Skip to main content
Pulmonary Circulation logoLink to Pulmonary Circulation
. 2016 Dec;6(4):448–464. doi: 10.1086/688908

Transcription factors, transcriptional coregulators, and epigenetic modulation in the control of pulmonary vascular cell phenotype: therapeutic implications for pulmonary hypertension (2015 Grover Conference series)

Soni S Pullamsetti 1,2,, Frédéric Perros 3, Prakash Chelladurai 1, Jason Yuan 4, Kurt Stenmark 5
PMCID: PMC5210074  PMID: 28090287

Abstract Abstract

Pulmonary hypertension (PH) is a complex and multifactorial disease involving genetic, epigenetic, and environmental factors. Numerous stimuli and pathological conditions facilitate severe vascular remodeling in PH by activation of a complex cascade of signaling pathways involving vascular cell proliferation, differentiation, and inflammation. Multiple signaling cascades modulate the activity of certain sequence-specific DNA-binding transcription factors (TFs) and coregulators that are critical for the transcriptional regulation of gene expression that facilitates PH-associated vascular cell phenotypes, as demonstrated by several studies summarized in this review. Past studies have largely focused on the role of the genetic component in the development of PH, while the presence of epigenetic alterations such as microRNAs, DNA methylation, histone levels, and histone deacetylases in PH is now also receiving increasing attention. Epigenetic regulation of chromatin structure is also recognized to influence gene expression in development or disease states. Therefore, a complete understanding of the mechanisms involved in altered gene expression in diseased cells is vital for the design of novel therapeutic strategies. Recent technological advances in DNA sequencing will provide a comprehensive improvement in our understanding of mechanisms involved in the development of PH. This review summarizes current concepts in TF and epigenetic control of cell phenotype in pulmonary vascular disease and discusses the current issues and possibilities in employing potential epigenetic or TF-based therapies for achieving complete reversal of PH.

Keywords: transcription factors, epigenetics, pulmonary arterial hypertension, histone deacetylases, epigenetics/transcription factor–based therapies


Transcription factors (TFs) are sequence-specific DNA-binding proteins that control the process of transcription. They contain one or more DNA-binding domains, which help them to attach to specific sequences of DNA adjacent to genes. TFs typically regulate gene expression by binding regulatory DNA elements called enhancers, an event that recruits cofactors, the general transcriptional machinery, and Pol II (RNA polymerase II) complexes to target genes.1-3 An active enhancer typically binds multiple TFs in a cooperative fashion and regulates transcription from core promoters, often via long-range genomic interactions that involve looping of DNA.3 In addition, TFs can bind directly to core promoter elements in proximity to transcriptional start sites to recruit transcriptional machinery and regulate gene expression.4

TFs are classified into two broad categories based on the functions they are involved in: (1) general TFs that are involved in basal transcriptional regulation and (2) regulatory, or gene-specific, TFs that contribute to differential gene expression. Regulatory TFs exert control over just one or a few genes and determine whether the gene is switched “on” or “off” by binding to regulatory sites, which may be located some distance from the coding region. They are crucial in ensuring that genes are expressed in the right cell at the right time and in the right amount, depending on the changing requirements of the organism.5,6

Growing evidence indicates that TFs require other molecules (e.g., coactivators and repressors, hereafter called “coregulators”) to regulate their activities.7 While it was logical to initially consider that there might be distinct use of specific coregulator complexes by distinct classes of TFs or by different members of a DNA-binding TF family, what has emerged is evidence of combinatorial usage.1,6,8 For example, many of the cofactors initially identified were based on their interactions with nuclear receptors, which have been found to play significant roles in mediating the actions of numerous classes of TFs.9 Conversely, specific TFs can use distinct combinations of cofactors, depending on cell type, promoter, DNA binding site, and the actions of various signaling pathways/ligands. Thus, coactivators/corepressors and sequence-specific TFs constitute distinct axes on a matrix for many potential combinatorial interactions that are used in a context-dependent manner to control functional gene expression.8,9 Recent advances suggest that many human diseases and disorders are associated with misregulation in TFs and TF coregulators, chromatin regulators, and noncoding RNAs (ncRNAs).

Importance of TFs and TF coregulators in the pathogenesis of pulmonary hypertension

Pulmonary hypertension (PH) is a deadly disease characterized by vasoconstriction and abnormal remodeling of pulmonary vessels, leading to a progressive increase in pulmonary artery pressure (PAP), and vascular stiffness, which culminates in right ventricular (RV) failure and premature death.10 Broadly speaking, upon vascular injury, deregulation of normal cellular processes in the pulmonary vasculature mediates proliferation, differentiation, inflammation, and cell death programs, which all together contribute to the development of PH. Numerous stimuli and pathological conditions (shear stress, hypoxia, oxidative stress, infection, HIV, and others) can result in PH by activating a complex cascade of signaling pathways.11,12 Ultimately, different signaling cascades converge on a common program targeting the activity of certain TFs.13 This leads to activation of a vascular gene program in the nucleus that is manifested finally as the PH vascular phenotype. An understanding of the altered gene expression in diseased cells lays the foundation for novel therapeutic strategies involving manipulation of gene expression. Targeting TFs represents one such innovation that can allow manipulation of gene activation and suppression in a specific fashion.

