Skip to main content
Cardiovascular Research logoLink to Cardiovascular Research
. 2018 Jan 27;114(4):551–564. doi: 10.1093/cvr/cvy004

Dynamic and diverse changes in the functional properties of vascular smooth muscle cells in pulmonary hypertension

Kurt R Stenmark 1,, Maria G Frid 1, Brian B Graham 2, Rubin M Tuder 2
PMCID: PMC5852549  PMID: 29385432

Abstract

Pulmonary hypertension (PH) is the end result of interaction between pulmonary vascular tone and a complex series of cellular and molecular events termed ‘vascular remodelling’. The remodelling process, which can involve the entirety of pulmonary arterial vasculature, almost universally involves medial thickening, driven by increased numbers and hypertrophy of its principal cellular constituent, smooth muscle cells (SMCs). It is noted, however that SMCs comprise heterogeneous populations of cells, which can exhibit markedly different proliferative, inflammatory, and extracellular matrix production changes during remodelling. We further consider that these functional changes in SMCs of different phenotype and their role in PH are dynamic and may undergo significant changes over time (which we will refer to as cellular plasticity); no single property can account for the complexity of the contribution of SMC to pulmonary vascular remodelling. Thus, the approaches used to pharmacologically manipulate PH by targeting the SMC phenotype(s) must take into account processes that underlie dominant phenotypes that drive the disease. We present evidence for time- and location-specific changes in SMC proliferation in various animal models of PH; we highlight the transient nature (rather than continuous) of SMC proliferation, emphasizing that the heterogenic SMC populations that reside in different locations along the pulmonary vascular tree exhibit distinct responses to the stresses associated with the development of PH. We also consider that cells that have often been termed ‘SMCs’ may arise from many origins, including endothelial cells, fibroblasts and resident or circulating progenitors, and thus may contribute via distinct signalling pathways to the remodelling process. Ultimately, PH is characterized by long-lived, apoptosis-resistant SMC. In line with this key pathogenic characteristic, we address the acquisition of a pro-inflammatory phenotype by SMC that is essential to the development of PH. We present evidence that metabolic alterations akin to those observed in cancer cells (cytoplasmic and mitochondrial) directly contribute to the phenotype of the SM and SM-like cells involved in PH. Finally, we raise the possibility that SMCs transition from a proliferative to a senescent, pro-inflammatory and metabolically active phenotype over time.

Keywords: Progenitor cells, Endothelial mesenchymal transition, Inflammation, Metabolism, Senescence

1. Introduction

Pulmonary hypertension (PH) is the end result of interaction between pulmonary vascular tone and a complex series of cellular and molecular events termed ‘vascular remodelling’. This remodelling involves the intimal, medial, and adventitial compartments of the pulmonary (arterial and venous) vessel wall and often the perivascular space as well. The remodelling process almost universally involves medial thickening, driven by increased numbers and hypertrophy of its principal cellular constituent, smooth muscle cells (SMCs); concomitant with these processes, SMCs exhibit enhanced chemokine and cytokine production, and alterations in extracellular matrix protein production and degradation. We consider that these structural and functional changes in SMCs and their role in PH are dynamic and may undergo significant changes over time (which we will refer to as cellular plasticity); no single property can account for the complexity of the contribution of SMCs to pulmonary vascular remodelling. Thus, the approaches used to pharmacologically manipulate PH by targeting the SMC phenotype(s) must take into account processes that underlie dominant phenotypes that drive the disease.

SMCs are heterogeneous in the normal pulmonary circulation. It has been shown that pulmonary artery (PA) SMCs are not uniform in phenotype throughout the pulmonary circulation, but rather are heterogeneous at both a single anatomical site (proximal vs. distal PAs) and along a specific vascular segment, exhibiting site-specific and unique responses to pathologic hypertensive stimuli.1–5 For instance, the cellular composition of the bovine PA media changes along its proximal to distal longitudinal axis from being phenotypically heterogeneous in the pulmonary trunk to being phenotypically uniform in distal resistance vessels (see Figure 1).6 The media of the proximal large PAs, including the main PA (MPA), is composed of a mosaic of more differentiated SMCs and less differentiated ‘SM-like’ cells (characterized by expression of α-SM-actin but not by other differentiation markers), while in the distal muscular PAs, the arterial media is composed of a phenotypically uniform and apparently well-differentiated SMC phenotype (including expression of SM-myosin heavy chains, SM h-caldesmon and metavinculin). These observations in vivo provided a ‘cellular basis’ for the different functional properties of vessels along the vascular tree and are consistent with the classic physiologic studies of Burton et al.7 These studies outlined that great arteries are structured to provide tensile strength and withstand high wall tension along with a certain degree of distensibility to accommodate stroke volume. The small arteries, on the other hand are exposed to much less wall stress and pulse pressure than the large arteries. In these distal arteries, under homeostatic conditions, the interaction of endothelial cells with contractile SMCs provide the tone essential for regulation of blood pressure and flow in the pulmonary circulation.

Figure 1.

Figure 1

(I) Schematic representation of the pulmonary arterial tree depicting levels, at which the sections were obtained. (II) The cellular composition of the bovine arterial media gradually changes along the longitudinal axis from phenotypically heterogeneous (proximal arteries) to more uniform (distal arteries). This is not immediately apparent on histologically stained tissue (H&E; left A, D, G, J, M), but becomes evident via immunostaining for SM-specific differentiation markers: meta-vinculin (middle: B, E, H, K, N) and smooth muscle myosin heavy chain B isoform (SM-MHC-B; right: C, F, I, L, O). From top to bottom: MPA (A–C), intra-lobar PAs with inner diameters of: 3000 μm (D–F), 1, 500 μm (G–I), 500 μm (J–L), and 100 μm (M–O). Scale, 100 μm. Adapted from reference.6

The purpose of this perspective is to (i) review the evidence for time- and location-specific changes in SMC proliferation in various models of PH, with special emphasis on the transient nature of SMC proliferation; (ii) emphasize that the heterogenic SMC populations that reside in different locations along the pulmonary vascular tree exhibit distinct responses to the stresses associated with the development of PH; (iii) review the potential sources of SM or SM-like cells that acquire a proliferative phenotype and/or contribute in other ways to the remodelling process, including endothelial cells (throughout the process of endothelial–mesenchymal transition (EndMT)), fibroblasts, and resident and circulating progenitors; (iv) review evidence of the pro-inflammatory phenotype of SMCs in PH; (v) present evidence that metabolic alterations (cytoplasmic and mitochondrial), which are akin to those observed in cancer cells directly contribute to the phenotype of the SM and SM-like cells involved; and (vi) present evidence supporting the possibility that SMCs transition from a proliferative to a senescent, pro-inflammatory and metabolically active phenotype over time.

2. SMC proliferation and hypertrophy in experimental animal models and in human PH

2.1 Time, location and species dependent effects

Increased medial thickness of the PAs is a well-established feature of PH in animal models and humans. Proliferation of resident medial SMCs is widely believed to play an important role in this thickening. Seminal studies in the late ‘70 s by Dr Meyrick and Dr Reid established site- and time-specific changes in the medial thickening and SMC proliferation that occurred in animal models of PH.8–11 In the hypoxic rat model of PH (Sprague-Dawley rats @ 230 g at hypoxia initiation), the first increases in thymidine incorporation occurred in the hilar vessels, yet these were predominately in the adventitial and endothelial compartments, with only minimal change (<2% labelling index) in the medial compartment. The authors also showed that thickening of the large vessels appeared to consistently follow the increase in PA pressure. However, medial changes and SMC proliferation in the intra-acinar arteries and vessels of the alveolar wall occurred later. Similar, but not identical observations were made in the monocrotaline rat (Sprague-Dawley) model of PH.9 Changes in SMC proliferation in hilar vessels also appeared to follow increases in PA pressure, but occurred later, whereas the time course of changes in the intra-acinar vessels occurred earlier because the authors speculated that the timing of SMC proliferation in this models PH was due to more severe endothelial injury by the alkaloid monocrotaline and associated interstitial inflammation than those observed in the hypoxic model. The most consistent observation with regard to SMCs in these two experimental PH models was extension of the ‘sleeve of SM-like cells to vessels that had been previously non-muscularized’. This response was believed, at the time, to be mediated by proliferation of intermediate ‘SMC-like cells’ and/or pericytes.8–11

Subsequent studies in other animal models of PH have largely supported these findings: in hypoxic mice, proliferation of SMCs was greatest in larger hilar vessels and occurred early (at 3–4 days) but then gradually diminished, and, as most studies demonstrated, after 4–5 weeks of hypoxia cell proliferation was minimal.12–14 Distal muscularization occurred early and consistently in the hypoxic mouse models, similar to that in rat.15,16 The Schistosoma mouse model also has proliferation of cells in the medial layer, and again SMC proliferation decreases with time.17 Further, these mouse and rat models are characterized by PAs in which the endothelial cells, though dysfunctional, line the PAs as single layer. This structural characteristic is also present in human disease associated with left ventricular dysfunction (WHO Group 2), hypoxia and interstitial injury (WHO Group 3). However, the paradigmatic pathology of severe PH in humans, characteristic of idiopathic and BMPR2 mutation associated pulmonary arterial hypertension, characterized by excessive luminal growth of endothelial cells, forming plexiform lesions, and significant but notably more mild expansion of the medial layer is not observed in these animal models.18,19 These findings underscore that PH is not a monolithic disease and much can be gained from dissecting the commonalities and differences among the multiple of forms of PH.

Recently, a Sugen + Hypoxia model of PH (based on the combination of the VEGF receptor blocker SU5416 and chronic hypoxia20,21) has been increasingly thought of as one of the better models to study human pulmonary arterial hypertension (PAH), combining suprasystemic levels of PA pressures with progressive plexiform-like lesions. It is noteworthy that, in this model, there is again evidence for only transient proliferation of SMCs in the large as well as small vessels, notably early in the course of the disease; importantly, this proliferation response wanes with time and is insignificant at later stages (Figure 2). This observation challenges the dominant dogma that SMC proliferation is the root cause of PH, most importantly, of the persistence and chronicity of SMC in vascular remodelling; it rather suggests that SMC proliferation may indeed be only a transient response and that, in established disease, alternative SMC intermediate phenotypes might play key roles in established disease.

Figure 2.

