factor 10 Route: A Key Pathway to Understand the Control and Regulation of Epithelial and Mesenchymal Cell-Lineage Formation during Lung Development and Repair after Injury. Scientifica 2014, 2014, 538379. [Google Scholar] [CrossRef]
  123. Al Alam, D.; El Agha, E.; Sakurai, R.; Kheirollahi, V.; Moiseenko, A.; Danopoulos, S.; Shrestha, A.; Schmoldt, C.; Quantius, J.; Herold, S.; et al. Evidence for the Involvement of Fibroblast Growth Factor 10 in Lipofibroblast Formation during Embryonic Lung Development. Development 2015, 142, 4139–4150. [Google Scholar] [CrossRef] [Green Version]
  124. McGowan, S.E. The Lipofibroblast: More than a Lipid-Storage Depot. Am. J. Physiol. Lung Cell. Mol. Physiol. 2019, 316, L869–L871. [Google Scholar] [CrossRef] [PubMed]
  125. Rehan, V.K.; Torday, J.S. The Lung Alveolar Lipofibroblast: An Evolutionary Strategy against Neonatal Hyperoxic Lung Injury. Antioxid. Redox Signal. 2014, 21, 1893–1904. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Gupte, V.V.; Ramasamy, S.K.; Reddy, R.; Lee, J.; Weinreb, P.H.; Violette, S.M.; Guenther, A.; Warburton, D.; Driscoll, B.; Minoo, P.; et al. Overexpression of Fibroblast Growth Factor-10 during Both Inflammatory and Fibrotic Phases Attenuates Bleomycin-Induced Pulmonary Fibrosis in Mice. Am. J. Respir. Crit. Care Med. 2009, 180, 424–436. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Yuan, T.; Volckaert, T.; Chanda, D.; Thannickal, V.J.; De Langhe, S.P. Fgf10 Signaling in Lung Development, Homeostasis, Disease, and Repair After Injury. Front. Genet. 2018, 9, 418. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Chao, C.M.; Moiseenko, A.; Kosanovic, D.; Rivetti, S.; El Agha, E.; Wilhelm, J.; Kampschulte, M.; Yahya, F.; Ehrhardt, H.; Zimmer, K.P.; et al. Impact of Fgf10 Deficiency on Pulmonary Vasculature Formation in a Mouse Model of Bronchopulmonary Dysplasia. Hum. Mol. Genet. 2019, 28, 1429–1444. [Google Scholar] [CrossRef]
  129. Tong, L.; Zhou, J.; Rong, L.; Seeley, E.J.; Pan, J.; Zhu, X.; Liu, J.; Wang, Q.; Tang, X.; Qu, J.; et al. Fibroblast Growth Factor-10 (FGF-10) Mobilizes Lung-Resident Mesenchymal Stem Cells and Protects against Acute Lung Injury. Sci. Rep. 2016, 6, 21642. [Google Scholar] [CrossRef] [Green Version]
  130. Taghizadeh, S.; Chao, C.-M.; Guenther, S.; Glaser, L.; Gersmann, L.; Michel, G.; Kraut, S.; Goth, K.; Koepke, J.; Heiner, M.; et al. FGF10 Triggers De Novo Alveologenesis in a Bronchopulmonary Dysplasia Model: Impact on Resident Mesenchymal Niche Cells. Stem Cells 2022, 40, 605–617. [Google Scholar] [CrossRef]
  131. Yu, P.; Wilhelm, K.; Dubrac, A.; Tung, J.K.; Alves, T.C.; Fang, J.S.; Xie, Y.; Zhu, J.; Chen, Z.; De Smet, F.; et al. FGF-Dependent Metabolic Control of Vascular Development. Nature 2017, 545, 224–228. [Google Scholar] [CrossRef] [Green Version]
  132. Khaki, M.; Salmanian, A.H.; Abtahi, H.; Ganji, A.; Mosayebi, G. Mesenchymal Stem Cells Differentiate to Endothelial Cells Using Recombinant Vascular Endothelial Growth Factor—A. Rep. Biochem. Mol. Biol. 2018, 6, 144–150. [Google Scholar]