Strategies targeting single growth factors, cytokines, or receptor tyrosine kinases (RTKs) might exhibit limited efficacy because of the redundancy of the multiple stimuli that activate pulmonary vascular endothelial, smooth muscle, and fibroblast proliferation, as is observed in this progressive disease, which exhibits certain overlapping characteristics with cancer. One strategy to overcome these limitations might be to target multiple RTKs in parallel; however, a broad side-effect profile may be the consequence. A further novel and emerging concept is to target central downstream effector molecules that integrate the multiple signals. Along this line, targeting TFs might hold promise to be effective in interfering with multifactorial disease processes, that is, to combine strong antiproliferative, anti-inflammatory, and proapoptotic effects with a high specificity for activated vascular smooth muscle cells (SMCs), activated adventitial fibroblasts, and degenerative endothelial cells.

Brief review of TFs implicated in PH

There are numerous TFs and transcriptional coactivators that have been implicated in PH and RV dysfunction (Table 1; Fig. 1). In this review, we focus on the TFs Forkhead box O (FoxO) and CBF1/RBP-Jκ (recombination signal binding protein for immunoglobulin kappa J region), hypoxic inducible factors (HIFs), the transcriptional coactivator pyruvate kinase isozyme PKM2, the corepressor CtBP1 (a member of the C-terminal binding protein [CtBP] family), and the Twist family bHLH transcription factor 1 (TWIST1).

Table 1.

Transcription factors in PH

Gene Full name Role in PH Cells/tissue samples investigated in PH References
PPARG1 Peroxisome proliferator–activated receptor γ Decreased expression in PH; PPARγ agonists might reverse pulmonary vascular remodeling IPAH lungs, SuHx-PH Ameshima et al.;14 Hansmann et al.15
HIF2A Hypoxia-inducible factor 2α Hif2a/Epas1 heterozygous mice were fully protected against PH and RVH; HIF2A-mediated upregulation of vasoconstrictors contributes to the development of hypoxic PH Rodent HPH Brusselmans et al.16
HIF1A Hypoxia-inducible factor 1α Normoxic HIF activation in FHR and human PAs from IPAH; partial deficiency of either HIF1α- or HIF2α-attenuated PAP and RVH Rodent HPH, FHR-PH, IPAH PASMCs Bonnet et al.17
NFATc2 Nuclear factor of activated T-cells, cytoplasmic, calcineurin-dependent 2 Activated in CD3+ lymphocytes; NFAT inhibition reverses MCT-induced established PAH IPAH PASMCs MCT-PH Bonnet et al.18
STAT3 Signal transducers and activators of transcription 3 Persistent activation of STAT3 was identified in IPAH PAECs IPAH Masri et al.19
HES5 Hes family bHLH transcription factor 5 High levels of HES5 in lung tissue correlated with worsening disease severity IPAH, rodent HPH Li et al.20
Smad8/Smad9 Mothers against decapentaplegic homolog 8 Abnormal vascular remodeling in Smad8 mutants Transgenic Huang et al.21
OCT4 Octamer-binding transcription factor 4 OCT4 isoforms were upregulated in PASMCs from human PAH IPAH PASMCs Firth et al.22
NFATc3 Nuclear factor of activated T-cells, cytoplasmic, calcineurin-dependent 3 Activated in neonatal mice exposed to chronic hypoxia; Nfatc3 KO mice do not show PA remodeling Rodent HPH Bierer et al.23
KLF5 Krüppel-like factor 5 KLF5 is upregulated in human and MCT-induced PAH IPAH lungs and PASMCs Courboulin et al.24
P53 Tumor protein p53 Gene deficiency promotes HPH and vascular remodeling Rodent HPH Mizuno et al.25
Lef1 Lymphoid enhancer factor 1 Upregulated in HPH in neonatal rats Rodent HPH Xu et al.26
PPARGC1A (PGC-1α) Peroxisome proliferator-activated receptor gamma coactivator 1α Upregulated mRNA expression IPAH blood samples Mata et al.27
NF-KB/RELA Nuclear factor κ-B NF-κB is activated in pulmonary macrophages, lymphocytes, endothelial cells, and PASMCs IPAH Price et al.28
GATA6 GATA-binding protein 6 Endothelial GATA-6 deficiency promotes PAH SSc-PAH, IPAH Ghatnekar et al.29
c-JUN, c-FOS Jun proto-oncogene, FBJ murine osteosarcoma viral oncogene homolog Higher levels of total and phosphorylated c-JUN and c-FOS in the vessel wall IPAH lungs Biasin et al.30
FOXO1 Forkhead box O1 FOXO1 expression is downregulated and inactivated; FOXO1 activation reverses the development of PH IPAH, MCT-PH, SuHx-PH Savai et al.31
KLF4 Krüppel-like factor 4 KLF4 gene and protein expression is reduced in lungs from patients with PAH; loss of endothelial KLF4 exacerbates pulmonary vessel muscularization IPAH lungs Shatat et al.32
MEF2 Myocyte enhancer factor 2 MEF2 transcriptional activity is impaired IPAH/FPAH PAECs Kim et al.33
TWIST1 Twist family bHLH transcription factor 1 EndoMT; neointimal and plexiform lesion formation; role in neomuscularization PAH, MCT-PH, SuHx-PH, Bmpr2+/− rats Ranchoux et al.34
SLUG Snail family zinc finger 2 EndoMT BMPR2 KO PAECs Hopper et al.35
Note