Figure 2

SMC proliferation is minimal in the Sugen + Hypoxia model of PH. (A) Percent medial thickness in PAs of rats treated with the VEGF receptor blocker SU5416 and exposed to chronic hypoxia for 1–3 weeks; (3 + 3) indicates the group of rats treated with SU5416+chronic hypoxia and then exposed to room air (Denver altitude) for an additional three weeks. *P < 0.01 (t-test) (n = 3 per time point and experimental group). (B) Expression of a nuclear proliferation marker PCNA is minimal, if any, in medial SMCs of SU5416+Hypoxia or Control rats, as outlined in A (n = 3 per time point and experimental group). (C) Staining for MIB1 (nuclear proliferation marker) in (i) a control human PA shows rare proliferating endothelial cells (arrow) and (ii) endothelial cells in a plexiform lesion (arrow) in a lung of a patient with idiopathic pulmonary arterial hypertension. Note lack of immunoreactivity (i.e. lack of proliferation) in the SMCs of the arterial media (arrowhead). *P < 0.01 (students t-test); Adapted from reference.20

Consistent with the observations in animal models wherein proliferation, particularly in SM-actin+ cells, seems to wane with time are studies of cellular proliferation using lung tissues from both idiopathic PAH (iPAH) and familial PAH (FPAH) patients compared with controls. The authors report, based on the pathological evaluation of a large numbers of lungs with PAH (and Group 3 PH), no evidence of SMC proliferation in established PH, with no significant Ki67 expression in α-SM-actin+ SMCs.22 Instead, other cell populations, including endothelial cells and inflammatory cells that concentrate around and within the pulmonary vascular lesions in PH.18 exhibit evidence of ongoing proliferation. Interestingly, there were significant perivascular accumulations of CD45+ and CD133+ cells, observations that have been supported by other studies,23 which were proliferating but without significant fusion with recruited circulating cells.22 This is consistent with the observations of Rich et al. in a patient with iPAH, where they used double-immunolabelling techniques for α-SM-actin and PCNA (marker of cell cycle progression), and showed that proliferation occurred almost exclusively in non-α-SM-actin expressing cells.24 Collectively, these observations challenge the concept that SMC proliferation continues unabated, while raising important questions as to what are the phenotypes of the proliferating cells in chronically hypertensive vessel wall, as these appear to contribute to sustaining the large cellular mass and ultimately to vascular obstruction.

2.2 Age and development effects on SMC proliferation

There are also age and/or development specific differences in the proliferative responses of pulmonary vascular wall cells to various stimuli involved in hypoxia induced PH. In neonatal calf models, SMCs have a much higher capacity for proliferation than cells from later stages of development in both large and small vessels compared with older weanling calves.25–28 It should be noted that there is a relatively high rate of proliferation in both the media (and adventitia) at birth, which decline over the first 14 days following birth.29 If this transition is interrupted by exposure of the animals to hypobaric hypoxia, there are very significant increases in both medial and adventitial cell proliferation (assessed by labelling index), which exceed the proliferative responses usually observed in rats exposed to hypoxia, even in rapidly growing rats (e.g. 8-weeks of age). Interestingly, in this neonatal calf model, at least within the medial compartment, the highest rates of proliferation occur in small muscular arteries and not in the larger hilar elastic vessels.29 Based on observations in this calf model and in SMC responses in young vs. old rats, matched neonatal and adult bovine SMCs were isolated from young and older animals and tested for differences in many parameters associated with growth capacity. Neonatal PA SMCs were smaller, grew faster, reached a higher plateau density and were less susceptible to senescence than adult PA SMCs. Further, they were more resistant to serum withdrawal, had spontaneous autocrine growth capacity, and were more responsive to IGF-1, PMA, and the combination.25,26 Studies in older calves also provided interesting information regarding the effects of more prolonged hypoxia that suggested that with time the structural changes in the media became more and more significant and actually were associated with the failure of PA pressure to return to normal upon oxygen breathing or vasodilator administration.27,30 Interestingly, medial SMCs of proximal and intermediate arteries were occasionally found to be in ‘advanced stages of degeneration’, with observations of sarcoplasm filled with large vacuoles, perinuclear oedema, degenerating mitochondria, myelin whorls and small intracellular particles of varying size and electron density. At this stage, cells appeared compressed perhaps by the marked accumulation of increased amounts of extracellular collagen and elastin fibres in the subintima and media. Concentric laminae of basement membrane material were sometimes noted surrounding SMCs. Similar changes in SMCs have been described in humans with PAH31 and in systemic hypertension.32 These observations raise questions regarding the state of SMCs in more advanced stages of PH where vascular reactivity is lost.27,30 They also support the idea that models of PH should include the use of animals across the spectrum of developmental stages, including later stages of the life span.

2.3 Heterogenic SMC subpopulations exhibit distinct growth capabilities in vivo and in vitro

As noted earlier, the vascular media in large mammals and humans has been demonstrated to be comprised of phenotypically heterogeneous SMC populations that potentially serve diverse roles in vascular homeostasis.4In vivo, these cells have distinctly different growth responses to hypoxia and growth factors, which drive most if not all pulmonary hypertensive responses, including pulmonary vascular remodelling2,5,6,33 (Figure 3). When isolated in culture, these cell subpopulations continue, at least for the first couple of passages, to maintain their distinct phenotype, as well as to display markedly different rates of proliferation6 (see Figure 4). These phenotypically distinct vascular SM-like cell populations appear to subserve different functions within the vascular media, based on observations of distinct ion channel expression and proliferative and matrix-producing capabilities in response to various stimuli, including hypoxia.1,2,6,33–37 Because each of these populations express α-SM-actin in culture, any one of these rapidly proliferating cells could contribute to ‘the highly proliferative phenotype’ that is often said to characterize ‘SMCs’ in the patient with PH.

Figure 3.

Figure 3

Phenotypically distinct SMC populations within the pulmonary arterial media of calves exhibit markedly different proliferative responses to hypoxia. (A) Phenotypic heterogeneity of pulmonary arterial media is demonstrated by immunostaining with antibodies against SM-myosin heavy chains. (B) Double-labelling with antibodies against a nuclear proliferative marker Ki67 (red, marked by arrowheads) and SMC differentiation marker meta-vinculin (green, marked by an asterisk) demonstrates that, in the PAs of calves with hypoxia-induced PH, proliferation (Ki67-positive cells) occurs almost exclusively in a less differentiated (metavinculin-negative) SMC-like subpopulation. (C) Quantitative analysis of proliferating cells expressing two distinct phenotypes, metavinculin-positive (green bars) and metavinculin-negative (red bars) (*P < 0.001; n = 3 calves in each group at each time point). Adapted from reference.33

Figure 4.

Figure 4

Cellular composition of tunica media of large proximal PA markedly differs from that of distal PA. Proximal (main) PA (top row) is characterized by profound heterogeneity of SMC populations, as reflected in cell morphology, phenotype and proliferative capabilities. In contrast, cellular composition and functional responses (proliferation) of distal PA (bottom row) are generally uniform. In the MPA, the heterogeneous pattern of cell arrangement allows the arterial media to be subdivided into three cellular ‘layers’: subendothelial (L1), middle (L2) and outer (L3). The outer media (L3) is comprised of two differently arranged cell populations: cells forming compact ‘clusters’ (‘C’) are oriented longitudinally, and cells in ‘interstitial’ (‘I’) areas between the clusters are oriented circumferentially. Adapted from reference.4

There is evidence to support the argument that these heterogenic cells are derived from distinct lineages and are not simply a common cell, exhibiting different states of differentiation.4,38,39 Little is known regarding the mechanisms that confer unique proliferative characteristics to specific cell populations that exist in the large PAs. It has been demonstrated that less differentiated, more proliferation-prone medial cells are characterized by exuberant responses to G-protein coupled receptor (GPCR) agonists, compared with differentiated medial SMCs that do not exhibit proliferative responses to hypoxia. For example, hypoxia-induced activation of GPCR, with subsequent signalling through Gαi and Gq-mediated activation of extracellular signal regulated kinase 1/2 (ERK1/2), has been shown as necessary for hypoxia-induced proliferation in a specific subset of cells (SM-like) in the subendothelial media.2 These observations support the idea that there are differences in receptor expression and/or intracellular signalling among various medial cell types.2 Differences in endothelin production and receptor expression and responses have been described in distinct cell populations derived from the ovine PA.40,41 Changes in the engagement of intracellular signalling pathways have also been described. For instance, increases in cAMP response element-binding protein expression have been shown to function as molecular determinants of SMC proliferative capability under hypoxic conditions, as have differences in protein kinase C.42,43 Collectively, these observations support the existence of phenotypically distinct medial cell populations with unique proliferative responses to environmental stimuli; these responses are driven by membrane bound receptors sensitive to hypoxic activation that engage specific intracellular signalling pathways. Thus, in various segments of the vessel wall, there may reside distinct SMC populations that can respond differently to injury or environmental queues, and thus play diverse roles in maintaining vascular homeostasis.

Studies of cell heterogeneity in the bovine and human species are supported by recent elegant studies in mice, using clonal cell labelling strategies with multi-color reporters to probe the behaviour and potential of individual and sibling mesenchymal cells during lung development.44 These studies demonstrate a surprising diversity of mesenchymal progenitor populations with different locations, patterns of migrations, recruitment mechanisms, and lineage boundaries. Even a more recent report by Majesky et al. suggests that heterogeneity exists among vascular cells even as they differentiate into what appear to be a ‘uniform’ vascular SMC phenotype.45 Thus, future studies are needed to determine the cell-autonomous and microenvironmental cues that orchestrate an assembly of diverse SMC populations with different proliferative capabilities at a particular site. Most importantly, it will be essential to determine the fate of these cells as they cease dividing even under the continuous presence of injurious stimuli.

2.4 Heterogeneity of matrix-producing capabilities of SMCs in PH

Marked increases in accumulation of collagen, elastin, fibronectin, tenascin-C and other ECM proteins are observed in humans with PAH and in experimental animal models of PH. The cellular mechanisms responsible for these changes appear to be dependent on the species examined and resident cellular composition of the arterial wall. For instance, in the rat model of hypoxic PH (and similarly, in the mouse), adventitial thickening of the large PAs occurs early, whereas thickening of the vascular media lags behind.12,46 The observed medial thickening in rodents has been suggested to be due primarily to SMC hypertrophy and increased matrix (elastin and collagen) deposition. In contrast, hypoxic PH models of larger mammals (e.g. calf and pig) demonstrate predominating early and dramatic medial thickening,37,47 which is believed to be due, in part, to increases in proliferation and thus expansion of phenotypically distinct, less differentiated medial SMC subpopulations.33 The species-specific differences may be explained by the fact that the cellular composition of proximal arteries in large mammals (including cow, lamb, pig and human) is far more complex than that of the rodent species.4,37,38,48 The arterial media in large mammals is a complex organ-like structure with multiple phenotypically distinct SMC populations, which subserve diverse cellular functions in health and disease.4,37,38,48 These findings support the notion that the heterogeneity of SMCs in pulmonary vasculature governs, at least in part, the pattern of abnormal matrix-producing capabilities that characterize chronic hypoxic forms of PH. For example, as we have demonstrated in our previous in vitro studies, using phenotypically distinct SMC populations isolated from the MPA of chronically hypoxic hypertensive neonatal calves, less differentiated PA-SMCs exhibited higher elastogenic responses to chronic hypoxic exposure.34

Importantly, various intrinsic (genetic, developmental, and epigenetic) differences in matrix-producing capabilities, as well as local and regional phenotypic heterogeneity of PA-SMCs, also regulate the pattern of remodelling of the tunica media in PH.

