Bronchopulmonary dysplasia-associated pulmonary hypertension: Comparison
Please note this is a comparison between Version 2 by Conner Chen and Version 1 by Saverio Bellusci.

       Bronchopulmonary dysplasia (BPD) is a chronic lung disease affecting preterm infants. More than 50 years after the first description of BPD by Northway, this chronic lung disease affecting many preterm infants is still less understood. Additonally, approximately 40% of preterm infants suffering from severe BPD also suffer from Bronchopulmonary dysplasia-associated pulmonary hypertension (BPD-PH), leading to a significant increase in total morbidity and mortality. Until today, there is no curative therapy for both BPD and BPD-PH available. It has become increasingly evident that growth factors are playing a central role in normal and pathologic development of the pulmonary vasculature. Thus, this review aims to summarize the recent evidence in our understanding of BPD-PH from a basic scientific point of view, focusing on the potential role of Fibroblast Growth Factor (FGF)/FGF10 signaling pathway contributing to disease development, progression and resolution.

  • Bronchopulmonary dysplasia
  • Pulmonary hypertension
  • Fgf signaling

Dear author, the following contents are excerpts from your papers. They are editable.

(Due to the lack of relevant professional knowledge, our editors cannot complete a complete entry by summarizing your paper, so if you are interested in this work. you may need to write some contents by yourself. A good entry will better present your ideas, research and results to other scholars. Readers will also be able to access your paper directly through entries.)

1. Introduction

There is a rising incidence of preterm births which is one of the leading cause of death in infants younger than 5 years of age [1,2]. Bronchopulmonary dysplasia (BPD) is a common complication for infants born before 30 weeks of gestational age. The incidence of BPD is approximately 40%. BPD contributes substantially to long-term morbidities and mortalities [3]. Currently, there is no curative therapy for BPD.

There is a rising incidence of preterm births which is one of the leading cause of death in infants younger than 5 years of age [1][2]. Bronchopulmonary dysplasia (BPD) is a common complication for infants born before 30 weeks of gestational age. The incidence of BPD is approximately 40%. BPD contributes substantially to long-term morbidities and mortalities [3]. Currently, there is no curative therapy for BPD.

BPD is a complex chronic lung disease caused by the interplay of lung development and pre- and postnatal injurious events such as ventilatory damages, oxygen toxicity, infectious stimuli, growth restriction, and repair/remodeling processes. Due to advances in the clinical management of preterm infants (e.g., use of exogenous surfactant, antenatal administration of steroids, more gentle non-invasive ventilation, and nutritional strategies in the clinical management of preterm infants) the mortality has reduced but the proportion of very early preterm infants has increased. During the so-called post-surfactant era, remarkable changes regarding the histomorphology of fatal BPD have been observed. The ”new“ BPD is characterized by simplification of alveolar formation and arrested microvascular development as compared with fibroproliferative processes mostly of the airways in ”old“ BPD. Whether these histomorphological changes result from different windows of injury (very early stages versus late stages of lung development) or from different treatment approaches (e.g., harsh mechanical ventilation with high oxygen versus non-invasive ventilation) is still unclear.

Histomorphologically, BPD is mainly characterized as alveolar simplification and pulmonary vascular remodeling. Pre- and postnatal pulmonary inflammatory responses (e.g., chorioamnionitis) are central risk factors for developing BPD. Pulmonary inflammatory responses can be induced by oxygen therapy or mechanical ventilation leading to the imbalance of proinflammatory and anti-inflammatory cytokines. This is associated with an accumulation of inflammatory cells in the lung. For example, it has been demonstrated that interleukin 10 (IL-10) and growth factors such as vascular endothelial growth factor alpha (VEGFA), platelet derived growth factor alpha (PDGFA), and fibroblast growth factors (FGFs) are decreased in tracheal aspirates and lung tissue [4,5,6].

Histomorphologically, BPD is mainly characterized as alveolar simplification and pulmonary vascular remodeling. Pre- and postnatal pulmonary inflammatory responses (e.g., chorioamnionitis) are central risk factors for developing BPD. Pulmonary inflammatory responses can be induced by oxygen therapy or mechanical ventilation leading to the imbalance of proinflammatory and anti-inflammatory cytokines. This is associated with an accumulation of inflammatory cells in the lung. For example, it has been demonstrated that interleukin 10 (IL-10) and growth factors such as vascular endothelial growth factor alpha (VEGFA), platelet derived growth factor alpha (PDGFA), and fibroblast growth factors (FGFs) are decreased in tracheal aspirates and lung tissue [4][5][6].

Pulmonary hypertension (PH) is a common and life-threatening complication in BPD. A recent meta-analysis including 1400 preterm infants revealed that the prevalence of PH increases correspondingly with the severity of BPD. The prevalence is estimated to be approximately 40% in preterm infants suffering from severe BPD [32,33].

Pulmonary hypertension (PH) is a common and life-threatening complication in BPD. A recent meta-analysis including 1400 preterm infants revealed that the prevalence of PH increases correspondingly with the severity of BPD. The prevalence is estimated to be approximately 40% in preterm infants suffering from severe BPD [7][8].

Pulmonary vascular remodeling (VR) underlying PH leads to (I) increased muscularization of already muscularized proximal and middle-sized vessels due to proliferation of existing vascular smooth muscle cells (VSMCs) and (II) 

de novo muscularization of normally non-muscularized distal vessels. The latter are typically the distal arterioles with diameters under 30 µm. VR results in increased pressure in the pulmonary circulation leading to an increased right ventricle (RV) afterload and potentially fatal RV hypertrophy (RVH) [34,35,36]. It needs to be mentioned that the cellular origin of the newly formed VSMCs in BPD is still elusive. The complications associated with BPD without PH includes a higher risk for developing susceptibility to infections of the upper and lower respiratory tract and asthma. By contrast, infants suffering from BPD-PH are of higher risk for oxygen dependency and right ventricle hypertrophy, which requires long-term medication and follow-up. In the worst case, these additional complications can lead to right heart failure. The lifelong morbidity of patients suffering from BPD-PH represents a substantial and growing burden for patients and health care systems [37,38].

muscularization of normally non-muscularized distal vessels. The latter are typically the distal arterioles with diameters under 30 µm. VR results in increased pressure in the pulmonary circulation leading to an increased right ventricle (RV) afterload and potentially fatal RV hypertrophy (RVH) [9][10][11]. It needs to be mentioned that the cellular origin of the newly formed VSMCs in BPD is still elusive. The complications associated with BPD without PH includes a higher risk for developing susceptibility to infections of the upper and lower respiratory tract and asthma. By contrast, infants suffering from BPD-PH are of higher risk for oxygen dependency and right ventricle hypertrophy, which requires long-term medication and follow-up. In the worst case, these additional complications can lead to right heart failure. The lifelong morbidity of patients suffering from BPD-PH represents a substantial and growing burden for patients and health care systems [12][13].

Although recent guidelines published by the American Heart Association (AHA), American Thoracic Society (ATS), the European Pediatric Pulmonary Vascular Disease Network (PVD), and the Pediatric Pulmonary Hypertension Network (PPHNet) encourage and improve the implementation of standardized diagnostic and treatment protocols, the molecular and cellular pathomechanisms underlying BPD-associated PH remain poorly characterized, reflected by the rather limited pharmacotherapeutic options available to mitigate but not cure BPD-PH [39,40,41]

Although recent guidelines published by the American Heart Association (AHA), American Thoracic Society (ATS), the European Pediatric Pulmonary Vascular Disease Network (PVD), and the Pediatric Pulmonary Hypertension Network (PPHNet) encourage and improve the implementation of standardized diagnostic and treatment protocols, the molecular and cellular pathomechanisms underlying BPD-associated PH remain poorly characterized, reflected by the rather limited pharmacotherapeutic options available to mitigate but not cure BPD-PH [14][15][16]

2. Development of Normal Pulmonary Vasculature and BPD-PH from the Basic Scientific Point-of-View

According to the Fifth World Symposium held in France, PH is clinically classified into five groups (Group 1, pulmonary arterial hypertension (PAH); Group 2, pulmonary hypertension due to left heart disease; Group 3, pulmonary hypertension due to chronic lung disease or hypoxia; Group 4, chronic thromboembolic pulmonary hypertension; and Group 5, pulmonary hypertension due to unclear multifactorial mechanisms) [42,43]. Groups 1, 3 and 4 are all defined as precapillary pulmonary hypertension [44]. Therefore, they share similar molecular and cellular mechanisms and histopathology. Pediatric PH has some common features with adult PH but also shows its own features. BPD-PH, characterized by impaired alveolar growth and distorted pulmonary vascular development, is categorized as a subgroup of Group 3 (Group 3.5, developmental lung disorders) [45]. Therefore, in the next section, we describe the different lung vascular growth abnormalities and the potential reasons of BPD-PH from the developmental angle.