bHLH: basic helix-loop-helix; EndoMT: endothelial-to-mesenchymal transition; FHR: fawn-hooded rat; HPH: hypoxia-induced PH; IPAH: idiopathic PAH; KO: knockout; MCT-PH: monocrotaline-induced PH; mRNA: messenger RNA; PA: pulmonary artery; PAECs: pulmonary arterial endothelial cells; PAH: pulmonary arterial hypertension; PAP: pulmonary artery pressure; PASMCs: pulmonary artery smooth muscle cells; PH: pulmonary hypertension; RVH: right ventricular hypertrophy; SSc-PAH: systemic sclerosis–associated PAH; SuHX-PH: hypoxia+SU5416–induced PAH (SU5416 is a vascular endothelial growth factor receptor [VEGFR-2] inhibitor).

Figure 1.

Figure 1

Role of transcription factors in the pathogenesis of PAH. Multiple pathological stimuli, such as hypoxia, shear stress, oxidative stress, mitogens, and inflammation (cytokines and chemokines), trigger downstream signaling cascades (MAPK, JNK, PI3K-Akt, JAK-STAT, ERK, PKA, PKG, PKC, etc.), which modulate the recruitment and activation of transcription factors that determine the stimulus-specific transcriptional responses in PAH. ERK: extracellular signal–regulated kinase; JAK-STAT: Janus kinase–signal transducer and activator of transcription; JNK: c-Jun N-terminal kinases; MAPK: mitogen-activated protein kinase; MEK: mitogen-activated protein kinase kinase; PAH: pulmonary arterial hypertension; PI3K-Akt: phosphatidylinositol 3-kinase-Akt; PKA: cAMP (cyclic adenosine monophosphate)-dependent protein kinase, or protein kinase A; PKC: protein kinase C; PKG: cGMP (cyclic guanosine monophosphate)-dependent protein kinase, or protein kinase G.

FoxO TFs

FoxO TFs belong to a family of transcriptional regulators characterized by a conserved DNA-binding domain termed the “forkhead box.”36 FoxO proteins mainly act as potent transcriptional activators by binding to the conserved consensus core recognition motif TTGTTTAC.37,38 FoxOs, when present in the nucleus and bound to promoters that contain the FoxO consensus motif, can act as transcriptional activators and repressors. In mammals, four FoxO isoforms have been identified: FoxO1, FoxO3, FoxO4, and FoxO6. Despite some redundant functions, genetic loss of individual FoxOs does result in specific phenotypes, suggesting divergent roles of FoxO isoforms.39 FoxOs control various cellular responses, including proliferation, apoptosis, DNA repair, and metabolism, and they have been implicated in vascular structural maintenance.40,41 FoxO activity is tightly controlled by posttranslational modifications (PTMs), such as phosphorylation, acetylation, methylation, ubiquitination, and binding of protein partners. These modifications alter transcriptional activity, DNA binding, subcellular localization, and protein stability. For example, phosphorylation of FoxO1 at Thr24 and Ser256 create binding motifs for the 14-3-3 scaffolding proteins, causing FoxO1 exclusion from the nucleus and promoting their cytoplasmic ubiquitination and degradation.37,42