3. Non-medial origins of SM-like cells in the pulmonary vascular wall

3.1 Endothelial-to-mesenchymal transition

Increases in the accumulation and persistence of SM-like cells in pulmonary circulation clearly occur in all settings of PH. However, the idea that these cells derive solely from local proliferation of one of the resident cell populations described earlier, has been challenged in recent years by experimental data demonstrating many possible sources of SM-like cells not only in the lung, but also in the heart, kidney and liver. These include endothelial cells undergoing EndoMT, resident vascular progenitor cells in the process of their differentiation, recruited circulating progenitors, and/or multifunctional pro-fibrotic/inflammatory cells (fibrocytes).49,50

The observation that certain vascular endothelial cells are capable of undergoing an EndoMT was initially demonstrated by Frid et al.51 More recently, studies by Ranchoux et al. and Hopper et al. combined to provide convincing experimental evidence that EndoMT occurs in the setting of PH in both humans and experimental animal models, and potentially constitutes a target against which specific therapeutic agents could be used to abrogate the process.52,53 Regulation of EndoMT can involve changes in growth factors, inflammatory signalling, and the mechanical in situ environment.54 At present, the best-described inducers of EndoMT are members of the transforming growth factor-beta (TGF-β) super family.55 Interestingly, these more recent studies have observed a link between BMPR2 deficiency and EndoMT. This is important because decreases in pulmonary BMPR2 expression, which can be observed not only in familial but also in acquired forms of PH, appear to precede EndoMT and severe vascular remodelling, as has been demonstrated in at least the monocrotaline model of PH.2,56 Yet, it remains unclear as to whether EndoMT is an essential and important contributor to the accumulation of SM-like cells in obstructive lung vascular lesions. For instance, histological assessment of lungs from patients with systemic sclerosis-associated PAH and from the sugen/hypoxia murine PH model, identify the presence of von Willebrand factor/α-SM-actin-positive transitional endothelial cells in only ∼5% of pulmonary vessels.57 Since, during EndoMT, endothelial markers are ultimately lost in newly generated mesenchymal cells making them almost indistinguishable from other SM-like cells, there is a possibility, as will be further discussed later, that mesenchymal cells in occlusive or even plexigenic lesions may also have been derived via the EndoMT process.

3.2 Resident and circulating stem cells

In the diseased systemic vessel wall, the notion that at least some SM-like cells arise from resident and/or circulating progenitor cells is now well established.50,58–60 Similarly, in pulmonary circulation, in the pathogenesis of vascular remodelling in PH, there is now good evidence for participation of both resident and circulating progenitors.

As noted at the beginning of this review, the appearance of new muscle in normally non-muscular regions of peripheral intra-acinar arteries seems to be universal in experimental animal models used to study PH. This was first thought to arise from multiplication of SMCs in the peripheral vasculature and their migration down the pulmonary vascular tree. However, Hislop and Reid,61 using electron microscopy, demonstrated that the new muscle more likely represented hypertrophy and/or metaplasia of SMC precursors normally present in the non-muscular regions of the artery. These pioneering observations paved the way for more recent elegant studies by the Grief lab, using a lineage tracing approach and showing that the new precursor cells participating in the extension of SM were actually a subset of PDGFR-β-expressing SM precursors.15,16 These investigators showed that, in mice, SM progenitor cells gave rise to the distal musculature as if ‘primed’ to do so.15,16 An important finding of these studies is the demonstration that the process of distal muscularization in hypoxic mice is clonal in nature; this offers unique opportunities to identify molecular signals that drive specific clones to expand. Moreover, these findings also suggest SMCs with distinct potentials for growth (like progenitor cells) vs. cell growth arrest, as seen with fully differentiated and long-lived cells.

Recently, Dierick et al. identified resident PW1+ progenitor cell populations located in both the lung parenchyma and in perivascular zones of small peripheral vessels, which show an early recruitment and a potential to differentiate into SM-like cells during chronic hypoxia-induced PH.62–64PW1 encodes a zinc finger protein that has been shown to regulate cell cycle and cell stress responses caused by inflammation and p53.6264 Interestingly, PW1 has also been shown to function as a transcription factor with a DNA-binding motif regulating a large array of genes involved in metabolic homeostasis.65 These cells are also characterized by the expression of CD34, c-Kit and PDGFRα, all markers used to identify vascular progenitor cells with the capability of differentiating into SM-like cells. Importantly, these studies confirmed a resident origin for at least these new SMCs that were not derived from circulating BM-derived progenitors. It appears that PW1 identifies multiple progenitor populations. Further studies are needed to determine the respective role of each of the PW1-expressing cell populations, with the ultimate goal of inhibiting mobilization, proliferation or differentiation of these cells as a potential therapeutic avenue to an early treatment of PH.

Davie et al.66 were among the first to examine the possibility that circulating progenitor cells contribute to the PH-associated pulmonary vascular remodelling. These initial studies documented the recruitment of cKit+ cells to the adventitial compartment but did not demonstrate unequivocally that these cells acquired SM-like properties. Shortly thereafter, Hayashida et al. used green fluorescent protein bone marrow (BM) transplanted (GFP-BMT) chimeric mice to investigate the possible role of BM-derived cells in hypoxia-induced pulmonary vascular remodelling.67 They demonstrated a significant increase in the number of GFP+ BM-derived cells in the hypoxic PA wall and showed an increase in the number of GFP/α-SMA expressing cells within the vascular media and adventitia. Subsequently, utilizing the calf model of PH, where, in the distal PA, control animals express a phenotypically uniform SMC composition, it was noted that the remodelled distal PA media of calves with severe hypoxia-induced PH comprises cells with a distinct phenotype characterized by the expression of haematopoietic (CD45), leukocyte/monocytic (CD11b, CD14), progenitor (cKit), and motility-associated (S100A4) cell markers68 (Figure 5). Consistent with these in vivo observations, primary cultures generated from the distal PA media of hypertensive calves yielded both differentiated SMCs as well as smaller, morphologically ‘rhomboidal’ cells that transiently expressed CD11b, expressed progenitor markers cKit, CD34, CD73 and, with time in culture, gained expression of α-SM-actin. It is possible that these progenitor cells account for the disease-acquired heterogeneity of the distal pulmonary vasculature.68 These SM-like cells (termed ‘R’-cells for their morphological rhomboidal appearance) exhibit higher mRNA expression for IL6, CCL2/MCP1 (Figure 6), and S100A4 (not shown) and are hyper-proliferative, even under serum-deprived conditions, compared with a population of differentiated SMCs grown from the same vessel (Figure 6). Thus, it is possible that cells from the circulation are recruited to the distal media that can differentiate to express α-SM-actin that remain distinct from resident medial SMCs. These cells also secrete growth factors including PDGF and CXCL12 that stimulate proliferation of resident SMCs ex vivo.

Figure 5.

Figure 5

Exposure of neonatal calves to chronic hypoxia induces, within the media of distal PAs, the appearance of cells phenotypically distinct from differentiated SMCs. (A) in control normoxic calves, distal arterial media is comprised of a phenotypically uniform SMC population (α-SM-actin; green). (B–F) in chronically hypoxic calves, a markedly thickened media of distal PAs is composed of a phenotypically heterogeneous cell population, where certain cells do not express α-SM-actin (B, arrows), other express low levels of α-SM-actin (C, inset), some cells express a leukocytic marker CD45 (D, small arrows pointing to the red-labelled cells; and large arrows point to cells that do not express either α-SM-actin or CD45). There are a few cells that express a progenitor marker cKit (E, green) within the media (large arrows) or at the medial-adventitial border (small arrows). Several cells within the media express S100A4 (F, arrows pointing to brown cells). (A–E) Cell nuclei are labelled in blue (DAPI). M, media; A, adventitia, AW, airway. Adapted from reference.68

Figure 6.

Figure 6

Ex vivo, morphologically ‘rhomboidal’ SM-like cells (‘R’-cells) [corresponding to cells, shown in Figure 5, expressing α-SM-actin (but not SM-myosin), cKit and S100A4] express higher mRNA levels for inflammatory mediators IL6 and CCL2/MCP1 compared with differentiated SMCs (‘S’-SMCs). Furthermore, R-cells exhibit autocrine, serum-independent growth (0.2% plasma-derived serum), whereas differentiated ‘S’-SMCs remain growth-arrested under these conditions (*P < 0.05, **P < 0.01). Adapted from reference.68

Another population of circulating progenitor cells involved in pulmonary vascular remodelling is comprised by CD133+ cells.69–72 These cells are capable of differentiating into haematopoietic, endothelial, SM, and neuronal cell types. In response to chronic hypoxia, circulating BM-derived CD34+CD133+Flk+ cells are recruited to the adventitial, medial, or intimal compartments where they assume mesenchymal, SM-like characteristics.69–72 Intriguingly, in vitro these cells have been reported to acquire the morphology and phenotype of the cells with which they are co-cultured.73 For example, when these CD133+ cells were co-cultured with human PA segments, they migrated through the intima and differentiated into SMCs.

Interestingly, transplantation of BM-derived CD133+ progenitor cells from patients with PAH into mice resulted in angio-proliferative pulmonary vascular remodelling, right ventricular failure and death.74 Recent studies have begun to elucidate the molecular mechanisms that underlie the expansion and differentiation of CD133+ cells into SM-like cells. Overexpression of glucose 6-phosphate dehydrogenase (G6PD) is required for and promotes CD133+ cell proliferation.75 G6PD activity upregulated expression of hypoxia inducible factor-1α (HIF1α), cyclin A and phospho-histone H3, thereby promoting CD133+ cell dedifferentiation and self-renewal.75 Expression of SMC contractile proteins in these cells is also dependent on G6PD activity. Interestingly, administration of dehydroepiandrostrone, which is known to block G6PD activity, inhibited accumulation of CD133+ cells around PAs and reduced formation of vascular obstructive lesions.75,76

Lastly, it should be noted, the BM-derived stem cells can effect vascular remodelling and stiffening through paracrine effects on SMCs or other cells in the vascular wall. Launay et al.77 demonstrated a causal link between BM-derived stem cells and serotonin (5-HT) in PH. They established that BM-derived stem cells are critical for PH development and further established a causal link between recruited cells and 5-HT by showing that blocking 5-HT2β receptor receptors (or genetically knocking them out specifically only in BM cells) abrogated PA development. Studies in BMPR2 mutant mice treated with 5-HT2β inhibitor also showed that 5-HT signalling in BM-derived cells is critical in PH development.78 Thus, more work interrogating BM-derived progenitor cell interactions with SMCs is needed in PH models.