According to the Fifth World Symposium held in France, PH is clinically classified into five groups (Group 1, pulmonary arterial hypertension (PAH); Group 2, pulmonary hypertension due to left heart disease; Group 3, pulmonary hypertension due to chronic lung disease or hypoxia; Group 4, chronic thromboembolic pulmonary hypertension; and Group 5, pulmonary hypertension due to unclear multifactorial mechanisms) [17][18]. Groups 1, 3 and 4 are all defined as precapillary pulmonary hypertension [19]. Therefore, they share similar molecular and cellular mechanisms and histopathology. Pediatric PH has some common features with adult PH but also shows its own features. BPD-PH, characterized by impaired alveolar growth and distorted pulmonary vascular development, is categorized as a subgroup of Group 3 (Group 3.5, developmental lung disorders) [20]. Therefore, in the next section, we describe the different lung vascular growth abnormalities and the potential reasons of BPD-PH from the developmental angle.

2.1. Normal Development of Pulmonary Vasculature

2.1. Normal Development of Pulmonary Vasculature

2.1.1. Lung Vasculature Formation and Maturation

Human lung development can be histologically divided into four distinguishable stages: embryonic and pseudoglandular stages (human, week 4–17 and mouse, E9.5–E16.5), canalicular stage (human, week 17–26 and mouse, E16.5–E17.5), saccular stage (human, week 26–36 and mouse, E17.5–PN5), and alveolar stage (human, week 36–8 years and mouse, PN5–PN30) [21][22].

The vasculature starts to develop as early as the lung buds evaginate from the foregut endoderm [23]. At E10, angioblasts and hematopoietic cells, localized at the distal mouse lung buds, form the blood islands through a processed termed vasculogenesis. The blood islands are comprised of circumferential layers of flattened angioblasts which represent primitive endothelial cells (ECs) and inner hematopoietic cells [24]. From E11 to E12, proximal vessels sprout from the main pulmonary trunk (proximal angiogenesis), run along the main conducting airways, as well as its many ramified branches. In addition, the distal blood islands increase markedly and are connected together to form the primitive capillary plexus (distal angiogenesis) [21]. During the late pseudoglandular stage, the capillary network, around lung buds, fuses with the proximal vessels. This capillary network expands significantly during the canalicular and saccular stages and gradually gets close to each other to become a double capillary layer between the adjacent lateral walls of distal sacs which form during the process of primary septa formation. During alveologenesis, concomitant with the formation of the secondary septa, the capillary network folds up to form new double capillary layers. These double capillary layers further evolve into a single capillary layer through a process called microvascular maturation. This process reduces the distance between the alveolar walls and the capillaries, thereby facilitating efficient gas exchange [25][26]. Microvascular maturation occurs concomitantly to alveologenesis and, in humans, lasts until young adulthood [27].

2.1.1. Lung Vasculature Formation and Maturation

Human lung development can be histologically divided into four distinguishable stages: embryonic and pseudoglandular stages (human, week 4–17 and mouse, E9.5–E16.5), canalicular stage (human, week 17–26 and mouse, E16.5–E17.5), saccular stage (human, week 26–36 and mouse, E17.5–PN5), and alveolar stage (human, week 36–8 years and mouse, PN5–PN30) [46,47].
The vasculature starts to develop as early as the lung buds evaginate from the foregut endoderm [48]. At E10, angioblasts and hematopoietic cells, localized at the distal mouse lung buds, form the blood islands through a processed termed vasculogenesis. The blood islands are comprised of circumferential layers of flattened angioblasts which represent primitive endothelial cells (ECs) and inner hematopoietic cells [49]. From E11 to E12, proximal vessels sprout from the main pulmonary trunk (proximal angiogenesis), run along the main conducting airways, as well as its many ramified branches. In addition, the distal blood islands increase markedly and are connected together to form the primitive capillary plexus (distal angiogenesis) [46]. During the late pseudoglandular stage, the capillary network, around lung buds, fuses with the proximal vessels. This capillary network expands significantly during the canalicular and saccular stages and gradually gets close to each other to become a double capillary layer between the adjacent lateral walls of distal sacs which form during the process of primary septa formation. During alveologenesis, concomitant with the formation of the secondary septa, the capillary network folds up to form new double capillary layers. These double capillary layers further evolve into a single capillary layer through a process called microvascular maturation. This process reduces the distance between the alveolar walls and the capillaries, thereby facilitating efficient gas exchange [50,51]. Microvascular maturation occurs concomitantly to alveologenesis and, in humans, lasts until young adulthood [52].

2.1.2. Pulmonary Vascular Development Needs Multicellular Crosstalks

Interactions among different cell types are indispensable for the formation and maturation of the vascular system during lung development. At the early stage of lung development, mesenchymal cells express Vegf, which interacts with its receptors, Vegfr1 (Flt-1), Vegfr2 (Flk-1), and Vegfr3 on endothelial cells (ECs), initiating and regulating the formation of vasculogenesis and angiogenesis [53,54]. Vegfr2 is the most important signaling receptor, which upon activation, initiates and promotes vasculogenesis and angiogenesis [55]. In contrast, Vegfr1 functions as a ligand trap, therefore, reducing the interaction between Vegf and Vegfr2, thereby decreasing the overexpansion of ECs [56]. From the late pseudoglandular stage to the canalicular stage, the cellular source of Vegf gradually shifts from the resident mesenchymal cells to the alveolar epithelial cells (mainly alveolar type 2 (AT2) cells). This results in further attraction of the capillaries towards the epithelium, thereby promoting the formation of primitive alveolar septa through EC-derived angiocrine factors, such as hepatocyte growth factor (Hgf) [57,58,59].

Interactions among different cell types are indispensable for the formation and maturation of the vascular system during lung development. At the early stage of lung development, mesenchymal cells express Vegf, which interacts with its receptors, Vegfr1 (Flt-1), Vegfr2 (Flk-1), and Vegfr3 on endothelial cells (ECs), initiating and regulating the formation of vasculogenesis and angiogenesis [28][29]. Vegfr2 is the most important signaling receptor, which upon activation, initiates and promotes vasculogenesis and angiogenesis [30]. In contrast, Vegfr1 functions as a ligand trap, therefore, reducing the interaction between Vegf and Vegfr2, thereby decreasing the overexpansion of ECs [31]. From the late pseudoglandular stage to the canalicular stage, the cellular source of Vegf gradually shifts from the resident mesenchymal cells to the alveolar epithelial cells (mainly alveolar type 2 (AT2) cells). This results in further attraction of the capillaries towards the epithelium, thereby promoting the formation of primitive alveolar septa through EC-derived angiocrine factors, such as hepatocyte growth factor (Hgf) [32][33][34].

Following the interaction between epithelium and mesenchyme, a population of mesenchymal cells differentiate into α-smooth muscle actin (Sma)-positive mural cells (VSMCs and pericytes), another component of the blood vessel wall [60]. VSMCs, which display a flattened, spindle, and dense structure, are usually present in the media of large vessels such as arteries, arterioles, and veins. From large vessels to capillaries, VSMCs are gradually getting sparse and changing into round, protruded cells, termed pericytes. They always embed in the basement membrane and adhere tightly to ECs [61,62]. (

Following the interaction between epithelium and mesenchyme, a population of mesenchymal cells differentiate into α-smooth muscle actin (Sma)-positive mural cells (VSMCs and pericytes), another component of the blood vessel wall [35]. VSMCs, which display a flattened, spindle, and dense structure, are usually present in the media of large vessels such as arteries, arterioles, and veins. From large vessels to capillaries, VSMCs are gradually getting sparse and changing into round, protruded cells, termed pericytes. They always embed in the basement membrane and adhere tightly to ECs [36][37]. (

Figure 1). In contrast to VSMCs, several markers including neuron-glial antigen-2 (Ng2), Cd146, α-Sma, Sm22, desmin, platelet-derived growth factor receptor-β (Pdgfr-β), aminopeptidase A and N, RGS5, and the promoter trap transgene XlacZ4 have been identified to label pericytes [62,63,64,65].

). In contrast to VSMCs, several markers including neuron-glial antigen-2 (Ng2), Cd146, α-Sma, Sm22, desmin, platelet-derived growth factor receptor-β (Pdgfr-β), aminopeptidase A and N, RGS5, and the promoter trap transgene XlacZ4 have been identified to label pericytes [38][39][40][41].

Figure 1.