  133. Ahn, S.Y.; Park, W.S.; Kim, Y.E.; Sung, D.K.; Sung, S.I.; Ahn, J.Y.; Chang, Y.S. Vascular Endothelial Growth Factor Mediates the Therapeutic Efficacy of Mesenchymal Stem Cell-Derived Extracellular Vesicles against Neonatal Hyperoxic Lung Injury. Exp. Mol. Med. 2018, 50, 1–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Kalikkot Thekkeveedu, R.; Guaman, M.C.; Shivanna, B. Bronchopulmonary Dysplasia: A Review of Pathogenesis and Pathophysiology. Respir. Med. 2017, 132, 170–177. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Munshi, U.K.; Niu, J.O.; Siddiq, M.M.; Parton, L.A. Elevation of Interleukin-8 and Interleukin-6 Precedes the Influx of Neutrophils in Tracheal Aspirates from Preterm Infants Who Develop Bronchopulmonary Dysplasia. Pediatr. Pulmonol. 1997, 24, 331–336. [Google Scholar] [CrossRef]
  136. Wright, C.J.; Kirpalani, H. Targeting Inflammation to Prevent Bronchopulmonary Dysplasia: Can New Insights Be Translated into Therapies? Pediatrics 2011, 128, 111–126. [Google Scholar] [CrossRef] [Green Version]
  137. Moodie, F.M.; Marwick, J.A.; Anderson, C.S.; Szulakowski, P.; Biswas, S.K.; Bauter, M.R.; Kilty, I.; Rahman, I. Oxidative Stress and Cigarette Smoke Alter Chromatin Remodeling but Differentially Regulate NF-KappaB Activation and Proinflammatory Cytokine Release in Alveolar Epithelial Cells. FASEB J. 2004, 18, 1897–1899. [Google Scholar] [CrossRef]
  138. Everhart, M.B.; Han, W.; Sherrill, T.P.; Arutiunov, M.; Polosukhin, V.V.; Burke, J.R.; Sadikot, R.T.; Christman, J.W.; Yull, F.E.; Blackwell, T.S. Duration and Intensity of NF-KappaB Activity Determine the Severity of Endotoxin-Induced Acute Lung Injury. J. Immunol. 2006, 176, 4995–5005. [Google Scholar] [CrossRef] [Green Version]
  139. Lawrence, T. The Nuclear Factor NF-KappaB Pathway in Inflammation. Cold Spring Harb. Perspect. Biol. 2009, 1, a001651. [Google Scholar] [CrossRef] [Green Version]
  140. Lawrence, T.; Fong, C. The Resolution of Inflammation: Anti-Inflammatory Roles for NF-KappaB. Int. J. Biochem. Cell Biol. 2010, 42, 519–523. [Google Scholar] [CrossRef]
  141. Barkett, M.; Gilmore, T.D. Control of Apoptosis by Rel/NF-KappaB Transcription Factors. Oncogene 1999, 18, 6910–6924. [Google Scholar] [CrossRef] [Green Version]
  142. Schmidt, D.; Textor, B.; Pein, O.T.; Licht, A.H.; Andrecht, S.; Sator-Schmitt, M.; Fusenig, N.E.; Angel, P.; Schorpp-Kistner, M. Critical Role for NF-KappaB-Induced JunB in VEGF Regulation and Tumor Angiogenesis. EMBO J. 2007, 26, 710–719. [Google Scholar] [CrossRef] [Green Version]
  143. Hinz, M.; Krappmann, D.; Eichten, A.; Heder, A.; Scheidereit, C.; Strauss, M. NF-KappaB Function in Growth Control: Regulation of Cyclin D1 Expression and G0/G1-to-S-Phase Transition. Mol. Cell. Biol. 1999, 19, 2690–2698. [Google Scholar] [CrossRef] [Green Version]
  144. Blackwell, T.S.; Hipps, A.N.; Yamamoto, Y.; Han, W.; Barham, W.J.; Ostrowski, M.C.; Yull, F.E.; Prince, L.S. NF-ΚB Signaling in Fetal Lung Macrophages Disrupts Airway Morphogenesis. J. Immunol. 2011, 187, 2740–2747. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Alvira, C.M.; Abate, A.; Yang, G.; Dennery, P.A.; Rabinovitch, M. Nuclear Factor-KappaB Activation in Neonatal Mouse Lung Protects against Lipopolysaccharide-Induced Inflammation. Am. J. Respir. Crit. Care Med. 2007, 175, 805–815. [Google Scholar] [CrossRef] [Green Version]
  146. Iosef, C.; Alastalo, T.P.; Hou, Y.; Chen, C.; Adams, E.S.; Lyu, S.C.; Cornfield, D.N.; Alvira, C.M. Inhibiting NF-KappaB in the Developing Lung Disrupts Angiogenesis and Alveolarization. Am. J. Physiol. Lung Cell. Mol. Physiol. 2012, 302, L1023–L1036. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Bourbia, A.; Cruz, M.A.; Rozycki, H.J. NF-KappaB in Tracheal Lavage Fluid from Intubated Premature Infants: Association with Inflammation, Oxygen, and Outcome. Arch. Dis. Child Fetal Neonatal Ed. 2006, 91, F36–F39. [Google Scholar] [CrossRef] [Green Version]