4. Pro-inflammatory phenotype of SMCs in PH

It is now apparent that PH is associated with classical cellular and molecular components of inflammation, which may direct specific SMC populations in their contribution to pulmonary vascular remodelling. Current evidence supports the idea that the emergence of the hyper-proliferative SM-like cell is nearly always observed in the context of local changes in the inflammatory milieu of the vessel wall; however, this same proinflammatory pulmonary environment can shape long-lived, non-proliferative SMCs as well (see discussion below on senescent phenotype). It is increasingly clear that inflammation is observed in all described experimental animal models of PH, as well as in humans with PAH and other forms of chronic PH.50,79,80 Several studies have demonstrated that sustained hypoxia (perhaps the best studied animal model related to recruitment of inflammatory and progenitor cells) induces a robust accumulation of leukocytes and mesenchymal progenitor cells in perivascular areas of the PAs.81–83 The majority of studies in PH have demonstrated that the principal inflammatory cell types recruited to and retained in the hypoxic lung vasculature are those of mononuclear origin. Recruitment and retention of these cells are critical for hypoxia-induced pulmonary vascular remodelling, as has been demonstrated by in vivo depletion of circulating mononuclear phagocytes using liposome-encapsulated clodronate, which abrogated the pulmonary vascular remodelling.84 These observations are consistent with studies in the systemic circulation showing that macrophages play an essential role in both outward and inward remodelling.85–87 Burke et al.83 used laser capture microdissection of the remodelled PAs from hypoxic hypertensive rats and found a progressive accumulation of monocytes/macrophages and dendritic cells but only a few T-cells and no B-cells or neutrophils. Upregulation of CXCL12/SDF-1, VEGF, C5, ICAM-1, osteopontin and TGF-β preceded mononuclear cell influx. The cellular source of these cytokines was not precisely determined in this study, but was likely a combination of both medial and adventitial cells. Both SMCs and fibroblasts produce chemokines and cytokines, including IL-1β, IL-6, CCL2/MCP-1 and CXCL10 capable of recruiting and activating monocytes/macrophages. However, by and large, in both the pulmonary and systemic circulations in the disease process, the macrophage infiltrate appears largely in the adventitia, sparing the media.79,85,88,89 Studies by several laboratories support a mechanistic role for cytokines and inflammation in the disease process by showing that chemical inhibition and/or genetic knockout of CXCL12/SDF-12, C-C chemokine receptor type 5, CXCR7 and IL-1β/MyD88 all led to attenuation of hypoxia and/or monocrotaline-induced PH with reduced inflammatory cell recruitment and inflammation.90–94

It seems highly likely that the inflammatory capacities of the SM-like cells in the diseased vessel wall are heterogeneous, similar to that previously noted for proliferating SMCs, and depend on their precise origin. In support of this are observations of a cell population that accumulates in the media of the distal vasculature of hypoxic hypertensive calves, which originates from circulation yet acquires expression of α-SM-actin, and exhibits a distinct pro-inflammatory phenotype (with augmented expression of IL-6 and CCL2/MCP-1), compared with the resident SMCs within the vessel wall. It is possible that these cells contribute to the continued inflammation observed in the distal vasculature. More studies are needed to define the inflammatory capabilities of various heterogenic populations of SMCs contributing to vascular disease.

Collectively, these studies point to the fact that acute, and more importantly non-resolving, inflammation plays a prominent role in irreversible pulmonary hypertensive process. It is possible that as SMCs shift from a proliferating state to cell arrest, they are maintained in a distinct phenotype characterized by abnormal pro-inflammatory cytokine production and a locked altered metabolic state. This distinct phenotypic state, which is discussed below, has been described in so-called senescent cells; their potential critical role in chronic stages of PH is beginning to be appreciated.

5. Role of metabolism in SMC phenotypic changes

All WHO Groups of PH, but particularly those in Group 1, are characterized by a complex panvasculopathy that recapitulates features similar to those observed in cancer, including excessive proliferation, apoptosis resistance, inflammation and a dramatic remodelling of the extracellular matrix.80,95–97 Borrowing from recent advancements in cancer98–100 and immune research,101–106 it has been suggested that PH phenotypes might be, in part, explained by a substantial metabolic/pro-inflammatory reprogramming of vascular wall cells, including adventitial fibroblasts and macrophages. In cancer research, it is now accepted that most oncogenes and mutated tumour suppressor genes play a primary role in metabolic regulation.107 Conversely, aberrant metabolism can promote epigenetic control of oncogenic re-programming, even in the absence of specific mutations, in response to hypoxic or cytokine-triggered stimuli, promoting a pro-inflammatory and pro-fibrotic phenotype that favours both hyper-proliferation and prolonged survival of cancer and cancer-associated cells; of note, a similar overall picture also applies to immunity99,104,105,108 Studies on the metabolic reprogramming of activated immune cells indicate that such a complex phenotype can be observed in the absence of specific genetic mutations.104 This observation prompted cardiovascular investigators to wonder whether similar molecular underpinnings contribute to PH aetiology, fuelling the debate around the metabolic theory of PH.109–111

Importantly, a chronic shift in energy production from mitochondrial oxidative phosphorylation to glycolysis has been described to occur in SM-like cells, as well as in endothelial cells and fibroblasts. This phenotype, originally described in cancer and termed the Warburg Effect, contributes to both PA SMC hyper-proliferation (notably early in the disease) and to resistance to apoptosis (with established disease).111,112 The observed metabolic shift towards glycolysis increases the availability of non-oxidized amino acids, lipids, and sugars, which are all necessary for rapid cell proliferation. Further, it has been described that, in PH, PA SMCs have hyperpolarized mitochondria, which produce less ROS.112,113 The reported decreases in mitochondrial ROS production are associated with the well described decrease in Kv channels, and attenuated activity and expression at the plasma membrane, increased intracellular calcium levels, and increased activity of transcription factors, including nuclear factor of activated T-cell and HIF-1α, leading to increased proliferation and decreased apoptosis. Again, these changes have been extensively reviewed and are relevant not only to the hyper-proliferation state, but also to apoptosis resistance of SM-like cells, endothelial cells, and certain populations of adventitial fibroblasts.114

A potential driver of altered SMC cellular metabolism is increased TGF-β signalling, which is associated with a glycolytic shift in other contexts including cancer.115,116 Altered TGF-β signalling via BMPR2 and other genetic mutations underlie the majority of familial cases of PAH. Kumar and colleagues recently reported that inflammation drives the recruitment of thrombospondin-1 (a known activator of latent TGF-β) expressing monocytes to the pulmonary adventitia, resulting in TGF-β activation and resulting in expansion of the pulmonary vessel medial layer.117 The Isenberg group has documented another function of thrombospondin-1 is signalling through CD47 to block nitric oxide signalling, which may also lead to SMC dysfunction.118 Sheppard and colleagues also recently showed that SMC-specific deletion of the TGF-βR2 receptor blocked development of the PH phenotype in a mouse model of scleroderma.119

6. Senescence phenotype of SMCs

Given that there is no evidence of continuous SMC proliferation in PH, it is conceivable that several of the early SMC mitogenic responders to a PH stimulus may eventually acquire a senescent phenotype. Senescent cells, which are long-lived, metabolically and functionally active cells, are unable to re-enter the mitotic cycle while resisting apoptosis or other cell death processes.

Senescence refers to a characteristic behaviour, first noticed in cultured cells, of long-lived cells, which are incapable of cell proliferation. Additional characteristics include increased cell size, expression of senescence associated β galactosidase activity (SA-βgal) and the cell cycle inhibitors p16Ink4a and p21WAF1/Cip1, and potential presence of the DNA damage response (DDR).120 There are several molecular processes linked to senescence, which however cannot account to the full range of experimental observations linked to this key cellular state. The most notable is the observation of telomere shortening in cultured cells that progressed to cell growth arrest. Telomere shortening likely triggers the DDR, as could also be triggered by strong mitogenic stimuli, and therefore causes senescence. The major pathophysiological implications of cellular senescence are the cell’s inability to further proliferate and their acquisition of an inflammatory phenotype (labelled senescence-associated secretory phenotype or SASP), largely characterized by overexpression of IL-1, IL-6 and IL-8.120

At the organismal level, senescence has been observed and related to processes of aging, which can be defined as loss of fecundity, increased susceptibility to disease, and enhanced risk of death.121 In summary, these events are due to loss of homeostatic mechanisms involved in organ structure and function. The relation between senescence and aging varies, being either considered as associative or contributory.120 Many age-related diseases have increased expression of cells with markers of senescence and some experimental age-related disease can be attenuated with depletion of p16Ink4a expressing cells122; however, it is unclear if the senescent cells play a role in age-related human diseases. Moreover, senescence may represent a pathogenetically relevant in vivo response that per se may not be associated with increased age, as in diseases such as lung fibrosis and PH.

The roles of senescence in lung diseases were initially identified in chronic obstructive pulmonary disease (COPD), notably in emphysematous tissue123 with subsequent support by evidence of telomere shortening in peripheral blood mononuclear cells from COPD patients.124 More recently, the role of senescence was expanded to pulmonary fibrosis,125 including a genetic link with the discovery of telomerase mutations in idiopathic pulmonary fibrosis.126,127 A causal role of senescence in COPD and pulmonary fibrosis is anticipated given the close association with increasing age of affected individuals.128 It is therefore not surprising that PH or pulmonary vascular remodelling in the setting of COPD has been linked to SMC senescence.129 In this study, telomere length assessed in peripheral blood mononuclear cells correlated inversely with IL6 levels and PA pressures assessed by echocardiography. These findings are in line with the burden of oxidative stress in smoking (which leads to of DNA damage,130,131 adduct-modified proteins132 and telomere shortening133). As mentioned previously, DNA damage is a critical determinant of senescence.

A potential role for PA SMC senescence in PH or pulmonary vascular disease was inferred from studies on COPD patients, with some patients having echocardiography-based evidence of mild PH.129 PAs of patients with mild COPD had increased numbers of SMCs positive for SA-βgal, p16Ink4a and p21WAF1/Cip1, reaching up to almost 48% of counted cells. At the same time, almost 50% of SMCs were positive for Ki67, implying that they were undergoing mitosis. This dichotomy of SMCs either in senescence or proliferation is surprising, as one might anticipate that the senescence phenotype would be more restricted given the wide range of SMC phenotypes; moreover, it is unclear from the study which specific SMC cell populations gave rise to the senescent-marker expressing cells. The study goes on demonstrating that, in culture, SMCs originating from COPD-PAs developed senescence in earlier passages than control SMCs and produced the SASP of cytokines. These cytokines could then drive increased proliferation and migration of the non-senescent SMCs. Whether the process of cellular senescence applies to PAH (a more severe disease, usually clustered as WHO Group 1 PAH134) remains unclear.

PAH (as compared with PH associated with COPD, known as WHO Group 3 PH) is considered a more proliferative process, notably in endothelial cells as they form plexiform lesions.135 Given the evidence of increased telomerase activity in proliferative processes (like malignancy and after vascular injury), one might predict that PAH SMCs might rather have increased telomerase activity as compared with controls or PH related to COPD. In PAH, telomerase reverse transcriptase (TERT) expression was increased in CD44+ SMCs located in the outer rim of remodelled PAs, but was not expressed in α-SMC actin-positive cells or KI67-negative cells. No control SMCs expressed TERT.136 Mice knocked out for TERT or wild type mice treated with a TERT inhibitor had attenuated PA pressures when challenged with hypoxia, while having increased expression of p16Ink4a and p21WAF1/Cip1 in pulmonary vascular SMC. Of note, these results are in opposition to those presented in human studies of COPD remodelled PAs, in which senescence is increased and promotes SMC growth.129 It is therefore unclear whether, in PAH, there is cellular senescence that might drive the overall pro-remodelling phenotype seen in PAH or even in hypoxia driven PH.

In summary, the potential conflictive roles of SMC senescence may be as conflictive as those delineated in the cancer field.120 On one hand, in proliferation-prone states, like in specific SMC populations in PAH or even early hypoxic PH, senescence may be a protective mechanism by disengaging the proliferative SMC process. On the other hand, in late disease when proliferation may no longer play a critical role, SMC senescence may be pathological by promoting persistent inflammation, recruitment of progenitor cells and a switch to matrix production.

7. Metabolic characteristics of cellular senescence

As alluded previously, several key features of metabolic adaptation may participate in the phenotype of endothelial cells and SMC in PH PAs. The predominant metabolic features involve aerobic glycolysis, which have been reviewed previously.97,137,138 As outlined earlier, hypertensive SMCs utilize glycolysis to generate a pro-inflammatory feedback interactions with pulmonary vascular fibroblasts, ultimately contributing to pulmonary vascular remodelling.