 A speculative model of structural changes of the pulmonary vessel wall in bronchopulmonary dysplasia-associated pulmonary hypertension (BPD-PH). Under a normal situation, pulmonary arterioles are wrapped by the following three layers of tunicae: The tunica intima consists of endothelium, basement membrane, and internal elastic tissue; the tunica media is comprised of smooth muscle cells and external elastic tissue; and connective tissues constitute the tunica adventitia. From large vessels to small vessels, tunica media and adventitia are gradually getting sparse and the smooth muscle cells gradually evolving into pericytes, which adhere tightly to capillary endothelial cells (ECs). While in BPD, due to multifactorial injuries, the endothelial cells are dysfunctional and eventually become apoptosis resistant. Smooth muscle cells, which mostly proliferated from resident smooth muscle cells (SMCs) are increased significantly, resulting in the thickening of tunica media and the muscularization of normally non-muscular vessels. Adventitia of pulmonary arterioles undergo α-Sma positive myofibroblast transition assisted by altered extracellular matrix breakdown and deposition. Pericytes disconnect with capillary ECs, leading to the loss of small capillaries, proliferate, and migrate into the mesenchyme and also contribute to a small population of smooth muscle-like cells.

Pericytes play a critical role in regulating microvascular and alveolar development. Pericytes, on the one hand, adhere to capillary ECs to regulate their function, proliferation, migration, and differentiation via several signaling pathways including Vegf/Vegfr, Pdgfb/Pdgfrb2, Tgfβ/Alk, S1p/Endoglin 1(Edg-1), Ang1/2/Tie2, Cadherin, and Notch signaling pathways [51,66,67,68,69,70]. On the other hand, pericytes also interact with AT2 cells to promote secondary septa formation through the Hippo pathway components Yap and Taz [71]. In addition, pericytes can also function as pulmonary stem cells, upon stimulation, transdifferentiating into VSMCs and myofibroblasts [72,73].

Pericytes play a critical role in regulating microvascular and alveolar development. Pericytes, on the one hand, adhere to capillary ECs to regulate their function, proliferation, migration, and differentiation via several signaling pathways including Vegf/Vegfr, Pdgfb/Pdgfrb2, Tgfβ/Alk, S1p/Endoglin 1(Edg-1), Ang1/2/Tie2, Cadherin, and Notch signaling pathways [42][43][44][45][46]. On the other hand, pericytes also interact with AT2 cells to promote secondary septa formation through the Hippo pathway components Yap and Taz [47]. In addition, pericytes can also function as pulmonary stem cells, upon stimulation, transdifferentiating into VSMCs and myofibroblasts [48][49].

Interactions between endothelial cells (ECs) and smooth muscle cells (SMCs) play a major role in pulmonary vasculature. Normal interplay among these two cell types control the homeostasis of the pulmonary circulation, whereas aberrant association can contribute to the pulmonary vascular diseases and pulmonary hypertension. It has been well established that the release of various vasoactive agents such as nitric oxide (NO) and endothelin-1 (Et−1) through paracrine signaling endothelial cells regulates smooth muscle cell activity. However, other non-paracrine signaling mediated crosstalk between EC and SMC (such as communication through myoendothelial junctions, as well as interaction via extracellular vesicles) exists, which is altered under the pathological condition, thus, causing an increase in vasocontractility and abnormal vascular proliferation, therefore, leading to vascular remodeling, right ventricular hypertrophy, and pulmonary hypertension [74].

Interactions between endothelial cells (ECs) and smooth muscle cells (SMCs) play a major role in pulmonary vasculature. Normal interplay among these two cell types control the homeostasis of the pulmonary circulation, whereas aberrant association can contribute to the pulmonary vascular diseases and pulmonary hypertension. It has been well established that the release of various vasoactive agents such as nitric oxide (NO) and endothelin-1 (Et−1) through paracrine signaling endothelial cells regulates smooth muscle cell activity. However, other non-paracrine signaling mediated crosstalk between EC and SMC (such as communication through myoendothelial junctions, as well as interaction via extracellular vesicles) exists, which is altered under the pathological condition, thus, causing an increase in vasocontractility and abnormal vascular proliferation, therefore, leading to vascular remodeling, right ventricular hypertrophy, and pulmonary hypertension [50].

2.2. Pathologic Development of Pulmonary Vasculature Leading to PH in BPD

2.2.1. Abnormalities of Pulmonary Vasculature Observed in BPD-PH

Impaired pulmonary vascular development, VR and PH are likely associated with interruption of vascular formation during lung development. However, not all premature infants develop BPD or BPD-PH. It has been claimed that genetic components (e.g., polymorphisms in matrix metallopeptidase 16 (

MMP16

) and SPARC (Osteonectin) Cwcv and Kazal like domains proteoglycan 2 (

SPOCK2) could play a role regarding the susceptibility for BPD [75].

) could play a role regarding the susceptibility for BPD [51][52].

We previously found that conditional deletion of phosphatase and tensin homologue (

Pten) in early embryonic mouse lung mesenchyme led to lethality at birth with disorganized alveolar capillary beds [76]. This phenotype is similar to the lethal alveolar capillary dysplasia phenotype observed in newborn babies. In support of developmental perturbations, BPD often occurs in preterm infants born before the alveolar stage, and thus susceptible to various injuries such as mechanical ventilation, hyperoxia, and airway inflammation. These, in turn, affect pulmonary vascular development, leading to vascular remodeling, PH, and impaired alveologenesis [77,78].

) in early embryonic mouse lung mesenchyme led to lethality at birth with disorganized alveolar capillary beds [53]. This phenotype is similar to the lethal alveolar capillary dysplasia phenotype observed in newborn babies. In support of developmental perturbations, BPD often occurs in preterm infants born before the alveolar stage, and thus susceptible to various injuries such as mechanical ventilation, hyperoxia, and airway inflammation. These, in turn, affect pulmonary vascular development, leading to vascular remodeling, PH, and impaired alveologenesis .

Bhatt et al. analyzed lung samples from infants who died with BPD versus infants who died from non-pulmonary causes and found a decreased expression of VEGF and platelet endothelial cell adhesion molecule-1 (PECAM, also termed CD31, endothelial marker), as well as a decreased staining density of alveolar capillaries in BPD infants, indicating that the development of the pulmonary microvasculature was disrupted in BPD patients [

4]. Consistent with previous results, we and others, utilizing BPD animal models, established by hyperoxia exposure, also showed a decrease of endothelial cells in capillaries and blood vessel numbers and an increase of α-Sma positive cells (VSMC) in the tunica media of pulmonary arterioles and normally non-muscularized precapillary arterioles [79,80,81]. A lineage tracing study indicated that the expansion of resident SMCs was the major source related to the thickening of the smooth muscle layer in adult PH [82]. An increased collagen and elastin expression associated with increased α-SMA-positive myofibroblasts, possibly due to endothelial to mesenchymal transition (EndMT), has also been proposed as a mechanism for the accumulation of myofibroblats in the adventitia of pulmonary arteries (

]. Consistent with previous results, we and others, utilizing BPD animal models, established by hyperoxia exposure, also showed a decrease of endothelial cells in capillaries and blood vessel numbers and an increase of α-Sma positive cells (VSMC) in the tunica media of pulmonary arterioles and normally non-muscularized precapillary arterioles [54][55][56]. A lineage tracing study indicated that the expansion of resident SMCs was the major source related to the thickening of the smooth muscle layer in adult PH [57]. An increased collagen and elastin expression associated with increased α-SMA-positive myofibroblasts, possibly due to endothelial to mesenchymal transition (EndMT), has also been proposed as a mechanism for the accumulation of myofibroblats in the adventitia of pulmonary arteries (

Figure 2) [83,84].

) [58][59].

Figure 2.

 Possible endothelial–mesenchymal and epithelial–mesenchymal interactions in BPD-PH. Disrupted endothelial Apelin, 

miR-503

, and 

miR-424

 results in an increased expression of FGF2 and induces the hyperproliferation of vessel smooth muscle cells (VSMC). An increase of α-SMA-positive myofibroblasts could be due to endothelial-mesenchymal transition (EndMT). The continuous high expression of 

miR-154

 in AT2 cells stimulate the activation of TGFβ1 signaling pathway, which results in an impaired alveologenesis. Decreased FGF10 expression in BPD, which is caused by the upregulation of 

miR-421

, SHH, and Spry2, leads to the downregulation of VEGFA and upregulation of α-SMA, ultimately resulting in an impaired angiogenesis and an increase of α-SMA positive cells. However, whether FGF10 acts directly on the mesenchymal stem cells (MSC) to induce them to differentiate into α-SMA positive cells or through activating TGFβ1 signaling pathway needs to be further investigated.