At the 2015 Grover Conference, one of us (SSP) presented evidence that among the FoxO family, FoxO1 is centrally involved in the hyperproliferative and apoptosis-resistant phenotype of pulmonary arterial SMCs (PASMCs), which represents a hallmark of PH. We observed that, in pulmonary vessels and PASMCs of human and experimental PH lungs, FoxO1 expression is downregulated and FoxO1 is inactivated via phosphorylation and nuclear exclusion. Importantly, stimulation of human PASMCs by growth factors and inflammatory cytokines typically involved in the pathogenesis of PH, such as PDGF (platelet-derived growth factor), IGF-1 (insulin-like growth factor), TNF-α (tumor necrosis factor alpha), and interleukin 6, led to downregulation of FoxO1 phosphorylation at Ser256 and Thr24, nuclear exclusion and inactivation, suggesting that FoxO1 is one of the central downstream TFs regulated by several PH stimuli. This postulated central role of FoxO1 in the pathogenesis of pulmonary arterial hypertension (PAH; i.e., group 1 PH) is further supported by in vitro and in vivo loss-of-function studies.31 These data indicate that depletion of FoxO1 specifically in SMCs is sufficient to induce PH and also synergizes with the hypertensive effects of hypoxia, resulting in more severe PH. Reconstitution of PASMC FoxO1 activity by an adenovirus carrying a phosphorylation-deficient FoxO1 mutant reversed the features of PAH both in vitro and in vivo. Transfection of PASMCs with FoxO1 fully normalized growth factor–driven abnormalities in proliferation, migration, and apoptosis in vitro. In monocrotaline (MCT)-induced PAH, inhalation of the adenovirus (after full establishment of disease) markedly reduced RV systolic pressure, pulmonary vascular resistance, and RV hypertrophy—the central clinical features of this disease. In parallel, adenovirus-treated lungs showed reversal of vascular remodeling and normalization of vascular cell hyperproliferation and cell cycle abnormalities to favor the quiescent PASMC phenotype.31

SSP’s group also demonstrated that the anti-neoplastic drug paclitaxel has a similar ability to reverse the PAH PASMC phenotype in vitro and the clinical features of established experimental PAH in vivo. Both intravenous and inhaled paclitaxel markedly improved hemodynamics, RV hypertrophy, RV function, and vascular remodeling in various experimental PAH models. The finding that several of these pathways converge on FoxO1 as a central downstream effector molecule in hyperproliferative PASMCs offers new options for therapeutic intervention. The preclinical evidence for the efficacy of this approach will have to be assessed further in clinical trials.

Importantly, understanding of other PTMs on FoxOs, such as acetylation and ubiquitination, in the PH disease setting and the impact of FoxOs on other cellular processes driving PH, such as metabolism and inflammation, is essential.13 In addition, the effects of FoxOs and FoxO-activating therapies directly on RV hypertrophy and failure should be studied.13,43 Further characterizing the genes and gene networks regulated by FoxO1 in PAH-PASMCs, Savai et al.31 found that FoxO1-regulated genes are associated with cell cycle control (cyclin D1, p27), apoptosis (BCL6, GADD45), and bone morphogenetic protein (BMP) signaling (BMP receptor 2 [BMPR2], ID1, ID3). However, the microarray analysis also revealed changes in several genes that are not prototype target genes of FoxO TFs. An attractive possibility to explain these changes would be that FoxO has effects on chromatin properties more generally. Emerging evidence indicates that FoxO, similar to FoxA, has the capacity to bind to and remodel compacted chromatin.44 Thus, in addition to regulating the expression of specific genes, FoxO PTMs, similar to histone PTMs, may regulate overall chromatin structure and participate in the epigenetic regulation of gene expression, which require deeper investigations.

Hypoxic inducible factors (HIFs)

HIFs are TFs and key regulators of the molecular response to hypoxia. The targets of HIFs include genes controlling vascularization, cellular proliferation, migration, and metabolism.45,46 HIFs have therefore been strongly implicated in the pathogenesis of various forms of PH. Studies have shown that mice that are homozygous for either of the HIF isoforms (complete deletion of HIF1α or HIF2α is embryonic lethal) exhibit attenuated pulmonary pressures and vascular remodeling after experimental exposure to chronic hypoxia.16,47 Further, recent studies show that conditional deletion of HIF1α in SMCs ameliorates the degree of remodeling in chronically hypoxic mice.48,49 In addition, Kapitsinou et al.50 have recently shown that endothelial HIF2α is required for the development of increased PAP in chronically hypoxic mice. These studies strongly support the importance of better understanding the role of HIFs in the pathogenesis of PH, which includes how they interact with other transcriptional factors, transcriptional coactivators, and repressors.

HIFs are heterodimeric TFs composed of an α-subunit (HIF1α, HIF2α, or HIF3α) and a β-subunit (HIF1β, also called the arylhydrocarbon receptor nuclear translocator [ARNT]). HIF is absolutely required for HIF-target gene activation under hypoxia, because knockdown of HIF1α and/or ARNT significantly reduces or completely blocks hypoxic induction of HIF-target genes. However, recent reports demonstrate that other TFs are also required for HIF-target gene expression. These factors have been found to physically interact with HIF1α or HIF2α in transcriptional complexes on HIF-target gene promoters, demonstrating the possibility that some of these factors may be components of what have been termed HIF enhanceosomes.51 The finding that HIFs interact with a diverse variety of cotranscriptional activators suggests a mechanism by which hypoxia activates HIF, as well as a number of physiologically and pathologically relevant signaling pathways, to induce HIF-target gene expression.51-53 In addition, there are examples in which it has been shown that other TFs function to selectively enhance expression of HIF1α or HIF2α target genes, demonstrating that distinct HIF1α or HIF2α enhanceosome complexes exist and are composed of different sets of HIF-specific cotranscriptionally activating factors on the promoters of HIF-target genes.