  148. Alvira, C.M. Nuclear Factor-Kappa-B Signaling in Lung Development and Disease: One Pathway, Numerous Functions. Birth Defects Res. Part A Clin. Mol. Teratol. 2014, 100, 202–216. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Ali, S.; Hirschfeld, A.F.; Mayer, M.L.; Fortuno, E.S., 3rd; Corbett, N.; Kaplan, M.; Wang, S.; Schneiderman, J.; Fjell, C.D.; Yan, J.; et al. Functional Genetic Variation in NFKBIA and Susceptibility to Childhood Asthma, Bronchiolitis, and Bronchopulmonary Dysplasia. J. Immunol. 2013, 190, 3949–3958. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  150. Reicherzer, T.; Häffner, S.; Shahzad, T.; Gronbach, J.; Mysliwietz, J.; Hübener, C.; Hasbargen, U.; Gertheiss, J.; Schulze, A.; Bellusci, S.; et al. Activation of the NF-KB Pathway Alters the Phenotype of MSCs in the Tracheal Aspirates of Preterm Infants with Severe BPD. Am. J. Physiol. Lung Cell. Mol. Physiol. 2018, 315, L87–L101. [Google Scholar] [CrossRef] [PubMed]
  151. Kaltschmidt, C.; Greiner, J.F.W.; Kaltschmidt, B. The Transcription Factor Nf-Κb in Stem Cells and Development. Cells 2021, 10, 2042. [Google Scholar] [CrossRef]
  152. Joel, M.D.M.; Yuan, J.; Wang, J.; Yan, Y.; Qian, H.; Zhang, X.; Xu, W.; Mao, F. MSC: Immunoregulatory Effects, Roles on Neutrophils and Evolving Clinical Potentials. Am. J. Transl. Res. 2019, 11, 3890. [Google Scholar] [PubMed]
  153. Prince, L.S. FGF10 and Human Lung Disease Across the Life Spectrum. Front. Genet. 2018, 9, 517. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Popova, A.P.; Bentley, J.K.; Cui, T.X.; Richardson, M.N.; Linn, M.J.; Lei, J.; Chen, Q.; Goldsmith, A.M.; Pryhuber, G.S.; Hershenson, M.B. Reduced Platelet-Derived Growth Factor Receptor Expression Is a Primary Feature of Human Bronchopulmonary Dysplasia. Am. J. Physiol. Lung Cell. Mol. Physiol. 2014, 307, L231–L239. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Pierro, M.; Ionescu, L.; Montemurro, T.; Vadivel, A.; Weissmann, G.; Oudit, G.; Emery, D.; Bodiga, S.; Eaton, F.; Peault, B.; et al. Short-Term, Long-Term and Paracrine Effect of Human Umbilical Cord-Derived Stem Cells in Lung Injury Prevention and Repair in Experimental Bronchopulmonary Dysplasia. Thorax 2013, 68, 475–484. [Google Scholar] [CrossRef] [Green Version]
  156. Chu, X.; Lingampally, A.; Moiseenko, A.; Kheirollahi, V.; Vazquez-Armendariz, A.I.; Koepke, J.; Khadim, A.; Kiliaris, G.; Shahriari Felordi, M.; Zabihi, M.; et al. GLI1+ Cells Are a Source of Repair-Supportive Mesenchymal Cells (RSMCs) during Airway Epithelial Regeneration. Cell. Mol. Life Sci. 2022, 79, 581. [Google Scholar] [CrossRef]
  157. Agostini, C. Stem Cell Therapy for Chronic Lung Diseases: Hope and Reality. Respir. Med. 2010, 104 (Suppl. 1), S86–S91. [Google Scholar] [CrossRef]
  158. Blaisdell, C.J.; Gail, D.B.; Nabel, E.G. National Heart, Lung, and Blood Institute Perspective: Lung Progenitor and Stem Cells—Gaps in Knowledge and Future Opportunities. Stem Cells 2009, 27, 2263–2270. [Google Scholar] [CrossRef] [Green Version]
  159. Sueblinvong, V.; Weiss, D.J. Stem Cells and Cell Therapy Approaches in Lung Biology and Diseases. Transl. Res. 2010, 156, 188–205. [Google Scholar] [CrossRef]