In addition, it has also been proposed that the pericytes play a potential role in the microvascular remodeling of PH. In a clinical study, Assaad et al. found that the numbers of pericytes associated with upregulated PDGF-B expression were significantly increased in pulmonary capillary hemangiomatosis (PCH) patients, a cause of PH [85]. Furthermore, Ricard et al. also showed an increased migration and proliferation of pericytes induced by FGF2 and IL-2, in human PH patients. Finally, increased TGFβ also promotes the transdifferentiation of pericytes into contractile α-SMA positive cells (

In addition, it has also been proposed that the pericytes play a potential role in the microvascular remodeling of PH. In a clinical study, Assaad et al. found that the numbers of pericytes associated with upregulated PDGF-B expression were significantly increased in pulmonary capillary hemangiomatosis (PCH) patients, a cause of PH [60]. Furthermore, Ricard et al. also showed an increased migration and proliferation of pericytes induced by FGF2 and IL-2, in human PH patients. Finally, increased TGFβ also promotes the transdifferentiation of pericytes into contractile α-SMA positive cells (

Figure 1) [86].

) [61].

2.2.2. Impaired Multicellular Interactions Disrupt Pulmonary Vascular Development in BPD-PH

Multiple studies aiming at elucidating the pathogenesis of pulmonary VR have demonstrated an impairment of multicellular interactions, which could have resulted from abnormal expression levels of signaling pathways involved in cell–cell interactions.

As aforementioned, the Vegf signaling pathway is one of the most important signaling pathways regulating epithelial-endothelial crosstalk during pulmonary vascular development. Multiple studies have demonstrated that Vegf and Vegfr2 expressions were significantly decreased, while the expression of soluble Fms-like tyrosine kinase 1 (sFlt-1), an endogenous antagonist to Vegf corresponding to a truncated form of the Vegf receptor acting as a dominant negative Vegf receptor, was significantly increased, in experimental BPD animal models, which lead to a reduction and disarrangement of the microvasculature [87,88].

As aforementioned, the Vegf signaling pathway is one of the most important signaling pathways regulating epithelial-endothelial crosstalk during pulmonary vascular development. Multiple studies have demonstrated that Vegf and Vegfr2 expressions were significantly decreased, while the expression of soluble Fms-like tyrosine kinase 1 (sFlt-1), an endogenous antagonist to Vegf corresponding to a truncated form of the Vegf receptor acting as a dominant negative Vegf receptor, was significantly increased, in experimental BPD animal models, which lead to a reduction and disarrangement of the microvasculature [62][63].

Other signaling pathways potentially involved in the angiogenic network include sonic hedgehog (Shh) and Sprouty2 (Spry2). Sprouty2 is an inhibitor of Fgf10. Fgf10 is known to be a major regulator of epithelial branching. Inhibition of Vegfr1-mediated signaling leads to upregulation of Spry2 in the epithelium, suggesting a downregulation of Fgf10 signaling. This is an essential evidence demonstrating the importance of endothelial-mesenchymal crosstalk. More evidence confirming the endothelial-epithelial crosstalk came from DeLisser and colleagues. 

Pecam1-deficient mice revealed disrupted endothelial cell formation associated with decreased alveolar simplification [22,89].

-deficient mice revealed disrupted endothelial cell formation associated with decreased alveolar simplification [64].

Furthermore, a recent study found a reduction of WNT5 in the pulmonary microvascular endothelial cells (PMVECs) of PH patients. Through exposing 

Wnt5a Endothelial cKO mice with chronic hypoxia, the authors demonstrated that loss of endothelium-derived Wnt5a impaired the endothelium–pericytes interaction, resulting in significant reduction, muscularization, and decreased pericyte coverage of microvessels [72]. Another study, conducted in adult PH, revealed that PH pericytes overexpressed C-X-C motif chemokine receptor-7 (CXCR-7) and TGFβRII, and as compared with control pericytes, they were more likely to proliferate, migrate, and differentiate into smooth muscle-like cells, indicating a significant role of endothelium–pericytes interaction in the process of PH (

Endothelial cKO mice with chronic hypoxia, the authors demonstrated that loss of endothelium-derived Wnt5a impaired the endothelium–pericytes interaction, resulting in significant reduction, muscularization, and decreased pericyte coverage of microvessels [48]. Another study, conducted in adult PH, revealed that PH pericytes overexpressed C-X-C motif chemokine receptor-7 (CXCR-7) and TGFβRII, and as compared with control pericytes, they were more likely to proliferate, migrate, and differentiate into smooth muscle-like cells, indicating a significant role of endothelium–pericytes interaction in the process of PH (

Figure 1) [90].

) [65].

Pericyte-myofibroblast transition (PMT) could represent another pathogenesis of pulmonary VR, consistent with the results conducted in kidney fibrosis [91]. Wang et al. found that lung pericytes differentiated into myofibroblast in idiopathic pulmonary fibrosis (IPF) patients through increasing NOTCH1/PDGFRβ/ROCK1 signaling pathway [92]. However, the exact role of pericytes in the pathogenesis of BPD-PH is still unclear and needs to be further explored.

Pericyte-myofibroblast transition (PMT) could represent another pathogenesis of pulmonary VR, consistent with the results conducted in kidney fibrosis [66]. Wang et al. found that lung pericytes differentiated into myofibroblast in idiopathic pulmonary fibrosis (IPF) patients through increasing NOTCH1/PDGFRβ/ROCK1 signaling pathway [67]. However, the exact role of pericytes in the pathogenesis of BPD-PH is still unclear and needs to be further explored.

PDGFA/PDGFRA is an important signaling pathway controlling the elastogenesis of secondary crest myofibroblast during alveologenesis [93]. Interestingly, reduced PDGFRA has been found both in BPD patients and in hyperoxia-induced BPD mouse model [5,94]. Moreover, Oak and colleagues found that attenuated PDGFRA expression due to hyperoxia exposure resulted in a decrease of VEGFA, which ultimately led to increased ECs apoptosis and reduced microvessel numbers [94].
It is important to notice that FGF10 signaling has a close interaction with the VEGF signaling pathway. Therefore, we propose that FGF10 signaling plays critical roles in the pathogenesis of BPD-PH (discussed in detail in Section 3 below).

2.3. MicroRNAs (miRs) May Be Associated with the Pathologic Vascular Development of BPD-PH

A growing number of evidences show that expression of numerous microRNAs (miRs), that normally regulate different signaling pathways mediating cellular crosstalk in lung development, are dysregulated in PH and BPD. Thompson and Lawrie summarized a list of miRs which had therapeutic effects on PH from hypoxia or monocrotaline (MCT)-induced PH in animal experiments [44]. Combining the differentially expressed miRs in BPD versus control with Thompson and Lawrie’s findings in PH, we identified several miRs which were all dysregulated and had the same changes in both BPD and PH (

A growing number of evidences show that expression of numerous microRNAs (miRs), that normally regulate different signaling pathways mediating cellular crosstalk in lung development, are dysregulated in PH and BPD. Thompson and Lawrie summarized a list of miRs which had therapeutic effects on PH from hypoxia or monocrotaline (MCT)-induced PH in animal experiments . Combining the differentially expressed miRs in BPD versus control with Thompson and Lawrie’s findings in PH, we identified several miRs which were all dysregulated and had the same changes in both BPD and PH (

Table 1) [95,96]. These dysregulated miRs could be involved in BPD-PH.

) [66][67]. These dysregulated miRs could be involved in BPD-PH.

Table 1. MicroRNAs (miRs) potentially involved in pulmonary vascular remodeling in BPD.

MicroRNAs (miRs) potentially involved in pulmonary vascular remodeling in BPD.

 