Incorporation of multiple factors in enhanceosome complexes allows regulation of target gene transcription in a way that is highly sensitive to a variety of stimuli and can result in controlled levels of target gene expression under a variety of conditions. In addition, these complexes can allow for cooperation between factors acting within the same or overlapping signaling pathways and allow for cooperative or synergistic activation of target gene transcription in a context-specific manner (Fig. 1).51

It is noteworthy that HIF transcriptional effects are modulated by epigenetic factors described in more detail below. It is now clear that HIF activates its target gene expression by recruiting histone acetylases CBP and p300 via their C- and N-terminal activation domains.54 Of importance in the similarity between many of the identified HIF cotranscriptional activators is their ability to bind and recruit the general coactivator proteins CBP and p300. It has been suggested, in fact, that HIF-dependent CBP and p300 recruitment contributes less to total CBP/p300 bound to HIF-target gene promoters than other coactivators, such as STAT3. These findings raise important questions for the future as to the specific functions of HIFs versus those of coactivators in transcriptional activation of HIF-target genes in PH and a myriad of other diseases including cancer.51

Transcription corepressor RBPJ/CSL

The Notch signaling pathway is a versatile regulator of cell fate decisions and plays an essential role for vascular development. Notch signaling is activated upon cell-to-cell contact as a result of interactions between Notch receptors and their ligands (Delta or Jagged).55 Upon ligand binding, the Notch receptors undergo proteolytic processing, which involves ADAM metalloproteases and the γ-secretase complex, resulting in the intracellular domain of Notch (NICD) being released from the cell membrane and shuttling to the nucleus. This process activates the major downstream nuclear target for Notch, the CSL family of DNA-binding factors (CBF1/RBP-Jκ in mammals, Su(H) in Drosophila and Lag-1 in Caenorhabditis elegans). In the absence of active Notch, RBPJ is thought to be primarily a transcriptional repressor that exists in complexes with corepressors.56 When bound to the active NICD, RBPJ recruits a coactivator complex, including a mastermind homolog (MAML1–3 in mammals), activates transcription of genes containing RBPJ binding sites, and drives a complex transcriptional program with phenotypic effects.57,58

Notch signaling plays a critical role in controlling proliferation and differentiation of PASMCs. Upregulated Notch ligands and Notch3 receptors in PASMCs have been reported to promote the development of pulmonary vascular remodeling in patients with PAH and in animals with experimental PH.20 At the 2015 Grover Conference, one of us (JY) explained how hypoxia, both acute and chronic, regulates Notch signaling and how hypoxia-mediated Notch signaling regulates cytosolic free Ca2+ concentration ([Ca2+]cyt).59,60

Because Notch3 and Notch3 NICD expression is also increased in hypoxia-induced PH (HPH) in mice, JY’s group investigated the role of Notch signaling in the responses to acute and chronic hypoxia. Intriguingly, inhibition of Notch signaling with DAPT, a γ-secretase inhibitor (GSI), significantly attenuated the amplitude of acute alveolar hypoxia-induced increase in PAP in isolated perfused/ventilated mouse lungs. In addition, DAPT reversed the development of HPH in mice,20,60 suggesting that Notch signaling is involved in the responses of the pulmonary vasculature to acute and chronic hypoxia.

Elevated [Ca2+]cyt is implicated in stimulating vascular SMC proliferation and inducing vasomotor tone and hence vasoconstriction. In PASMCs, [Ca2+]cyt can be increased by Ca2+ release from the intracellular stores and Ca2+ influx through plasmalemma Ca2+ channels.61 In PASMCs, increased [Ca2+]cyt may occur by activation of various Ca2+-permeable channels, including (1) activation of receptor-operated (or ligand-gated) Ca2+ channels (ROCs) that are opened, independent of membrane potential, by agonist-mediated activation of membrane receptors; (2) activation of store-operated Ca2+ channels (SOCs) that are opened, independent of membrane potential, by depletion of Ca2+ from intracellular stores; (3) activation of voltage-dependent Ca2+ channels that are opened by membrane depolarization; (4) activation of inositol triphosphate (IP3) receptor–mediated Ca2+ release from the IP3-sensitive sarcoplasmic reticulum (SR); and (5) activation of ryanodine receptor–mediated Ca2+ release from ryanodine-sensitive SR. Previous work demonstrated that the resting [Ca2+]cyt is increased and that expression and activity of ROCs and SOCs are both upregulated in PASMCs isolated from patients with idiopathic PAH (IPAH).62-64 Furthermore, it was found that extracellular Ca2+ induces a large increase in [Ca2+]cyt in IPAH PASMCs and functionally upregulated extracellular Ca2+-sensing receptor (CaSR) is involved in the enhanced Ca2+ influx and proliferation in IPAH PASMCs.65