Interestingly, senescent cells are largely glycolytic as well.139 This metabolic adaptation would serve, as in highly proliferating cells, to provide carbon intermediates for macromolecular synthesis, most notably of proteins and phospholipids 140 (as nucleic acids are dispensable due to growth arrest). However, the teleological reasoning behind the preferential use of glycolysis vs. mitochondria metabolism in the maintenance of senescence remains unclear. One unexplored reason would reside in the preservation of the SASP phenotype, notably of IL6 production and STAT3 activation.

Moreover, key metabolic alterations associated with or causing senescence shape the metabolic landscape of these cells. ADP and AMP levels increase in relation to ATP, causing activation of the enzyme AMPkinase. AMPkinase can promote cell growth arrest via activation of p53 and via promotion of p16Ink4a and p21WAF1/Cip1 stabilization. Moreover, the levels of NAD+ (in relation to its reduced form NADH) decrease and thus trigger a multi-pronged molecular process that favours senescence: reduction of NAD+ leads to decrease the DNA damage repair activity of PARP; furthermore, reduced NAD+ decreases the activity of SIRT 3 and 5 (which with SIRT6), which also contribute to senescence.

Mitochondria mass increases in senescent cells and plays a key role in the acquisition of the SASP phenotype, whereas depletion of mitochondria DNA, while promoting senescence, blocks SASP. However, the role of mitochondria in senescence is complex, in that ROS originating from mitochondrial oxidative phosphorylation promotes senescence, possibly via DDR.

As the role of senescence in accounting for the unique heterogeneity of SMC phenotypes in PH remains largely unexplored, the angle of the metabolic interface between the different SMC phenotypes might offer unique insights into how to dissect this complex interaction. Senescent cells define their organ microenvironment in that these cells lose proliferative and repopulation properties; however, they also affect via paracrine interactions how the microenvironment reacts to proliferative cues.

8. Conclusion

The spectrum of SMC origin and phenotype, particularly in disease, is evolving (Figure 7) and, though complex, may lend itself to potential therapeutic targeting. To reach this goal, it is critical that specific PA SMC phenotypes be ascribed specific roles in the disease process, including the temporal relation with the course of disease. Incorporation of cell fate mapping using key transcription markers characteristic of specific phenotypes allied to clonal markers, will provide important tools to delineate specific contributions and the relative important of the different types of PA SMCs. Other pulmonary fields, including those addressing origins and phenotypes of macrophages and type II cells, are paradigms that have utilized these tools, which can serve as a framework to be applied to PA SMCs. At present, we do not know of any studies, which have specifically tried to target the functional phenotype of a specific subset of SMC or SM-like cells in PH. Certainly there continues to be great interest in inhibiting SMC proliferation. Several agents have been utilized in animal models with success including inhibitors of receptor tyrosine kinase, mTOR, p38, CDK4/6 among others. Again, a subset of human iPAH patients responded clinically to Imatinib treatment. Future work will define the subset(s) of patients who may respond to these therapies at a point in time when SMC proliferation is contributing significantly to disease pathogenesis.

Figure 7.

Figure 7

Potential SMC origins and phenotypes in PH. Central image shows vascular remodelling in medium-sized PA, characterized by thickening of tunica media (arrow) with lymphocytic cluster of inflammatory cells, largely lymphocytes (short arrow).

Conflict of interest: none declared.

Funding

This work was supported by National Institutes of Health grants [R03HL133306 to B.B.G., R01HL135872 to B.B.G., R01HL114887 to K.R.S., R24HL123767 to R.M.T., P01HL014985 to B.B.G., K.R.S., R.M.T.]; and Department of Defense Grant [PR140977 to K.R.S.].