References

  1. Blencowe, H.; Cousens, S.; Oestergaard, M.Z.; Chou, D.; Moller, A.B.; Narwal, R.; Adler, A.; Vera Garcia, C.; Rohde, S.; Say, L.; et al. National, regional, and worldwide estimates of preterm birth rates in the year 2010 with time trends since 1990 for selected countries: A systematic analysis and implications. Lancet 2012, 379, 2162–2172.
  2. Chawanpaiboon, S.; Vogel, J.P.; Moller, A.B.; Lumbiganon, P.; Petzold, M.; Hogan, D.; Landoulsi, S.; Jampathong, N.; Kongwattanakul, K.; Laopaiboon, M.; et al. Global, regional, and national estimates of levels of preterm birth in 2014: A systematic review and modelling analysis. Lancet Glob. Health 2019, 7, e37–e46.
  3. Stoll, B.J.; Hansen, N.I.; Bell, E.F.; Walsh, M.C.; Carlo, W.A.; Shankaran, S.; Laptook, A.R.; Sanchez, P.J.; Van Meurs, K.P.; Wyckoff, M.; et al. Trends in Care Practices, Morbidity, and Mortality of Extremely Preterm Neonates, 1993–2012. JAMA 2015, 314, 1039–1051.
  4. Bhatt, A.J.; Pryhuber, G.S.; Huyck, H.; Watkins, R.H.; Metlay, L.A.; Maniscalco, W.M. Disrupted pulmonary vasculature and decreased vascular endothelial growth factor, Flt-1, and TIE-2 in human infants dying with bronchopulmonary dysplasia. Am. J. Respir. Crit. Care Med. 2001, 164, 1971–1980.
  5. 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 C 2014, 307, L231–L239.
  6. Speer, C.P. Inflammation and bronchopulmonary dysplasia: A continuing story. Semin. Fetal Neonat. Med. 2006, 11, 354–362.
  7. Al-Ghanem, G.; Shah, P.; Thomas, S.; Banfield, L.; El Helou, S.; Fusch, C.; Mukerji, A. Bronchopulmonary dysplasia and pulmonary hypertension: A meta-analysis. J. Perinatol. 2017, 37, 414–419.
  8. Arjaans, S.; Zwart, E.A.H.; Ploegstra, M.J.; Bos, A.F.; Kooi, E.M.W.; Hillege, H.L.; Berger, R.M.F. Identification of gaps in the current knowledge on pulmonary hypertension in extremely preterm infants: A systematic review and meta-analysis. Paediatr. Perinat. Epidemiol. 2018, 32, 258–267.
  9. Sheikh, A.Q.; Lighthouse, J.K.; Greif, D.M. Recapitulation of developing artery muscularization in pulmonary hypertension. Cell Rep. 2014, 6, 809–817.
  10. Shimoda, L.A.; Laurie, S.S. Vascular remodeling in pulmonary hypertension. J. Mol. Med. Berl. 2013, 91, 297–309.
  11. Tan, W.; Madhavan, K.; Hunter, K.S.; Park, D.; Stenmark, K.R. Vascular stiffening in pulmonary hypertension: Cause or consequence? (2013 Grover Conference series). Pulm. Circ. 2014, 4, 560–580.
  12. Frank, D.B.; Crystal, M.A.; Morales, D.L.; Gerald, K.; Hanna, B.D.; Mallory, G.B., Jr.; Rossano, J.W. Trends in pediatric pulmonary hypertension-related hospitalizations in the United States from 2000–2009. Pulm. Circ. 2015, 5, 339–348.
  13. Haworth, S.G.; Hislop, A.A. Treatment and survival in children with pulmonary arterial hypertension: The UK Pulmonary Hypertension Service for Children 2001–2006. Heart 2009, 95, 312–317.
  14. Abman, S.H.; Hansmann, G.; Archer, S.L.; Ivy, D.D.; Adatia, I.; Chung, W.K.; Hanna, B.D.; Rosenzweig, E.B.; Raj, J.U.; Cornfield, D.; et al. Pediatric Pulmonary Hypertension: Guidelines From the American Heart Association and American Thoracic Society. Circulation 2015, 132, 2037–2099.
  15. Krishnan, U.; Feinstein, J.A.; Adatia, I.; Austin, E.D.; Mullen, M.P.; Hopper, R.K.; Hanna, B.; Romer, L.; Keller, R.L.; Fineman, J.; et al. Evaluation and Management of Pulmonary Hypertension in Children with Bronchopulmonary Dysplasia. J. Pediatr. 2017, 188, 24–34.
  16. Hansmann, G.; Apitz, C.; Abdul-Khaliq, H.; Alastalo, T.P.; Beerbaum, P.; Bonnet, D.; Dubowy, K.O.; Gorenflo, M.; Hager, A.; Hilgendorff, A.; et al. Executive summary. Expert consensus statement on the diagnosis and treatment of paediatric pulmonary hypertension. The European Paediatric Pulmonary Vascular Disease Network, endorsed by ISHLT and DGPK. Heart 2016, 102 (Suppl. S2), ii86–ii100.
  17. Simonneau, G.; Gatzoulis, M.A.; Adatia, I.; Celermajer, D.; Denton, C.; Ghofrani, A.; Gomez Sanchez, M.A.; Krishna Kumar, R.; Landzberg, M.; Machado, R.F.; et al. Updated clinical classification of pulmonary hypertension. J. Am. Coll. Cardiol. 2013, 62, D34–D41, doi:10.1016/j.jacc.2013.10.029.
  18. Ezekian, J.E.; Hill, K.D. Management of Pulmonary Arterial Hypertension in the Pediatric Patient. Curr. Cardiol. Rep. 2019, 21, 162, doi:10.1007/s11886-019-1229-2.
  19. Thompson, A.A.R.; Lawrie, A. Targeting Vascular Remodeling to Treat Pulmonary Arterial Hypertension. Trends Mol. Med. 2017, 23, 31–45, doi:10.1016/j.molmed.2016.11.005.
  20. Rosenzweig, E.B.; Abman, S.H.; Adatia, I.; Beghetti, M.; Bonnet, D.; Haworth, S.; Ivy, D.D.; Berger, R.M.F. Paediatric pulmonary arterial hypertension: Updates on definition, classification, diagnostics and management. Eur. Respir. J. 2019, 53, doi:10.1183/13993003.01916-2018.
  21. Mammoto, A.; Mammoto, T. Vascular Niche in Lung Alveolar Development, Homeostasis, and Regeneration. Front. Bioeng. Biotechnol. 2019, 7, 318, doi:10.3389/fbioe.2019.00318.
  22. Chao, C.M.; El Agha, E.; Tiozzo, C.; Minoo, P.; Bellusci, S. A breath of fresh air on the mesenchyme: Impact of impaired mesenchymal development on the pathogenesis of bronchopulmonary dysplasia. Front. Med. Lausanne 2015, 2, 27, doi:10.3389/fmed.2015.00027.
  23. Gebb, S.A.; Shannon, J.M. Tissue interactions mediate early events in pulmonary vasculogenesis. Dev. Dyn. 2000, 217, 159–169, doi:10.1002/(SICI)1097-0177(200002)217:2<159::AID-DVDY3>3.0.CO;2-9.
  24. Schittny, J.C. Development of the lung. Cell Tissue Res. 2017, 367, 427–444, doi:10.1007/s00441-016-2545-0.
  25. Stenmark, K.R.; Abman, S.H. Lung vascular development: Implications for the pathogenesis of bronchopulmonary dysplasia. Annu. Rev. Physiol. 2005, 67, 623–661, doi:10.1146/annurev.physiol.67.040403.102229.
  26. Sweeney, M.; Foldes, G. It Takes Two: Endothelial-Perivascular Cell Cross-Talk in Vascular Development and Disease. Front. Cardiovasc. Med. 2018, 5, 154, doi:10.3389/fcvm.2018.00154.
  27. Schittny, J.C.; Mund, S.I.; Stampanoni, M. Evidence and structural mechanism for late lung alveolarization. Am. J. Physiol. Lung Cell. Mol. Physiol. 2008, 294, L246–L254, doi:10.1152/ajplung.00296.2007.
  28. Greenberg, J.M.; Thompson, F.Y.; Brooks, S.K.; Shannon, J.M.; McCormick-Shannon, K.; Cameron, J.E.; Mallory, B.P.; Akeson, A.L. Mesenchymal expression of vascular endothelial growth factors D and A defines vascular patterning in developing lung. Dev. Dyn. 2002, 224, 144–153, doi:10.1002/dvdy.10095.
  29. White, A.C.; Lavine, K.J.; Ornitz, D.M. FGF9 and SHH regulate mesenchymal Vegfa expression and development of the pulmonary capillary network. Development 2007, 134, 3743–3752, doi:10.1242/dev.004879.