Along similar lines, in HPH, increased expression of SOCs and enhanced store-operated Ca2+ entry (SOCE) was shown to contribute to increased [Ca2+]cyt.64,66 SOCE is known to be important for cell proliferation and vascular remodeling in PH, and studies from JY’s lab have demonstrated increased expression of several proteins involved in SOCE, such as canonical transient receptor potential (TRPC) channels (TRPC3, TRPC6), stromal interaction molecule 2 (STIM2), and Orai2, in patients with PH.62,64,67,68 Understanding the molecular mechanisms that regulate [Ca2+]cyt and PASMC proliferation is critical in understanding the pathogenesis of HPH.

Hypoxic pulmonary vasoconstriction (HPV) is an important physiological response that optimizes the ventilation/perfusion ratio.69 In order to determine the involvement of Notch signaling in acute HPV, isolated perfused/ventilated mouse lungs and human PASMCs were treated with Notch ligand Jagged-1. Treatment with Jagged-1 resulted in a significant increase in PAP, demonstrating that Jagged-1 can mimic acute hypoxia to cause pulmonary vasoconstriction. In addition, interestingly, both short-term (15–30 min) and long-term (48 hours to days) activation of Notch by Jagged-1 enhances SOCE, which is closely associated with the level of NICD in PASMCs. Importantly, the short and rapid response to activation of Notch signaling in PASMCs can be explained by NICD functional interaction with TRPC6, leading to TRPC6 activation, enhanced SOCE, and an increase in [Ca2+]cyt in PASMCs. Moreover, employing several genetic and pharmacological approaches, Sylvester and colleagues69 elegantly demonstrated that TRPC6 is required for SOCE and acute HPV in mice and that inhibition of Notch signaling markedly attenuates hypoxia-enhanced SOCE and acute HPV. However, the marked decrease of acute HPV mediated by Notch inhibition is not observed in TRPC6-deficient (TRPC6−/−) mice, confirming the noncanonical form of Notch signaling, that is, NICD-TRPC6 interaction under acute hypoxic exposure.59,60

Chronic hypoxia causes vascular remodeling, which is central to the pathogenesis of HPH.70 As it was previously shown that Notch3 and SOCs (TRPC6, Orai1/2) are upregulated in PH induced by chronic hypoxia, it is interesting to decipher whether and how Notch signaling regulates SOCs or TRPC6 specifically in chronic hypoxic situations. Strikingly, JY’s group found that Notch signaling activation transcriptionally upregulates TRPC6, Orai1/2, and STIM2 via increased binding of RBP-Jκ with NICD, leading to enhanced SOCE under chronic hypoxia. Finally, pharmacological inhibition of Notch signaling attenuates TRPC6 expression, hypoxia-enhanced SOCE, and the development of HPH.59,60 These studies suggest a novel dual role for Notch signaling on TRPC6 via direct functional interaction with TRPC6 and transcriptional upregulation of TRPC6, thereby maintaining increased levels of [Ca2+]cyt, that plays an important role in both acute HPV and HPH. Furthermore, recent work from this group suggests that Notch signaling regulates not only [Ca2+]cyt but also Ca2+ influx via regulation of CaSR function. Activation of Notch signaling by Jagged-1 enhances CaSR function and thus regulates Ca2+ influx and [Ca2+]cyt. Together, these data demonstrate the essential role of Notch-RBPJ signaling in initiation and perpetuation of HPH.

Transcriptional coactivator PKM2

PKM1 and PKM2 are pyruvate kinase isoforms expressed in different types of cells and tissues. Pyruvate kinase regulates the final rate-limiting step of glycolysis by catalyzing the transfer of a phosphate group from phosphoenolpyruvate to adenosine diphosphate to produce pyruvate and adenosine triphosphate (ATP),71 which suggests their major functions in glycolysis. In addition to its well-known role in glycolysis, PKM2 has also been reported to be involved in other cellular functions. Importantly, PKM2 has recently been found to translocate into the nucleus upon mitogenic and oncogenic stimulation.72,73 In the nucleus, PKM2 functions as a transcriptional coactivator and a protein kinase that phosphorylates histones, highlighting the crucial role of PKM2 in the epigenetic regulation of gene transcription that is important for the G1-S phase transition and the “Warburg effect” (which states that most cancer cells produce energy by a high level of glycolysis, followed by lactic acid fermentation).74,75 PKM2 functions as a coactivator that stimulates HIF-1 transactivation of target genes encoding GLUT1, LDHA, and PDK1 in cancer cells.76 In addition to the crucial role of PKM2 in the G1-S phase, it phosphorylates important cell cycle regulators, such as the spindle checkpoint protein Bub3, to regulate chromatid segregation and the mitotic checkpoint during mitosis, and myosin light chain 2 (MLC2, encoded by MYL2) to initiate cytokinesis, leading to enhanced and governed tumor cell proliferation.77 In line with its functions, PKM2 expression has been found to be increased in all cancer types examined.78,79