References

  • 1. Frid MG, Aldashev AA, Dempsey EC, Stenmark KR.. Smooth muscle cells isolated from discrete compartments of the mature vascular media exhibit unique phenotypes and distinct growth capabilities. Circ Res 1997;81:940–952. [DOI] [PubMed] [Google Scholar]
  • 2. Frid MG, Aldashev AA, Nemenoff RA, Higashito R, Westcott JY, Stenmark KR.. Subendothelial cells from normal bovine arteries exhibit autonomous growth and constitutively activated intracellular signaling. Arterioscler Thromb Vasc Biol 1999;19:2884–2893. [DOI] [PubMed] [Google Scholar]
  • 3. Frid MG, Dempsey EC, Durmowicz AG, Stenmark KR.. Smooth muscle cell heterogeneity in pulmonary and systemic vessels. Importance in vascular disease. Arterioscler Thromb Vasc Biol 1997;17:1203–1209. [DOI] [PubMed] [Google Scholar]
  • 4. Frid MG, Moiseeva EP, Stenmark KR.. Multiple phenotypically distinct smooth muscle cell populations exist in the adult and developing bovine pulmonary arterial media in vivo. Circ Res 1994;75:669–681. [DOI] [PubMed] [Google Scholar]
  • 5. Prosser IW, Stenmark KR, Suthar M, Crouch EC, Mecham RP, Parks WC.. Regional heterogeneity of elastin and collagen gene expression in intralobar arteries in response to hypoxic pulmonary hypertension as demonstrated by in situ hybridization. Am J Pathol 1989;135:1073–1088. [PMC free article] [PubMed] [Google Scholar]
  • 6. Stiebellehner L, Frid MG, Reeves JT, Low RB, Gnanasekharan M, Stenmark KR.. Bovine distal pulmonary arterial media is composed of a uniform population of well-differentiated smooth muscle cells with low proliferative capabilities. Am J Physiol Lung Cell Mol Physiol 2003;285:L819–L828. [DOI] [PubMed] [Google Scholar]
  • 7. Burton AC. Relation of structure to function of the tissues of the wall of blood vessels. Physiol Rev 1954;34:619–642. [DOI] [PubMed] [Google Scholar]
  • 8. Meyrick B, Reid L.. Hypoxia and incorporation of 3H-thymidine by cells of the rat pulmonary arteries and alveolar wall. Am J Pathol 1979;96:51–70. [PMC free article] [PubMed] [Google Scholar]
  • 9. Meyrick B, Reid L.. Development of pulmonary arterial changes in rats fed Crotalaria spectabilis. Am J Pathol 1979;94:37–50. [PMC free article] [PubMed] [Google Scholar]
  • 10. Meyrick B, Hislop A, Reid L.. Pulmonary arteries of the normal rat: the thick walled oblique muscle segment. J Anat 1978;125:209–221. [PMC free article] [PubMed] [Google Scholar]
  • 11. Meyrick B, Reid L.. The effect of continued hypoxia on rat pulmonary arterial circulation. An ultrastructural study. Lab Invest 1978;38:188–200. [PubMed] [Google Scholar]
  • 12. Paddenberg R, Stieger P, von Lilien AL, Faulhammer P, Goldenberg A, Tillmanns HH, Kummer W, Braun-Dullaeus RC.. Rapamycin attenuates hypoxia-induced pulmonary vascular remodeling and right ventricular hypertrophy in mice. Respir Res 2007;8:15.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Quinlan TR, Li D, Laubach VE, Shesely EG, Zhou N, Johns RA.. eNOS-deficient mice show reduced pulmonary vascular proliferation and remodeling to chronic hypoxia. Am J Physiol Lung Cell Mol Physiol 2000;279:L641–L650. [DOI] [PubMed] [Google Scholar]
  • 14. Nozik-Grayck E, Suliman HB, Majka S, Albietz J, Van Rheen Z, Roush K, Stenmark KR.. Lung EC-SOD overexpression attenuates hypoxic induction of Egr-1 and chronic hypoxic pulmonary vascular remodeling. Am J Physiol Lung Cell Mol Physiol 2008;295:L422–L430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Sheikh AQ, Lighthouse JK, Greif DM.. Recapitulation of developing artery muscularization in pulmonary hypertension. Cell Rep 2014;6:809–817. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Sheikh AQ, Misra A, Rosas IO, Adams RH, Greif DM.. Smooth muscle cell progenitors are primed to muscularize in pulmonary hypertension. Sci Transl Med 2015;7:308ra159.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Graham BB, Chabon J, Gebreab L, Poole J, Debella E, Davis L, Tanaka T, Sanders L, Dropcho N, Bandeira A, Vandivier RW, Champion HC, Butrous G, Wang XJ, Wynn TA, Tuder RM.. Transforming growth factor-beta signaling promotes pulmonary hypertension caused by Schistosoma mansoni. Circulation 2013;128:1354–1364. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Tuder RM, Groves BM, Badesch DB, Voelkel NF.. Exuberant endothelial cell growth and elements of inflammation are present in plexiform lesions of pulmonary hypertension. Am J Pathol 1994;144:275–285. [PMC free article] [PubMed] [Google Scholar]
  • 19. Stacher E, Graham BB, Hunt JM, Gandjeva A, Groshong SD, McLaughlin VV, Jessup M, Grizzle WE, Aldred MA, Cool CD, Tuder RM.. Modern age pathology of pulmonary arterial hypertension. Am J Respir Crit Care Med 2012;186:261–272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Taraseviciene-Stewart L, Kasahara Y, Alger L, Hirth P, Mc MGG, Waltenberger J, Voelkel NF, Tuder RM.. Inhibition of the VEGF receptor 2 combined with chronic hypoxia causes cell death-dependent pulmonary endothelial cell proliferation and severe pulmonary hypertension. Faseb J 2001;15:427–438. [DOI] [PubMed] [Google Scholar]
  • 21. Abe K, Toba M, Alzoubi A, Ito M, Fagan KA, Cool CD, Voelkel NF, McMurtry IF, Oka M.. Formation of plexiform lesions in experimental severe pulmonary arterial hypertension. Circulation 2010;121:2747–2754. [DOI] [PubMed] [Google Scholar]
  • 22. Majka SM, Skokan M, Wheeler L, Harral J, Gladson S, Burnham E, Loyd JE, Stenmark KR, Varella-Garcia M, West J.. Evidence for cell fusion is absent in vascular lesions associated with pulmonary arterial hypertension. Am J Physiol Lung Cell Mol Physiol 2008;295:L1028–L1039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Foris V, Kovacs G, Marsh LM, Balint Z, Totsch M, Avian A, Douschan P, Ghanim B, Klepetko W, Olschewski A, Olschewski H.. CD133+ cells in pulmonary arterial hypertension. Eur Respir J 2016;48:459–469. [DOI] [PubMed] [Google Scholar]
  • 24. Rich S, Pogoriler J, Husain AN, Toth PT, Gomberg-Maitland M, Archer SL.. Long-term effects of epoprostenol on the pulmonary vasculature in idiopathic pulmonary arterial hypertension. Chest 2010;138:1234–1239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Dempsey EC, Badesch DB, Dobyns EL, Stenmark KR.. Enhanced growth capacity of neonatal pulmonary artery smooth muscle cells in vitro: dependence on cell size, time from birth, insulin-like growth factor I, and auto-activation of protein kinase C. J Cell Physiol 1994;160:469–481. [DOI] [PubMed] [Google Scholar]
  • 26. Dempsey EC, Das M, Frid MG, Stenmark KR.. Unique growth properties of neonatal pulmonary vascular cells: importance of time- and site-specific responses, cell-cell interaction, and synergy. J Perinatol 1996;16:S2–11. [PubMed] [Google Scholar]
  • 27. Jaenke RS, Alexander AF.. Fine structural alterations of bovine peripheral pulmonary arteries in hypoxia-induced hypertension. Am J Pathol 1973;73:377–398. [PMC free article] [PubMed] [Google Scholar]
  • 28. Reeves JT, Leathers JE.. Postnatal development of pulmonary and bronchial arterial circulations in the calf and the effects of chronic hypoxia. Anat Rec 1967;157:641–655. [DOI] [PubMed] [Google Scholar]
  • 29. Belknap JK, Orton EC, Ensley B, Tucker A, Stenmark KR.. Hypoxia increases bromodeoxyuridine labeling indices in bovine neonatal pulmonary arteries. Am J Respir Cell Mol Biol 1997;16:366–371. [DOI] [PubMed] [Google Scholar]
  • 30. Alexander AF, Jensen R.. Pulmonary vascular pathology of bovine high mountain disease. Am J Vet Res 1963;24:1098–1111. [PubMed] [Google Scholar]
  • 31. Esterly JA, Glagov S, Ferguson DJ.. Morphogenesis of intimal obliterative hyperplasia of small arteries in experimental pulmonary hypertension. An ultrastructural study of the role of smooth-muscle cells. Am J Pathol 1968;52:325–347. [PMC free article] [PubMed] [Google Scholar]
  • 32. Biava C, West M.. Lipofuscin-like granules in vascular smooth muscle and juxtaglomerular cells of human kidneys. Am J Pathol 1965;47:287–313. [PMC free article] [PubMed] [Google Scholar]
  • 33. Wohrley JD, Frid MG, Moiseeva EP, Orton EC, Belknap JK, Stenmark KR.. Hypoxia selectively induces proliferation in a specific subpopulation of smooth muscle cells in the bovine neonatal pulmonary arterial media. J Clin Invest 1995;96:273–281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Durmowicz AG, Frid MG, Wohrley JD, Stenmark KR.. Expression and localization of tropoelastin mRNA in the developing bovine pulmonary artery is dependent on vascular cell phenotype. Am J Respir Cell Mol Biol 1996;14:569–576. [DOI] [PubMed] [Google Scholar]
  • 35. Durmowicz AG, Parks WC, Hyde DM, Mecham RP, Stenmark KR.. Persistence, re-expression, and induction of pulmonary arterial fibronectin, tropoelastin, and type I procollagen mRNA expression in neonatal hypoxic pulmonary hypertension. Am J Pathol 1994;145:1411–1420. [PMC free article] [PubMed] [Google Scholar]
  • 36. Stenmark KR. Cell-, age-, and phenotype-dependent differences in the control of gene expression. Am J Physiol Lung Cell Mol Physiol 2001;281:L762–L765. [DOI] [PubMed] [Google Scholar]
  • 37. Stenmark KR, Fagan KA, Frid MG.. Hypoxia-induced pulmonary vascular remodeling: cellular and molecular mechanisms. Circ Res 2006;99:675–691. [DOI] [PubMed] [Google Scholar]
  • 38. Yoshida T, Owens GK.. Molecular determinants of vascular smooth muscle cell diversity. Circ Res 2005;96:280–291. [DOI] [PubMed] [Google Scholar]
  • 39. Majesky MW. Vascular smooth muscle diversity: insights from developmental biology. Curr Atheroscler Rep 2003;5:208–213. [DOI] [PubMed] [Google Scholar]
  • 40. Tchekneva E, Lawrence ML, Meyrick B.. Cell-specific differences in ET-1 system in adjacent layers of main pulmonary artery. A new source of ET-1. Am J Physiol Lung Cell Mol Physiol 2000;278:L813–L821. [DOI] [PubMed] [Google Scholar]
  • 41. Balyakina EV, Chen D, Lawrence ML, Manning S, Parker RE, Shappell SB, Meyrick B.. ET-1 receptor gene expression and distribution in L1 and L2 cells from hypertensive sheep pulmonary artery. Am J Physiol Lung Cell Mol Physiol 2002;283:L42–L51. [DOI] [PubMed] [Google Scholar]
  • 42. Klemm DJ, Watson PA, Frid MG, Dempsey EC, Schaack J, Colton LA, Nesterova A, Stenmark KR, Reusch JE.. cAMP response element-binding protein content is a molecular determinant of smooth muscle cell proliferation and migration. J Biol Chem 2001;276:46132–46141. [DOI] [PubMed] [Google Scholar]
  • 43. Dempsey EC, Frid MG, Aldashev AA, Das M, Stenmark KR.. Heterogeneity in the proliferative response of bovine pulmonary artery smooth muscle cells to mitogens and hypoxia: importance of protein kinase C. Can J Physiol Pharmacol 1997;75:936–944. [PubMed] [Google Scholar]
  • 44. Kumar ME, Bogard PE, Espinoza FH, Menke DB, Kingsley DM, Krasnow MA.. Mesenchymal cells. Defining a mesenchymal progenitor niche at single-cell resolution. Science 2014;346:1258810.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Majesky MW, Horita H, Ostriker A, Lu S, Regan JN, Bagchi A, Dong XR, Poczobutt J, Nemenoff RA, Weiser-Evans MC.. Differentiated smooth muscle cells generate a subpopulation of resident vascular progenitor cells in the adventitia regulated by Klf4. Circ Res 2017;120:296–311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Jones R, Reid LM, Vascular remodeling in clinical and experimental pulmonary hypertension In Bishop JE, Reeves JT, Laurent GJ, eds. Pulmonary Vascular Remodeling. London: Portland Press, 1995. pp. 47–115. [Google Scholar]
  • 47. Stenmark KR, Mecham RP.. Cellular and molecular mechanisms of pulmonary vascular remodeling. Annu Rev Physiol 1997;59:89–144. [DOI] [PubMed] [Google Scholar]
  • 48. Hao H, Ropraz P, Verin V, Camenzind E, Geinoz A, Pepper MS, Gabbiani G, Bochaton PML.. Heterogeneity of smooth muscle cell populations cultured from pig coronary artery. Arterioscler Thromb Vasc Biol 2002;22:1093–1099. [DOI] [PubMed] [Google Scholar]
  • 49. Arciniegas E, Frid MG, Douglas IS, Stenmark KR.. Perspectives on endothelial-to-mesenchymal transition: potential contribution to vascular remodeling in chronic pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 2007;293:L1–L8. [DOI] [PubMed] [Google Scholar]