  30. Shalaby, F.; Rossant, J.; Yamaguchi, T.P.; Gertsenstein, M.; Wu, X.F.; Breitman, M.L.; Schuh, A.C. Failure of blood-island formation and vasculogenesis in Flk-1-deficient mice. Nature 1995, 376, 62–66, doi:10.1038/376062a0.
  31. Fong, G.H.; Rossant, J.; Gertsenstein, M.; Breitman, M.L. Role of the Flt-1 receptor tyrosine kinase in regulating the assembly of vascular endothelium. Nature 1995, 376, 66–70, doi:10.1038/376066a0.
  32. Yamamoto, H.; Yun, E.J.; Gerber, H.P.; Ferrara, N.; Whitsett, J.A.; Vu, T.H. Epithelial-Vascular cross talk mediated by VEGF-A and HGF signaling directs primary septae formation during distal lung morphogenesis. Dev. Biol. 2007, 308, 44–53, doi:10.1016/j.ydbio.2007.04.042.
  33. Zhao, L.; Wang, K.; Ferrara, N.; Vu, T.H. Vascular endothelial growth factor co-ordinates proper development of lung epithelium and vasculature. Mech. Dev. 2005, 122, 877–886, doi:10.1016/j.mod.2005.04.001.
  34. Yang, J.; Hernandez, B.J.; Martinez Alanis, D.; Narvaez del Pilar, O.; Vila-Ellis, L.; Akiyama, H.; Evans, S.E.; Ostrin, E.J.; Chen, J. The development and plasticity of alveolar type 1 cells. Development 2016, 143, 54–65, doi:10.1242/dev.130005.
  35. Greif, D.M.; Kumar, M.; Lighthouse, J.K.; Hum, J.; An, A.; Ding, L.; Red-Horse, K.; Espinoza, F.H.; Olson, L.; Offermanns, S.; et al. Radial construction of an arterial wall. Dev. Cell. 2012, 23, 482–493, doi:10.1016/j.devcel.2012.07.009.
  36. Bergers, G.; Song, S. The role of pericytes in blood-vessel formation and maintenance. Neuro Oncol. 2005, 7, 452–464, doi:10.1215/S1152851705000232.
  37. Armulik, A.; Genove, G.; Betsholtz, C. Pericytes: Developmental, physiological, and pathological perspectives, problems, and promises. Dev. Cell. 2011, 21, 193–215, doi:10.1016/j.devcel.2011.07.001.
  38. Crisan, M.; Yap, S.; Casteilla, L.; Chen, C.W.; Corselli, M.; Park, T.S.; Andriolo, G.; Sun, B.; Zheng, B.; Zhang, L.; et al. A perivascular origin for mesenchymal stem cells in multiple human organs. Cell Stem Cell 2008, 3, 301–313, doi:10.1016/j.stem.2008.07.003.
  39. Paiva, A.E.; Lousado, L.; Guerra, D.A.P.; Azevedo, P.O.; Sena, I.F.G.; Andreotti, J.P.; Santos, G.S.P.; Goncalves, R.; Mintz, A.; Birbrair, A. Pericytes in the Premetastatic Niche. Cancer Res. 2018, 78, 2779–2786, doi:10.1158/0008-5472.CAN-17-3883.
  40. Kramann, R.; Schneider, R.K.; DiRocco, D.P.; Machado, F.; Fleig, S.; Bondzie, P.A.; Henderson, J.M.; Ebert, B.L.; Humphreys, B.D. Perivascular Gli1+ progenitors are key contributors to injury-induced organ fibrosis. Cell Stem Cell 2015, 16, 51–66, doi:10.1016/j.stem.2014.11.004.
  41. Eilken, H.M.; Dieguez-Hurtado, R.; Schmidt, I.; Nakayama, M.; Jeong, H.W.; Arf, H.; Adams, S.; Ferrara, N.; Adams, R.H. Pericytes regulate VEGF-induced endothelial sprouting through VEGFR1. Nat. Commun. 2017, 8, 1574, doi:10.1038/s41467-017-01738-3.
  42. Hellstrom, M.; Kalen, M.; Lindahl, P.; Abramsson, A.; Betsholtz, C. Role of PDGF-B and PDGFR-beta in recruitment of vascular smooth muscle cells and pericytes during embryonic blood vessel formation in the mouse. Development 1999, 126, 3047–3055.
  43. Jarad, M.; Kuczynski, E.A.; Morrison, J.; Viloria-Petit, A.M.; Coomber, B.L. Release of endothelial cell associated VEGFR2 during TGF-beta modulated angiogenesis in vitro. BMC Cell Biol. 2017, 18, 10, doi:10.1186/s12860-017-0127-y.
  44. Wakui, S.; Yokoo, K.; Muto, T.; Suzuki, Y.; Takahashi, H.; Furusato, M.; Hano, H.; Endou, H.; Kanai, Y. Localization of Ang-1, -2, Tie-2, and VEGF expression at endothelial-pericyte interdigitation in rat angiogenesis. Lab. Invest. 2006, 86, 1172–1184, doi:10.1038/labinvest.3700476.
  45. Armulik, A.; Abramsson, A.; Betsholtz, C. Endothelial/Pericyte interactions. Circ. Res. 2005, 97, 512–523, doi:10.1161/01.RES.0000182903.16652.d7.
  46. Kato, K.; Dieguez-Hurtado, R.; Park, D.Y.; Hong, S.P.; Kato-Azuma, S.; Adams, S.; Stehling, M.; Trappmann, B.; Wrana, J.L.; Koh, G.Y.; et al. Pulmonary pericytes regulate lung morphogenesis. Nat. Commun. 2018, 9, 2448, doi:10.1038/s41467-018-04913-2.
  47. Yuan, K.; Shamskhou, E.A.; Orcholski, M.E.; Nathan, A.; Reddy, S.; Honda, H.; Mani, V.; Zeng, Y.; Ozen, M.O.; Wang, L.; et al. Loss of Endothelium-Derived Wnt5a Is Associated With Reduced Pericyte Recruitment and Small Vessel Loss in Pulmonary Arterial Hypertension. Circulation 2019, 139, 1710–1724, doi:10.1161/CIRCULATIONAHA.118.037642.
  48. Shammout, B.; Johnson, J.R. Pericytes in Chronic Lung Disease. Adv. Exp. Med. Biol. 2019, 1147, 299–317, doi:10.1007/978-3-030-16908-4_14.
  49. Gao, Y.; Chen, T.; Raj, J.U. Endothelial and Smooth Muscle Cell Interactions in the Pathobiology of Pulmonary Hypertension. Am. J. Respir. Cell. Mol. Biol. 2016, 54, 451–460, doi:10.1165/rcmb.2015-0323TR.
  50. Hadchouel, A.; Durrmeyer, X.; Bouzigon, E.; Incitti, R.; Huusko, J.; Jarreau, P.H.; Lenclen, R.; Demenais, F.; Franco-Montoya, M.L.; Layouni, I.; et al. Identification of SPOCK2 as a susceptibility gene for bronchopulmonary dysplasia. Am. J. Respir. Crit. Care Med. 2011, 184, 1164–1170, doi:10.1164/rccm.201103-0548OC.
  51. 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, doi:10.1093/hmg/ddy439.
  52. Yee, M.; White, R.J.; Awad, H.A.; Bates, W.A.; McGrath-Morrow, S.A.; O’Reilly, M.A. Neonatal hyperoxia causes pulmonary vascular disease and shortens life span in aging mice. Am. J. Pathol. 2011, 178, 2601–2610, doi:10.1016/j.ajpath.2011.02.010.
  53. Tiozzo, C.; Carraro, G.; Al Alam, D.; Baptista, S.; Danopoulos, S.; Li, A.; Lavarreda-Pearce, M.; Li, C.; De Langhe, S.; Chan, B.; et al. Mesodermal Pten inactivation leads to alveolar capillary dysplasia- like phenotype. J. Clin. Invest. 2012, 122, 3862–3872, doi:10.1172/JCI61334.
  54. Abman, S.H. Bronchopulmonary dysplasia: A vascular hypothesis. Am. J. Respir. Crit. Care Med. 2001, 164, 1755–1756, doi:10.1164/ajrccm.164.10.2109111c.
  55. Baker, C.D.; Abman, S.H. Impaired pulmonary vascular development in bronchopulmonary dysplasia. Neonatology 2015, 107, 344–351, doi:10.1159/000381129.
  56. Greco, F.; Wiegert, S.; Baumann, P.; Wellmann, S.; Pellegrini, G.; Cannizzaro, V. Hyperoxia-Induced lung structure-function relation, vessel rarefaction, and cardiac hypertrophy in an infant rat model. J. Transl. Med. 2019, 17, 91, doi:10.1186/s12967-019-1843-1.
  57. Crnkovic, S.; Marsh, L.M.; El Agha, E.; Voswinckel, R.; Ghanim, B.; Klepetko, W.; Stacher-Priehse, E.; Olschewski, H.; Bloch, W.; Bellusci, S.; et al. Resident cell lineages are preserved in pulmonary vascular remodeling. J. Pathol. 2018, 244, 485–498, doi:10.1002/path.5044.