  160. Murphy, S.; Lim, R.; Dickinson, H.; Acharya, R.; Rosli, S.; Jenkin, G.; Wallace, E. Human Amnion Epithelial Cells Prevent Bleomycin-Induced Lung Injury and Preserve Lung Function. Cell Transplant. 2011, 20, 909–923. [Google Scholar] [CrossRef]
  161. Aguilar, S.; Scotton, C.J.; McNulty, 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]
  162. 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, 1855–1863. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Iyer, S.S.; Torres-Gonzalez, E.; Neujahr, D.C.; Kwon, M.; Brigham, K.L.; Jones, D.P.; Mora, A.L.; Rojas, M. Effect of Bone Marrow-Derived Mesenchymal Stem Cells on Endotoxin-Induced Oxidation of Plasma Cysteine and Glutathione in Mice. Stem Cells Int. 2010, 2010, 868076. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Tropea, K.A.; Leder, E.; Aslam, M.; Lau, A.N.; Raiser, D.M.; Lee, J.H.; Balasubramaniam, V.; Fredenburgh, L.E.; Alex Mitsialis, S.; Kourembanas, S.; et al. Bronchioalveolar Stem Cells Increase after Mesenchymal Stromal Cell Treatment in a Mouse Model of Bronchopulmonary Dysplasia. Am. J. Physiol. Lung Cell. Mol. Physiol. 2012, 302, L829–L837. [Google Scholar] [CrossRef] [PubMed]
  165. Aslam, M.; Baveja, R.; Liang, O.D.; Fernandez-Gonzalez, A.; Lee, C.; Mitsialis, S.A.; Kourembanas, S. Bone Marrow Stromal Cells Attenuate Lung Injury in a Murine Model of Neonatal Chronic Lung Disease. Am. J. Respir. Crit. Care Med. 2009, 180, 1122–1130. [Google Scholar] [CrossRef] [Green Version]
  166. Zhang, X.; Wang, H.; Shi, Y.; Peng, W.; Zhang, S.; Zhang, W.; Xu, J.; Mei, Y.; Feng, Z. Role of Bone Marrow-Derived Mesenchymal Stem Cells in the Prevention of Hyperoxia-Induced Lung Injury in Newborn Mice. Cell Biol. Int. 2012, 36, 589–594. [Google Scholar] [CrossRef]
  167. Hansmann, G.; Fernandez-Gonzalez, A.; Aslam, M.; Vitali, S.H.; Martin, T.; Mitsialis, S.A.; Kourembanas, S. Mesenchymal Stem Cell-Mediated Reversal of Bronchopulmonary Dysplasia and Associated Pulmonary Hypertension. Pulm. Circ. 2012, 2, 170–181. [Google Scholar] [CrossRef] [Green Version]
  168. Vosdoganes, P.; Hodges, R.J.; Lim, R.; Westover, A.J.; Acharya, R.Y.; Wallace, E.M.; Moss, T.J. Human Amnion Epithelial Cells as a Treatment for Inflammation-Induced Fetal Lung Injury in Sheep. Am. J. Obstet. Gynecol. 2011, 205, 156-e26. [Google Scholar] [CrossRef]
  169. Popova, A.P.; Bentley, J.K.; Anyanwu, A.C.; Richardson, M.N.; Linn, M.J.; Lei, J.; Wong, E.J.; Goldsmith, A.M.; Pryhuber, G.S.; Hershenson, M.B. Glycogen Synthase Kinase-3beta/Beta-Catenin Signaling Regulates Neonatal Lung Mesenchymal Stromal Cell Myofibroblastic Differentiation. Am. J. Physiol. Lung Cell. Mol. Physiol. 2012, 303, L439–L448. [Google Scholar] [CrossRef]
  170. Chang, Y.S.; Ahn, S.Y.; Yoo, H.S.; Sung, S.I.; Choi, S.J.; Oh, W.I.; Park, W.S. Mesenchymal Stem Cells for Bronchopulmonary Dysplasia: Phase 1 Dose-Escalation Clinical Trial. J. Pediatr. 2014, 164, 966–972.e6. [Google Scholar] [CrossRef]
  171. Lim, R.; Malhotra, A.; Tan, J.; Chan, S.T.; Lau, S.; Zhu, D.; Mockler, J.C.; Wallace, E.M. First-In-Human Administration of Allogeneic Amnion Cells in Premature Infants With Bronchopulmonary Dysplasia: A Safety Study. Stem Cells Transl. Med. 2018, 7, 628–635. [Google Scholar] [CrossRef] [Green Version]