  • 50. Yeager ME, Frid MG, Stenmark KR.. Progenitor cells in pulmonary vascular remodeling. Pulm Circ 2011;1:3–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Frid MG, Kale VA, Stenmark KR.. Mature vascular endothelium can give rise to smooth muscle cells via endothelial-mesenchymal transdifferentiation: in vitro analysis. Circ Res 2002;90:1189–1196. [DOI] [PubMed] [Google Scholar]
  • 52. Ranchoux B, Antigny F, Rucker-Martin C, Hautefort A, Pechoux C, Bogaard HJ, Dorfmuller P, Remy S, Lecerf F, Plante S, Chat S, Fadel E, Houssaini A, Anegon I, Adnot S, Simonneau G, Humbert M, Cohen-Kaminsky S, Perros F.. Endothelial-to-mesenchymal transition in pulmonary hypertension. Circulation 2015;131:1006–1018. [DOI] [PubMed] [Google Scholar]
  • 53. Hopper RK, Moonen JR, Diebold I, Cao A, Rhodes CJ, Tojais NF, Hennigs JK, Gu M, Wang L, Rabinovitch M.. In pulmonary arterial hypertension, reduced BMPR2 promotes endothelial-to-mesenchymal transition via HMGA1 and its target slug. Circulation 2016;133:1783–1794. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Stenmark KR, Frid M, Perros F.. Endothelial-to-mesenchymal transition: an evolving paradigm and a promising therapeutic target in PAH . Circulation 2016;133:1734–1737. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Pardali E, Goumans MJ, ten Dijke P.. Signaling by members of the TGF-beta family in vascular morphogenesis and disease. Trends Cell Biol 2010;20:556–567. [DOI] [PubMed] [Google Scholar]
  • 56. Long L, Crosby A, Yang X, Southwood M, Upton PD, Kim DK, Morrell NW.. Altered bone morphogenetic protein and transforming growth factor-beta signaling in rat models of pulmonary hypertension: potential for activin receptor-like kinase-5 inhibition in prevention and progression of disease. Circulation 2009;119:566–576. [DOI] [PubMed] [Google Scholar]
  • 57. Good RB, Gilbane AJ, Trinder SL, Denton CP, Coghlan G, Abraham DJ, Holmes AM.. Endothelial to mesenchymal transition contributes to endothelial dysfunction in pulmonary arterial hypertension. Am J Pathol 2015;185:1850–1858. [DOI] [PubMed] [Google Scholar]
  • 58. Chong JJ, Reinecke H, Iwata M, Torok-Storb B, Stempien-Otero A, Murry CE.. Progenitor cells identified by PDGFR-alpha expression in the developing and diseased human heart. Stem Cells Dev 2013;22:1932–1943. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Leach DF, Nagarkatti M, Nagarkatti P, Cui T.. Functional states of resident vascular stem cells and vascular remodeling. Front Biol 2015;10:387–397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Psaltis PJ, Simari RD.. Vascular wall progenitor cells in health and disease. Circ Res 2015;116:1392–1412. [DOI] [PubMed] [Google Scholar]
  • 61. Hislop A, Reid L.. New findings in pulmonary arteries of rats with hypoxia-induced pulmonary hypertension. Br J Exp Pathol 1976;57:542–554. [PMC free article] [PubMed] [Google Scholar]
  • 62. Dierick F, Hery T, Hoareau-Coudert B, Mougenot N, Monceau V, Claude C, Crisan M, Besson V, Dorfmuller P, Marodon G, Fadel E, Humbert M, Yaniz-Galende E, Hulot JS, Marazzi G, Sassoon D, Soubrier F, Nadaud S.. Resident PW1+ progenitor cells participate in vascular remodeling during pulmonary arterial hypertension. Circ Res 2016;118:822–833. [DOI] [PubMed] [Google Scholar]
  • 63. Relaix F, Wei X, Li W, Pan J, Lin Y, Bowtell DD, Sassoon DA, Wu X.. Pw1/Peg3 is a potential cell death mediator and cooperates with Siah1a in p53-mediated apoptosis. Proc Natl Acad Sci U S A 2000;97:2105–2110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Relaix F, Wei XJ, Wu X, Sassoon DA.. Peg3/Pw1 is an imprinted gene involved in the TNF-NFkappaB signal transduction pathway. Nat Genet 1998;18:287–291. [DOI] [PubMed] [Google Scholar]
  • 65. Thiaville MM, Huang JM, Kim H, Ekram MB, Roh TY, Kim J.. DNA-binding motif and target genes of the imprinted transcription factor PEG3. Gene 2013;512:314–320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Davie NJ, Crossno JT Jr., Frid MG, Hofmeister SE, Reeves JT, Hyde DM, Carpenter TC, Brunetti JA, McNiece IK, Stenmark KR.. Hypoxia-induced pulmonary artery adventitial remodeling and neovascularization: contribution of progenitor cells. Am J Physiol Lung Cell Mol Physiol 2004;286:L668–L678. [DOI] [PubMed] [Google Scholar]
  • 67. Hayashida K, Fujita J, Miyake Y, Kawada H, Ando K, Ogawa S, Fukuda K.. Bone marrow-derived cells contribute to pulmonary vascular remodeling in hypoxia-induced pulmonary hypertension. Chest 2005;127:1793–1798. [DOI] [PubMed] [Google Scholar]
  • 68. Frid MG, Li M, Gnanasekharan M, Burke DL, Fragoso M, Strassheim D, Sylman JL, Stenmark KR.. Sustained hypoxia leads to the emergence of cells with enhanced growth, migratory, and promitogenic potentials within the distal pulmonary artery wall. Am J Physiol Lung Cell Mol Physiol 2009;297:L1059–L1072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Schwarz J. Emerging role of c-kit+ progenitor cells in pulmonary hypertension. Am J Respir Crit Care Med 2011;184:5–7. [DOI] [PubMed] [Google Scholar]
  • 70. Montani D, Perros F, Gambaryan N, Girerd B, Dorfmuller P, Price LC, Huertas A, Hammad H, Lambrecht B, Simonneau G, Launay JM, Cohen-Kaminsky S, Humbert M.. C-kit-positive cells accumulate in remodeled vessels of idiopathic pulmonary arterial hypertension. Am J Respir Crit Care Med 2011;184:116–123. [DOI] [PubMed] [Google Scholar]
  • 71. Yao W, Firth AL, Sacks RS, Ogawa A, Auger WR, Fedullo PF, Madani MM, Lin GY, Sakakibara N, Thistlethwaite PA, Jamieson SW, Rubin LJ, Yuan JX.. Identification of putative endothelial progenitor cells (CD34+CD133+Flk-1+) in endarterectomized tissue of patients with chronic thromboembolic pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 2009;296:L870–L878. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Toshner M, Voswinckel R, Southwood M, Al-Lamki R, Howard LS, Marchesan D, Yang J, Suntharalingam J, Soon E, Exley A, Stewart S, Hecker M, Zhu Z, Gehling U, Seeger W, Pepke-Zaba J, Morrell NW.. Evidence of dysfunction of endothelial progenitors in pulmonary arterial hypertension. Am J Respir Crit Care Med 2009;180:780–787. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Diez M, Barbera JA, Ferrer E, Fernandez-Lloris R, Pizarro S, Roca J, Peinado VI.. Plasticity of CD133+ cells: role in pulmonary vascular remodeling. Cardiovasc Res 2007;76:517–527. [DOI] [PubMed] [Google Scholar]
  • 74. Asosingh K, Farha S, Lichtin A, Graham B, George D, Aldred M, Hazen SL, Loyd J, Tuder R, Erzurum SC.. Pulmonary vascular disease in mice xenografted with human BM progenitors from patients with pulmonary arterial hypertension. Blood 2012;120:1218–1227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Chettimada S, Joshi SR, Alzoubi A, Gebb SA, McMurtry IF, Gupte R, Gupte SA.. 6-phosphate dehydrogenase plays a critical role in hypoxia-induced CD133+ progenitor cells self-renewal and stimulates their accumulation in the lungs of pulmonary hypertensive rats. Am J Physiol Lung Cell Mol Physiol 2014;307:L545–L556. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Chettimada S, Gupte R, Rawat D, Gebb SA, McMurtry IF, Gupte SA.. Hypoxia-induced glucose-6-phosphate dehydrogenase overexpression and -activation in pulmonary artery smooth muscle cells: implication in pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 2015;308:L287–L300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Launay JM, Herve P, Callebert J, Mallat Z, Collet C, Doly S, Belmer A, Diaz SL, Hatia S, Cote F, Humbert M, Maroteaux L.. Serotonin 5-HT2B receptors are required for bone-marrow contribution to pulmonary arterial hypertension. Blood 2012;119:1772–1780. [DOI] [PubMed] [Google Scholar]
  • 78. West JD, Carrier EJ, Bloodworth NC, Schroer AK, Chen P, Ryzhova LM, Gladson S, Shay S, Hutcheson JD, Merryman WD, Kuwana M.. Serotonin 2B receptor antagonism prevents heritable pulmonary arterial hypertension. PLoS One 2016;11:e0148657.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Stenmark KR, Meyrick B, Galie N, Mooi WJ, McMurtry IF.. Animal models of pulmonary arterial hypertension: the hope for etiological discovery and pharmacological cure. Am J Physiol Lung Cell Mol Physiol 2009;297:L1013–L1032. [DOI] [PubMed] [Google Scholar]
  • 80. Tuder RM, Archer SL, Dorfmuller P, Erzurum SC, Guignabert C, Michelakis E, Rabinovitch M, Schermuly R, Stenmark KR, Morrell NW.. Relevant issues in the pathology and pathobiology of pulmonary hypertension. J Am Coll Cardiol 2013;62:D4–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Yu L, Hales CA.. Silencing of sodium-hydrogen exchanger 1 attenuates the proliferation, hypertrophy, and migration of pulmonary artery smooth muscle cells via E2F1. Am J Respir Cell Mol Biol 2011;45:923–930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Farha S, Asosingh K, Xu W, Sharp J, George D, Comhair S, Park M, Tang WH, Loyd JE, Theil K, Tubbs R, Hsi E, Lichtin A, Erzurum SC.. Hypoxia-inducible factors in human pulmonary arterial hypertension: a link to the intrinsic myeloid abnormalities. Blood 2011;117:3485–3493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Burke DL, Frid MG, Kunrath CL, Karoor V, Anwar A, Wagner BD, Strassheim D, Stenmark KR.. Sustained hypoxia promotes the development of a pulmonary artery-specific chronic inflammatory microenvironment. Am J Physiol Lung Cell Mol Physiol 2009;297:L238–L250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Frid MG, Brunetti JA, Burke DL, Carpenter TC, Davie NJ, Reeves JT, Roedersheimer MT, van Rooijen N, Stenmark KR.. Hypoxia-induced pulmonary vascular remodeling requires recruitment of circulating mesenchymal precursors of a monocyte/macrophage lineage. Am J Pathol 2006;168:659–669. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Tellides G, Pober JS.. Inflammatory and immune responses in the arterial media. Circ Res 2015;116:312–322. [DOI] [PubMed] [Google Scholar]
  • 86. Tang PC, Qin L, Zielonka J, Zhou J, Matte-Martone C, Bergaya S, van Rooijen N, Shlomchik WD, Min W, Sessa WC, Pober JS, Tellides G.. MyD88-dependent, superoxide-initiated inflammation is necessary for flow-mediated inward remodeling of conduit arteries. J Exp Med 2008;205:3159–3171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Zhou J, Tang PC, Qin L, Gayed PM, Li W, Skokos EA, Kyriakides TR, Pober JS, Tellides G.. CXCR3-dependent accumulation and activation of perivascular macrophages is necessary for homeostatic arterial remodeling to hemodynamic stresses. J Exp Med 2010;207:1951–1966. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Kuang SQ, Geng L, Prakash SK, Cao JM, Guo S, Villamizar C, Kwartler CS, Peters AM, Brasier AR, Milewicz DM.. Aortic remodeling after transverse aortic constriction in mice is attenuated with AT1 receptor blockade. Arterioscler Thromb Vasc Biol 2013;33:2172–2179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Savai R, Pullamsetti SS, Kolbe J, Bieniek E, Voswinckel R, Fink L, Scheed A, Ritter C, Dahal BK, Vater A, Klussmann S, Ghofrani HA, Weissmann N, Klepetko W, Banat GA, Seeger W, Grimminger F, Schermuly RT.. Immune and inflammatory cell involvement in the pathology of idiopathic pulmonary arterial hypertension. Am J Respir Crit Care Med 2012;186:897–908. [DOI] [PubMed] [Google Scholar]
  • 90. Amsellem V, Lipskaia L, Abid S, Poupel L, Houssaini A, Quarck R, Marcos E, Mouraret N, Parpaleix A, Bobe R, Gary-Bobo G, Saker M, Dubois-Randé J-L, Gladwin MT, Norris KA, Delcroix M, Combadière C, Adnot S.. CCR5 as a treatment target in pulmonary arterial hypertension. Circulation 2014;130:880–891. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Gambaryan N, Perros F, Montani D, Cohen-Kaminsky S, Mazmanian M, Renaud JF, Simonneau G, Lombet A, Humbert M.. Targeting of c-kit+ haematopoietic progenitor cells prevents hypoxic pulmonary hypertension. Eur Respir J 2011;37:1392–1399. [DOI] [PubMed] [Google Scholar]
  • 92. Sartina E, Suguihara C, Ramchandran S, Nwajei P, Rodriguez M, Torres E, Hehre D, Devia C, Walters MJ, Penfold ME, Young KC.. Antagonism of CXCR7 attenuates chronic hypoxia-induced pulmonary hypertension. Pediatr Res 2012;71:682–688. [DOI] [PubMed] [Google Scholar]