  58. Stacher, E.; Graham, B.B.; Hunt, J.M.; Gandjeva, A.; Groshong, S.D.; McLaughlin, V.V.; Jessup, M.; Grizzle, W.E.; Aldred, M.A.; Cool, C.D.; et al. Modern age pathology of pulmonary arterial hypertension. Am. J. Respir. Crit. Care Med. 2012, 186, 261–272, doi:10.1164/rccm.201201-0164OC.
  59. Stenmark, K.R.; Frid, M.; Perros, F. Endothelial-To-Mesenchymal Transition: An Evolving Paradigm and a Promising Therapeutic Target in PAH. Circulation 2016, 133, 1734–1737, doi:10.1161/CIRCULATIONAHA.116.022479.
  60. Assaad, A.M.; Kawut, S.M.; Arcasoy, S.M.; Rosenzweig, E.B.; Wilt, J.S.; Sonett, J.R.; Borczuk, A.C. Platelet-Derived growth factor is increased in pulmonary capillary hemangiomatosis. Chest 2007, 131, 850–855, doi:10.1378/chest.06-1680.
  61. Ricard, N.; Tu, L.; Le Hiress, M.; Huertas, A.; Phan, C.; Thuillet, R.; Sattler, C.; Fadel, E.; Seferian, A.; Montani, D.; et al. Increased pericyte coverage mediated by endothelial-derived fibroblast growth factor-2 and interleukin-6 is a source of smooth muscle-like cells in pulmonary hypertension. Circulation 2014, 129, 1586–1597, doi:10.1161/CIRCULATIONAHA.113.007469.
  62. Lin, S.L.; Chang, F.C.; Schrimpf, C.; Chen, Y.T.; Wu, C.F.; Wu, V.C.; Chiang, W.C.; Kuhnert, F.; Kuo, C.J.; Chen, Y.M.; et al. Targeting endothelium-pericyte cross talk by inhibiting VEGF receptor signaling attenuates kidney microvascular rarefaction and fibrosis. Am. J. Pathol. 2011, 178, 911–923, doi:10.1016/j.ajpath.2010.10.012.
  63. Wang, Y.C.; Chen, Q.; Luo, J.M.; Nie, J.; Meng, Q.H.; Shuai, W.; Xie, H.; Xia, J.M.; Wang, H. Notch1 promotes the pericyte-myofibroblast transition in idiopathic pulmonary fibrosis through the PDGFR/ROCK1 signal pathway. Exp. Mol. Med. 2019, 51, 1–11, doi:10.1038/s12276-019-0228-0.
  64. Li, C.; Lee, M.K.; Gao, F.; Webster, S.; Di, H.; Duan, J.; Yang, C.Y.; Bhopal, N.; Peinado, N.; Pryhuber, G.; et al. Secondary crest myofibroblast PDGFRalpha controls the elastogenesis pathway via a secondary tier of signaling networks during alveologenesis. Development 2019, 146, doi:10.1242/dev.176354.
  65. Oak, P.; Pritzke, T.; Thiel, I.; Koschlig, M.; Mous, D.S.; Windhorst, A.; Jain, N.; Eickelberg, O.; Foerster, K.; Schulze, A.; et al. Attenuated PDGF signaling drives alveolar and microvascular defects in neonatal chronic lung disease. EMBO Mol. Med. 2017, 9, 1504–1520, doi:10.15252/emmm.201607308.
  66. Xing, Y.; Fu, J.; Yang, H.; Yao, L.; Qiao, L.; Du, Y.; Xue, X. MicroRNA expression profiles and target prediction in neonatal Wistar rat lungs during the development of bronchopulmonary dysplasia. Int. J. Mol. Med. 2015, 36, 1253–1263, doi:10.3892/ijmm.2015.2347.
  67. Yang, Y.; Qiu, J.; Kan, Q.; Zhou, X.G.; Zhou, X.Y. MicroRNA expression profiling studies on bronchopulmonary dysplasia: A systematic review and meta-analysis. Genet. Mol. Res. 2013, 12, 5195–5206, doi:10.4238/2013.October.30.4.
  68. Yang, S.; Banerjee, S.; Freitas, A.; Cui, H.; Xie, N.; Abraham, E.; Liu, G. miR-21 regulates chronic hypoxia-induced pulmonary vascular remodeling. Am. J. Physiol. Lung Cell. Mol. Physiol. 2012, 302, L521–L529, doi:10.1152/ajplung.00316.2011.
  69. Bhaskaran, M.; Xi, D.; Wang, Y.; Huang, C.; Narasaraju, T.; Shu, W.; Zhao, C.; Xiao, X.; More, S.; Breshears, M.; et al. Identification of microRNAs changed in the neonatal lungs in response to hyperoxia exposure. Physiol. Genomics 2012, 44, 970–980, doi:10.1152/physiolgenomics.00145.2011.
  70. Chen, X.; Talati, M.; Fessel, J.P.; Hemnes, A.R.; Gladson, S.; French, J.; Shay, S.; Trammell, A.; Phillips, J.A.; Hamid, R.; et al. Estrogen Metabolite 16alpha-Hydroxyestrone Exacerbates Bone Morphogenetic Protein Receptor Type II-Associated Pulmonary Arterial Hypertension Through MicroRNA-29-Mediated Modulation of Cellular Metabolism. Circulation 2016, 133, 82–97, doi:10.1161/CIRCULATIONAHA.115.016133.
  71. Hu, Y.; Xie, L.; Yu, J.; Fu, H.; Zhou, D.; Liu, H. Inhibition of microRNA-29a alleviates hyperoxia-induced bronchopulmonary dysplasia in neonatal mice via upregulation of GAB1. Mol. Med. 2019, 26, 3, doi:10.1186/s10020-019-0127-9.
  72. Dong, J.; Carey, W.A.; Abel, S.; Collura, C.; Jiang, G.; Tomaszek, S.; Sutor, S.; Roden, A.C.; Asmann, Y.W.; Prakash, Y.S.; et al. MicroRNA-mRNA interactions in a murine model of hyperoxia-induced bronchopulmonary dysplasia. BMC Genomics 2012, 13, 204, doi:10.1186/1471-2164-13-204.
  73. Syed, M.; Das, P.; Pawar, A.; Aghai, Z.H.; Kaskinen, A.; Zhuang, Z.W.; Ambalavanan, N.; Pryhuber, G.; Andersson, S.; Bhandari, V. Hyperoxia causes miR-34a-mediated injury via angiopoietin-1 in neonatal lungs. Nat. Commun. 2017, 8, 1173, doi:10.1038/s41467-017-01349-y.
  74. Ruiz-Camp, J.; Quantius, J.; Lignelli, E.; Arndt, P.F.; Palumbo, F.; Nardiello, C.; Surate Solaligue, D.E.; Sakkas, E.; Mizikova, I.; Rodriguez-Castillo, J.A.; et al. Targeting miR-34a/Pdgfra interactions partially corrects alveologenesis in experimental bronchopulmonary dysplasia. EMBO Mol. Med. 2019, 11, doi:10.15252/emmm.201809448.
  75. Potus, F.; Ruffenach, G.; Dahou, A.; Thebault, C.; Breuils-Bonnet, S.; Tremblay, E.; Nadeau, V.; Paradis, R.; Graydon, C.; Wong, R.; et al. Downregulation of MicroRNA-126 Contributes to the Failing Right Ventricle in Pulmonary Arterial Hypertension. Circulation 2015, 132, 932–943, doi:10.1161/CIRCULATIONAHA.115.016382.
  76. Bertero, T.; Cottrill, K.A.; Lu, Y.; Haeger, C.M.; Dieffenbach, P.; Annis, S.; Hale, A.; Bhat, B.; Kaimal, V.; Zhang, Y.Y.; et al. Matrix Remodeling Promotes Pulmonary Hypertension through Feedback Mechanoactivation of the YAP/TAZ-miR-130/301 Circuit. Cell Rep. 2015, 13, 1016–1032, doi:10.1016/j.celrep.2015.09.049.
  77. Chao, C.M.; Carraro, G.; Rako, Z.A.; Kolck, J.; Sedighi, J.; Zimmermann, V.; Moiseenko, A.; Wilhelm, J.; Young, B.M.; Chong, L.; et al. Failure to Down-Regulate miR-154 Expression in Early Postnatal Mouse Lung Epithelium Suppresses Alveologenesis, with Changes in Tgf-beta Signaling Similar to those Induced by Exposure to Hyperoxia. Cells 2020, 9, doi:10.3390/cells9040859.
  78. Bernardo, B.C.; Nguyen, S.S.; Gao, X.M.; Tham, Y.K.; Ooi, J.Y.; Patterson, N.L.; Kiriazis, H.; Su, Y.; Thomas, C.J.; Lin, R.C.; et al. Inhibition of miR-154 Protects Against Cardiac Dysfunction and Fibrosis in a Mouse Model of Pressure Overload. Sci. Rep. 2016, 6, 22442, doi:10.1038/srep22442.
  79. Milosevic, J.; Pandit, K.; Magister, M.; Rabinovich, E.; Ellwanger, D.C.; Yu, G.; Vuga, L.J.; Weksler, B.; Benos, P.V.; Gibson, K.F.; et al. Profibrotic role of miR-154 in pulmonary fibrosis. Am. J. Respir. Cell Mol. Biol. 2012, 47, 879–887, doi:10.1165/rcmb.2011-0377OC.