At the 2015 Grover Conference, one of us (KS) briefly reviewed his group’s recent findings regarding the metabolic state of pulmonary arterial adventitial fibroblasts (PAAFs) isolated from hypertensive calves and humans with IPAH, the metabolic functions of PKM2, and the mechanisms that regulate PKM2 expression and its glycolytic enzymatic activity in these cells. Previous studies from the group suggested that some of the earliest pathophysiological changes in PH are observed in the pulmonary artery adventitia and include robust proliferation of fibroblasts and accumulation of macrophages.80-83 It is increasingly recognized that increased proliferation and inflammatory activation can occur in the context of changes in cellular metabolism toward aerobic glycolysis (the Warburg effect), and these metabolic changes have been reported in PH in pulmonary arterial endothelial cells (PAECs), PASMCs, and, most recently, PAAFs and have formed the basis for a “metabolic theory of PH.”84,85 However, the mechanisms that link metabolic reprogramming to inflammatory and proliferative pathways in PH remain largely unknown.

Elegant work employing RNA-Seq transcriptome profiling and nuclear magnetic resonance– and mass spectrometry–based metabolomics analysis showed significant metabolic reprogramming to aerobic glycolysis in PH-fibroblasts (PH-fibs). Glycolytic changes included sugar phosphates, 13C-glycolysis signature, 13C-lactate, d-glucose and glucose uptake, glyceraldehyde 3-phosphate, pyruvate, and lactate production. In addition, PH-fibs were characterized by alterations of redox homeostasis, as indicated by increased amounts of glutathione (GSH) and glutathione disulfide (GSSG), decreased GSH/GSSG ratios, evidence of utilization of the NADPH (nicotinamide adenine dinucleotide phosphate)-generating pentose phosphate pathway, and increased phosphogluconate and ribose phosphate (K. Stenmark, unpublished data). These data provide strong evidence that in vivo and in vitro aerobic glycolysis is a metabolic adaptation in PAAFs in the setting of chronic hypoxia as well as in IPAH. Metabolic data here suggest that, in a Warburg-like fashion, incomplete glucose oxidation affords PH-fibs building blocks to support the anaplerotic reactions (amino acid anabolism, fatty acid synthesis, urea cycle, gluconeogenesis) necessary to sustain proliferation, counteract oxidative stress, and promote resistance to apoptosis.

Importantly, these authors found an isoform switch from PKM1 to PKM2 in PH-fibs. Knockdown of PKM2 decreases PH-fib proliferation and lactate production, providing a link between transcriptional regulation and the metabolic status of the cell. The PKM1-to-PKM2 switch (i.e., PKM2 expression) is regulated at the level of transcribed PKM pre–messenger RNA (mRNA) by splicing factors. Polypyrimidine tract–binding protein 1 (PTBP1) is upregulated in PH-fibs by microRNA 124 (miR124) and subsequently binds to splicing signals that flank PKM exon 9, repressing the inclusion of exon 9 and thus promoting an enhanced expression of the PKM2 isoform. Knockdown of PTBP1, miR124 mimics, and histone deacetylase (HDAC) inhibitors reverses the PKM2-PKM1 switch in PH-fibs and reduces lactate production, suggesting that epigenetic regulation is an important mechanism in controlling PKM splicing in PH.

Transcriptional corepressor CtBP1

In the context of increased glycolysis, where increases in NADH/NAD (ratio of reduced to total nicotinamide adenine dinucleotide) are frequently observed, the functions of the CtBP family members, traditionally considered transcriptional corepressors, must also be considered. CtBPs comprise a dimeric family of transcriptional repressors encoded by two paralogous genes (CTBP1 and CTBP2) that have major roles in animal cell development and cancer and have been suggested to be “sensors” of the metabolic state of cells that can modulate many aspects of cell phenotype.86 Overexpression of CtBP is observed in a number of cancers, including prostate, ovarian, colon, and breast.74,87,88 Studies in cancer and the nervous system revealed that CtBPs promote cellular survival primarily through repression of Bcl-2 family members and other proapoptotic molecules (PERP, Bax, Bik, Puma, p21, and Noxa), as well as tumor suppressors (e.g., p16INK4a, p15INK4b).86,88,89 Importantly, there is emerging evidence that CtBPs may also function to modulate the inflammatory response.90 Further, a recent genome-wide analysis in breast cancer provided a more comprehensive description of CtBP repression targets.88,91 CtBP targets were categorized into three main categories: genes that regulate cell renewal and pluripotency, genes that control genome stability, and genes that regulate epithelial differentiation and prevent epithelial-to-mesenchymal transition, all processes of potential importance in PH pathogenesis.88,91 At the 2015 Grover Conference, one of us (KS) presented evidence for CtBP1 activation and control of PAAF proliferation, apoptosis resistance, and inflammatory gene expression in PAAFs derived from animals and humans with PH.