  • 93. Young KC, Torres E, Hatzistergos KE, Hehre D, Suguihara C, Hare JM.. Inhibition of the SDF-1/CXCR4 axis attenuates neonatal hypoxia-induced pulmonary hypertension. Circ Res 2009;104:1293–1301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Parpaleix A, Amsellem V, Houssaini A, Abid S, Breau M, Marcos E, Sawaki D, Delcroix M, Quarck R, Maillard A, Couillin I, Ryffel B, Adnot S.. Role of interleukin-1 receptor 1/MyD88 signalling in the development and progression of pulmonary hypertension. Eur Respir J 2016;48:470–483. [DOI] [PubMed] [Google Scholar]
  • 95. Cottrill KA, Chan SY.. Metabolic dysfunction in pulmonary hypertension: the expanding relevance of the Warburg effect. Eur J Clin Invest 2013;43:855–865. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Hanahan D, Weinberg RA.. Hallmarks of cancer: the next generation. Cell 2011;144:646–674. [DOI] [PubMed] [Google Scholar]
  • 97. Tuder RM, Davis LA, Graham BB.. Targeting energetic metabolism. Am J Respir Crit Care Med 2012;185:260–266. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Doherty JR, Cleveland JL.. Targeting lactate metabolism for cancer therapeutics. J Clin Invest 2013;123:3685–3692. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Martinez-Outschoorn UE, Lisanti MP, Sotgia F.. Catabolic cancer-associated fibroblasts transfer energy and biomass to anabolic cancer cells, fueling tumor growth. Semin Cancer Biol 2014;25:47–60. [DOI] [PubMed] [Google Scholar]
  • 100. Wellen KE, Thompson CB.. A two-way street: reciprocal regulation of metabolism and signalling. Nat Rev Mol Cell Biol 2012;13:270–276. [DOI] [PubMed] [Google Scholar]
  • 101. Marelli-Berg FM, Fu H, Mauro C.. Molecular mechanisms of metabolic reprogramming in proliferating cells: implications for T-cell-mediated immunity. Immunology 2012;136:363–369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Mills E, O’Neill LA.. Succinate: a metabolic signal in inflammation. Trends Cell Biol 2014;24:313–320. [DOI] [PubMed] [Google Scholar]
  • 103. Mills EL, Kelly B, Logan A, Costa ASH, Varma M, Bryant CE, Tourlomousis P, Däbritz JHM, Gottlieb E, Latorre I, Corr SC, McManus G, Ryan D, Jacobs HT, Szibor M, Xavier RJ, Braun T, Frezza C, Murphy MP, O’neill LA.. Succinate dehydrogenase supports metabolic repurposing of mitochondria to drive inflammatory macrophages. Cell 2016;167:457–470. e413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. O'Neill LA, Kishton RJ, Rathmell JA.. Guide to immunometabolism for immunologists. Nat Rev Immunol 2016;16:553–565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. O'Neill LA, Pearce EJ.. Immunometabolism governs dendritic cell and macrophage function. J Exp Med 2016;213:15–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Wang R, Dillon CP, Shi LZ, Milasta S, Carter R, Finkelstein D, McCormick LL, Fitzgerald P, Chi H, Munger J, Green DR.. The transcription factor Myc controls metabolic reprogramming upon T lymphocyte activation. Immunity 2011;35:871–882. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Levine AJ, Puzio-Kuter AM.. The control of the metabolic switch in cancers by oncogenes and tumor suppressor genes. Science 2010;330:1340–1344. [DOI] [PubMed] [Google Scholar]
  • 108. Hirschey MD, DeBerardinis RJ, Diehl AM, Drew JE, Frezza C, Green MF, Jones LW, Ko YH, Le A, Lea MA, Locasale JW, Longo VD, Lyssiotis CA, McDonnell E, Mehrmohamadi M, Michelotti G, Muralidhar V, Murphy MP, Pedersen PL, Poore B, Raffaghello L, Rathmell JC, Sivanand S, Vander Heiden MG, Wellen KE, Target VT.. Dysregulated metabolism contributes to oncogenesis. Semin Cancer Biol 2015;35:S129–S150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. El Kasmi KC, Stenmark KR.. Contribution of metabolic reprogramming to macrophage plasticity and function. Semin Immunol 2015;27:267–275. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Paulin R, Michelakis ED.. The metabolic theory of pulmonary arterial hypertension. Circ Res 2014;115:148–164. [DOI] [PubMed] [Google Scholar]
  • 111. Sutendra G, Michelakis ED.. The metabolic basis of pulmonary arterial hypertension. Cell Metab 2014;19:558–573. [DOI] [PubMed] [Google Scholar]
  • 112. Ryan J, Dasgupta A, Huston J, Chen KH, Archer SL.. Mitochondrial dynamics in pulmonary arterial hypertension. J Mol Med 2015;93:229–242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113. Dromparis P, Michelakis ED.. Mitochondria in vascular health and disease. Annu Rev Physiol 2013;75:95–126. [DOI] [PubMed] [Google Scholar]
  • 114. D’Alessandro A, El Kasmi K, Plecita-Hlavata L, Jezek P, Li M, Zhang H, Gupte SA, Stenmark KR.. Hallmarks of pulmonary hypertension: mesenchymal and inflammatory cell metabolic reprogramming. Antioxid Redox Signal 2018;28:230–250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Guido C, Whitaker-Menezes D, Capparelli C, Balliet R, Lin Z, Pestell RG, Howell A, Aquila S, Ando S, Martinez-Outschoorn U, Sotgia F, Lisanti MP.. Metabolic reprogramming of cancer-associated fibroblasts by TGF-beta drives tumor growth: connecting TGF-beta signaling with “Warburg-like” cancer metabolism and L-lactate production. Cell Cycle 2012;11:3019–3035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Jiang L, Deberardinis R, Boothman DA.. The cancer cell ‘energy grid’: tGF-beta1 signaling coordinates metabolism for migration. Mol Cell Oncol 2015;2:e981994.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Kumar R, Mickael C, Kassa B, Gebreab L, Robinson JC, Koyanagi DE, Sanders L, Barthel L, Meadows C, Fox D, Irwin D, Li M, McKeon BA, Riddle S, Dale Brown R, Morgan LE, Evans CM, Hernandez-Saavedra D, Bandeira A, Maloney JP, Bull TM, Janssen WJ, Stenmark KR, Tuder RM, Graham BB.. TGF-beta activation by bone marrow-derived thrombospondin-1 causes Schistosoma- and hypoxia-induced pulmonary hypertension. Nat Commun 2017;8:15494.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Rogers NM, Sharifi-Sanjani M, Yao M, Ghimire K, Bienes-Martinez R, Mutchler SM, Knupp HE, Baust J, Novelli EM, Ross M, St Croix C, Kutten JC, Czajka CA, Sembrat JC, Rojas M, Labrousse-Arias D, Bachman TN, Vanderpool RR, Zuckerbraun BS, Champion HC, Mora AL, Straub AC, Bilonick RA, Calzada MJ, Isenberg JS.. TSP1-CD47 signaling is upregulated in clinical pulmonary hypertension and contributes to pulmonary arterial vasculopathy and dysfunction. Cardiovasc Res 2017;113:15–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Tsujino K, Reed NI, Atakilit A, Ren X, Sheppard D.. Transforming growth factor-beta plays divergent roles in modulating vascular remodeling, inflammation, and pulmonary fibrosis in a murine model of scleroderma. Am J Physiol Lung Cell Mol Physiol 2017;312:L22–L31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120. Rodier F, Campisi J.. Four faces of cellular senescence. J Cell Biol 2011;192:547–556. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121. Rando TA, Chang HY.. Aging, rejuvenation, and epigenetic reprogramming: resetting the aging clock. Cell 2012;148:46–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Jeon OH, Kim C, Laberge RM, Demaria M, Rathod S, Vasserot AP, Chung JW, Kim DH, Poon Y, David N, Baker DJ, van Deursen JM, Campisi J, Elisseeff JH.. Local clearance of senescent cells attenuates the development of post-traumatic osteoarthritis and creates a pro-regenerative environment. Nat Med 2017;23:775–781. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. Aoshiba K, Nagai A.. Senescence hypothesis for the pathogenetic mechanism of chronic obstructive pulmonary disease. Proc Am Thorac Soc 2009;6:596–601. [DOI] [PubMed] [Google Scholar]
  • 124. Savale L, Chaouat A, Bastuji-Garin S, Marcos E, Boyer L, Maitre B, Sarni M, Housset B, Weitzenblum E, Matrat M, Le Corvoisier P, Rideau D, Boczkowski J, Dubois-Randé J-L, Chouaid C, Adnot S.. Shortened telomeres in circulating leukocytes of patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med 2009;179:566–571. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125. Thannickal VJ, Murthy M, Balch WE, Chandel NS, Meiners S, Eickelberg O, Selman M, Pardo A, White ES, Levy BD, Busse PJ, Tuder RM, Antony VB, Sznajder JI, Budinger GR.. Blue journal conference. Aging and susceptibility to lung disease. Am J Respir Crit Care Med 2015;191:261–269. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Armanios MY, Chen JJ, Cogan JD, Alder JK, Ingersoll RG, Markin C, Lawson WE, Xie M, Vulto I, Phillips JA III, Lansdorp PM, Greider CW, Loyd JE.. Telomerase mutations in families with idiopathic pulmonary fibrosis. N Engl J Med 2007;356:1317–1326. [DOI] [PubMed] [Google Scholar]
  • 127. Alder JK, Chen JJ-L, Lancaster L, Danoff S, Su S-C, Cogan JD, Vulto I, Xie M, Qi X, Tuder RM, Phillips JA, Lansdorp PM, Loyd JE, Armanios MY.. Short telomeres are a risk factor for idiopathic pulmonary fibrosis. Proc Natl Acad Sci U S A 2008;105:13051–13056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128. Tuder RM. Aging and cigarette smoke: fueling the fire. Am J Respir Crit Care Med 2006;174:490–491. [DOI] [PubMed] [Google Scholar]
  • 129. Noureddine H, Gary-Bobo G, Alifano M, Marcos E, Saker M, Vienney N, Amsellem V, Maitre B, Chaouat A, Chouaid C, Dubois-Rande JL, Damotte D, Adnot S.. Pulmonary artery smooth muscle cell senescence is a pathogenic mechanism for pulmonary hypertension in chronic lung disease. Circ Res 2011;109:543–553. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130. Deslee G, Woods JC, Moore C, Conradi SH, Gierada DS, Atkinson JJ, Battaile JT, Liu L, Patterson GA, Adair-Kirk TL, Holtzman MJ, Pierce RA.. Oxidative damage to nucleic acids in severe emphysema. Chest 2009;135:965–974. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131. Pastukh VM, Zhang L, Ruchko MV, Gorodnya O, Bardwell GC, Tuder RM, Gillespie MN.. Oxidative DNA damage in lung tissue from patients with COPD is clustered in functionally significant sequences. Int J Chron Obstruct Pulmon Dis 2011;6:209–217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132. Rahman I, van Schadewijk AA, Crowther AJ, Hiemstra PS, Stolk J, MacNee W, De Boer WI.. 4-Hydroxy-2-nonenal, a specific lipid peroxidation product, is elevated in lungs of patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med 2002;166:490–495. [DOI] [PubMed] [Google Scholar]
  • 133. Serra V, Grune T, Sitte N, Saretzki G, von Zglinicki T.. Telomere length as a marker of oxidative stress in primary human fibroblast cultures. Ann N Y Acad Sci 2000;908:327–330. [DOI] [PubMed] [Google Scholar]
  • 134. Simonneau G, Robbins IM, Beghetti M, Channick RN, Delcroix M, Denton CP, Elliott CG, Gaine SP, Gladwin MT, Jing ZC, Krowka MJ, Langleben D, Nakanishi N, Souza R.. Updated clinical classification of pulmonary hypertension. J Am Coll Cardiol 2009;54:S43–S54. [DOI] [PubMed] [Google Scholar]
  • 135. Tuder RM, Stacher E, Robinson J, Kumar R, Graham BB.. Pathology of pulmonary hypertension. Clin Chest Med 2013;34:639–650. [DOI] [PubMed] [Google Scholar]
  • 136. Mouraret N, Houssaini A, Abid S, Quarck R, Marcos E, Parpaleix A, Gary-Bobo G, Dubois-Rande JL, Derumeaux G, Boczkowski J, Delcroix M, Blasco MA, Lipskaia L, Amsellem V, Adnot S.. Role for telomerase in pulmonary hypertension. Circulation 2015;131:742–755. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137. Archer SL, Gomberg-Maitland M, Maitland ML, Rich S, Garcia JG, Weir EK.. Mitochondrial metabolism, redox signaling, and fusion: a mitochondria-ROS-HIF-1alpha-Kv1.5 O2-sensing pathway at the intersection of pulmonary hypertension and cancer. Am J Physiol Heart Circ Physiol 2008;294:H570–H578. [DOI] [PubMed] [Google Scholar]
  • 138. Sutendra G, Michelakis ED.. Pulmonary arterial hypertension: challenges in translational research and a vision for change. Sci Transl Med 2013;5:208sr5.. [DOI] [PubMed] [Google Scholar]
  • 139. Wiley CD, Campisi J.. From Ancient Pathways to Aging Cells-Connecting Metabolism and Cellular Senescence. Cell Metab 2016;23:1013–1021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140. Lunt SY, Vander Heiden MG.. Aerobic glycolysis: meeting the metabolic requirements of cell proliferation. Annu Rev Cell Dev Biol 2011;27:441–464. [DOI] [PubMed] [Google Scholar]

Articles from Cardiovascular Research are provided here courtesy of Oxford University Press

RESOURCES