  80. Yuan, H.S.; Xiong, D.Q.; Huang, F.; Cui, J.; Luo, H. MicroRNA-421 inhibition alleviates bronchopulmonary dysplasia in a mouse model via targeting Fgf10. J. Cell. Biochem. 2019, 120, 16876–16887, doi:10.1002/jcb.28945.
  81. Kim, J.; Kang, Y.; Kojima, Y.; Lighthouse, J.K.; Hu, X.; Aldred, M.A.; McLean, D.L.; Park, H.; Comhair, S.A.; Greif, D.M.; et al. An endothelial apelin-FGF link mediated by miR-424 and miR-503 is disrupted in pulmonary arterial hypertension. Nat. Med. 2013, 19, 74–82, doi:10.1038/nm.3040.
  82. Chen, J.; Cui, X.; Li, L.; Qu, J.; Raj, J.U.; Gou, D. MiR-339 inhibits proliferation of pulmonary artery smooth muscle cell by targeting FGF signaling. Physiol. Rep. 2017, 5, doi:10.14814/phy2.13441.
  83. Jones, M.R.; Lingampally, A.; Dilai, S.; Shrestha, A.; Stripp, B.; Helmbacher, F.; Chen, C.; Chao, C.M.; Bellusci, S. Characterization of Tg(Etv4-GFP) and Etv5 (RFP) Reporter Lines in the Context of Fibroblast Growth Factor 10 Signaling During Mouse Embryonic Lung Development. Front. Genet. 2019, 10, 178, doi:10.3389/fgene.2019.00178.
  84. Mailleux, A.A.; Kelly, R.; Veltmaat, J.M.; De Langhe, S.P.; Zaffran, S.; Thiery, J.P.; Bellusci, S. Fgf10 expression identifies parabronchial smooth muscle cell progenitors and is required for their entry into the smooth muscle cell lineage. Development 2005, 132, 2157–2166, doi:10.1242/dev.01795.
  85. Ohuchi, H.; Hori, Y.; Yamasaki, M.; Harada, H.; Sekine, K.; Kato, S.; Itoh, N. FGF10 acts as a major ligand for FGF receptor 2 IIIb in mouse multi-organ development. Biochem. Biophys. Res. Commun. 2000, 277, 643–649, doi:10.1006/bbrc.2000.3721.
  86. 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, doi:10.1164/rccm.200811-1794OC.
  87. Volckaert, T.; Dill, E.; Campbell, A.; Tiozzo, C.; Majka, S.; Bellusci, S.; De Langhe, S.P. Parabronchial smooth muscle constitutes an airway epithelial stem cell niche in the mouse lung after injury. J. Clin. Invest. 2011, 121, 4409–4419, doi:10.1172/JCI58097.
  88. El Agha, E.; Moiseenko, A.; Kheirollahi, V.; De Langhe, S.; Crnkovic, S.; Kwapiszewska, G.; Szibor, M.; Kosanovic, D.; Schwind, F.; Schermuly, R.T.; et al. Two-Way Conversion between Lipogenic and Myogenic Fibroblastic Phenotypes Marks the Progression and Resolution of Lung Fibrosis. Cell Stem Cell 2017, 20, 261–273, doi:10.1016/j.stem.2016.10.004.
  89. Chao, C.M.; Yahya, F.; Moiseenko, A.; Tiozzo, C.; Shrestha, A.; Ahmadvand, N.; El Agha, E.; Quantius, J.; Dilai, S.; Kheirollahi, V.; et al. Fgf10 deficiency is causative for lethality in a mouse model of bronchopulmonary dysplasia. J. Pathol. 2017, 241, 91–103, doi:10.1002/path.4834.
  90. Scott, C.L.; Walker, D.J.; Cwiklinski, E.; Tait, C.; Tee, A.R.; Land, S.C. Control of HIF-1{alpha} and vascular signaling in fetal lung involves cross talk between mTORC1 and the FGF-10/FGFR2b/Spry2 airway branching periodicity clock. Am. J. Physiol. Lung Cell. Mol. Physiol. 2010, 299, L455–L471, doi:10.1152/ajplung.00348.2009.
  91. Del Moral, P.M.; Sala, F.G.; Tefft, D.; Shi, W.; Keshet, E.; Bellusci, S.; Warburton, D. VEGF-A signaling through Flk-1 is a critical facilitator of early embryonic lung epithelial to endothelial crosstalk and branching morphogenesis. Dev. Biol. 2006, 290, 177–188, doi:10.1016/j.ydbio.2005.11.022.
  92. Ramasamy, S.K.; Mailleux, A.A.; Gupte, V.V.; Mata, F.; Sala, F.G.; Veltmaat, J.M.; Del Moral, P.M.; De Langhe, S.; Parsa, S.; Kelly, L.K.; et al. Fgf10 dosage is critical for the amplification of epithelial cell progenitors and for the formation of multiple mesenchymal lineages during lung development. Dev. Biol. 2007, 307, 237–247, doi:10.1016/j.ydbio.2007.04.033.
  93. Benjamin, J.T.; Smith, R.J.; Halloran, B.A.; Day, T.J.; Kelly, D.R.; Prince, L.S. FGF-10 is decreased in bronchopulmonary dysplasia and suppressed by Toll-like receptor activation. Am. J. Physiol. Lung. Cell. Mol. Physiol. 2007, 292, L550–L558, doi:10.1152/ajplung.00329.2006.
  94. Seghezzi, G.; Patel, S.; Ren, C.J.; Gualandris, A.; Pintucci, G.; Robbins, E.S.; Shapiro, R.L.; Galloway, A.C.; Rifkin, D.B.; Mignatti, P. Fibroblast growth factor-2 (FGF-2) induces vascular endothelial growth factor (VEGF) expression in the endothelial cells of forming capillaries: An autocrine mechanism contributing to angiogenesis. J. Cell Biol. 1998, 141, 1659–1673, doi:10.1083/jcb.141.7.1659.
  95. Gospodarowicz, D.; Brown, K.D.; Birdwell, C.R.; Zetter, B.R. Control of proliferation of human vascular endothelial cells. Characterization of the response of human umbilical vein endothelial cells to fibroblast growth factor, epidermal growth factor, and thrombin. J. Cell Biol. 1978, 77, 774–788, doi:10.1083/jcb.77.3.774.
  96. Claus, P.; Grothe, C. Molecular cloning and developmental expression of rat fibroblast growth factor receptor 3. Histochem. Cell Biol. 2001, 115, 147–155, doi:10.1007/s004180000215.
  97. Shin, J.W.; Min, M.; Larrieu-Lahargue, F.; Canron, X.; Kunstfeld, R.; Nguyen, L.; Henderson, J.E.; Bikfalvi, A.; Detmar, M.; Hong, Y.K. Prox1 promotes lineage-specific expression of fibroblast growth factor (FGF) receptor-3 in lymphatic endothelium: A role for FGF signaling in lymphangiogenesis. Mol. Biol. Cell 2006, 17, 576–584, doi:10.1091/mbc.e05-04-0368.
  98. Lieu, C.; Heymach, J.; Overman, M.; Tran, H.; Kopetz, S. Beyond VEGF: Inhibition of the fibroblast growth factor pathway and antiangiogenesis. Clin. Cancer Res. 2011, 17, 6130–6139, doi:10.1158/1078-0432.CCR-11-0659.
  99. Murakami, M.; Elfenbein, A.; Simons, M. Non-Canonical fibroblast growth factor signalling in angiogenesis. Cardiovasc. Res. 2008, 78, 223–231, doi:10.1093/cvr/cvm086.
  100. Murakami, M.; Nguyen, L.T.; Hatanaka, K.; Schachterle, W.; Chen, P.Y.; Zhuang, Z.W.; Black, B.L.; Simons, M. FGF-Dependent regulation of VEGF receptor 2 expression in mice. J. Clin. Invest. 2011, 121, 2668–2678, doi:10.1172/JCI44762.
  101. Hatanaka, K.; Lanahan, A.A.; Murakami, M.; Simons, M. Fibroblast growth factor signaling potentiates VE-cadherin stability at adherens junctions by regulating SHP2. PLoS ONE 2012, 7, e37600, doi:10.1371/journal.pone.0037600.
  102. Oladipupo, S.S.; Smith, C.; Santeford, A.; Park, C.; Sene, A.; Wiley, L.A.; Osei-Owusu, P.; Hsu, J.; Zapata, N.; Liu, F.; et al. Endothelial cell FGF signaling is required for injury response but not for vascular homeostasis. Proc. Natl. Acad. Sci. USA 2014, 111, 13379–13384, doi:10.1073/pnas.1324235111.
  103. House, S.L.; Castro, A.M.; Lupu, T.S.; Weinheimer, C.; Smith, C.; Kovacs, A.; Ornitz, D.M. Endothelial fibroblast growth factor receptor signaling is required for vascular remodeling following cardiac ischemia-reperfusion injury. Am. J. Physiol. Heart Circ. Physiol. 2016, 310, H559–H571, doi:10.1152/ajpheart.00758.2015.
More