  172. Malhotra, A.; Lim, R.; Mockler, J.C.; Wallace, E.M. Two-Year Outcomes of Infants Enrolled in the First-in-Human Study of Amnion Cells for Bronchopulmonary Dysplasia. Stem Cells Transl. Med. 2020, 9, 289–294. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Baker, E.K.; Malhotra, A.; Lim, R.; Jacobs, S.E.; Hooper, S.B.; Davis, P.G.; Wallace, E.M. Human Amnion Cells for the Prevention of Bronchopulmonary Dysplasia: A Protocol for a Phase I Dose Escalation Study. BMJ Open 2019, 9, e026265. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 2. Schematic representation of the regenerative potential of the lung stem/progenitor cells (A) MSCs support the stem cell niche under physiological conditions and upon injury; basal cells differentiate in the secretory and ciliated cells. Variant cells can contribute to the secretory cell pool, and the secretory cells can come from the BASCs and secretory cells themselves. BASCs can differentiate in ciliated cells as well. AT2 cells can differentiate from the BASCs, and they represent the primary source of AT1 cells, and additionally they can maintain themselves; MSCs contribute to suppressing inflammation, inhibiting immune cells, such as monocytes, neutrophils, and lymphocytes (decreased level of pro-inflammatory molecules, TNF-α, IL-6 e.g.,) (B) MSCs give rise to the entire mesenchymal compartment, including lipofibroblasts (LIFs) and myofibroblasts (MYFs), with the activation of the lipogenic or myogenic program that can contribute to the progression or the resolution of injuries.
Figure 2. Schematic representation of the regenerative potential of the lung stem/progenitor cells (A) MSCs support the stem cell niche under physiological conditions and upon injury; basal cells differentiate in the secretory and ciliated cells. Variant cells can contribute to the secretory cell pool, and the secretory cells can come from the BASCs and secretory cells themselves. BASCs can differentiate in ciliated cells as well. AT2 cells can differentiate from the BASCs, and they represent the primary source of AT1 cells, and additionally they can maintain themselves; MSCs contribute to suppressing inflammation, inhibiting immune cells, such as monocytes, neutrophils, and lymphocytes (decreased level of pro-inflammatory molecules, TNF-α, IL-6 e.g.,) (B) MSCs give rise to the entire mesenchymal compartment, including lipofibroblasts (LIFs) and myofibroblasts (MYFs), with the activation of the lipogenic or myogenic program that can contribute to the progression or the resolution of injuries.
Ijms 24 11229 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Marega, M.; El-Merhie, N.; Gökyildirim, M.Y.; Orth, V.; Bellusci, S.; Chao, C.-M. Stem/Progenitor Cells and Related Therapy in Bronchopulmonary Dysplasia. Int. J. Mol. Sci. 2023, 24, 11229. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms241311229

AMA Style

Marega M, El-Merhie N, Gökyildirim MY, Orth V, Bellusci S, Chao C-M. Stem/Progenitor Cells and Related Therapy in Bronchopulmonary Dysplasia. International Journal of Molecular Sciences. 2023; 24(13):11229. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms241311229

Chicago/Turabian Style

Marega, Manuela, Natalia El-Merhie, Mira Y. Gökyildirim, Valerie Orth, Saverio Bellusci, and Cho-Ming Chao. 2023. "Stem/Progenitor Cells and Related Therapy in Bronchopulmonary Dysplasia" International Journal of Molecular Sciences 24, no. 13: 11229. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms241311229

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