TF TWIST1

TWIST1 was recently demonstrated to play a crucial role in the pulmonary vascular remodeling responsible for human and experimental PAH through a process called endothelial-to-mesenchymal transition (EndoMT).34,92 Interestingly, BMPR2 expression negatively correlated with Twist1 expression. The Twist proteins (Twist1 and Twist2) are highly conserved developmental proteins with key roles for transcriptional regulation in mesenchymal cell lineages. They belong to the superfamily of basic helix-loop-helix (bHLH) proteins and exhibit bifunctional roles as both activators and repressors of gene transcription. The Twist proteins are expressed at low levels in adult tissues but may become abundantly reexpressed in cells undergoing malignant transformation. This observation prompted extensive research on the roles of Twist proteins in cancer progression and metastasis. Twist1 suppresses BMP-induced osteoblast differentiation, and downregulation of endogenous Twist1 enhances BMP signaling. Maximal inhibition of BMP signaling was observed when Twist1 was bound to E47, which markedly enhanced the stability of Twist1. Co-immunoprecipitation assays revealed that Twist1 formed a complex with Smad4 and HDAC.93

Very recent studies also indicate a novel role for Twist1 as a potential regulator of adipose tissue remodeling and inflammation. Several studies have suggested that Twist genes are important determinants of obesity, fat distribution, and remodeling capacity of different adipose depots. Moreover, Twist1 expression is strongly correlated with body mass index and insulin resistance in humans.94 Interestingly, mice overexpressing Twist1 mutants in cardiomyocytes developed pathological cardiac remodeling. Twist1 may play a growth-inhibitory role in the postnatal heart, and phosphorylation may release their inhibitory effects, leading to hypertrophy.95 Interestingly, the genes involved in oxidative phosphorylation and the Krebs (citrate) cycle were found with reduced levels in the hearts of mice overexpressing Twist1 mutants, suggesting that decreased metabolism may result in heart failure. Twist1 play also critical roles in diverse developmental systems, including myogenesis. Several experiments have demonstrated its role in inhibition of muscle cell differentiation. Overexpression of Twist1 can reverse muscle cell differentiation in the presence of growth factors.96 Hence, Twist1 is potentially involved in all pulmonary and systemic abnormalities found in PAH. Regarding EndoMT as responsible for neointima and plexiform lesion formation and distal neomuscularization, Hopper et al.35 also demonstrated that another TF might be involved. They showed in vitro that loss of BMPR2 in PAECs leads to heightened expression of the chromatin architectural factor high-mobility group AT-hook 1 (HMGA1), which increases the TF Slug. HMGA1 binds to DNA and may promote binding of additional pro-EndoMT TFs.35

Targeting TFs: promising new strategies for PH therapy?

These studies strongly support the idea that modulation of TF and TF coregulator activity affects several pathological processes underlying PH and hence offers the possibility of future therapeutic interventions. Hence, the design of therapeutic agents targeted at pro–pulmonary hypertensive TFs regulating the initial expression should be the ultimate goal. Another approach is promoting the activity of anti–pulmonary hypertensive TFs. This will provide a protective mechanism to the cells in order to prevent further hypertensive derangement of gene expression. However, TFs are often considered chemically intractable because they lack surface involutions and hydrophobic pockets for high-affinity binding of small molecules and nuclear localization. Nonetheless, compounds have been developed to target TFs at different levels of function, including DNA binding, protein-protein interactions, and epigenetics. Direct inhibition of TF expression (via RNA interference or microRNAs [miRs]) and DNA binding (via oligodeoxynucleotide or pyrrole imidazole polyamides) are progressing into clinical trials with safer and more effective delivery systems. As mentioned above, miR124 mimics are useful tools to regulate PKM activity and thereby PH phenotype (K. Stenmark, unpublished data). For these approaches to enter the clinic arena, novel delivery systems must be developed.

Novel approaches in drug design that will selectively affect particular TFs and suppress their activity can be of great therapeutic interest. TFs have ligand-binding, dimerization, transeffector, DNA-binding, nuclear-localization, and regulatory domains, which may be targeted directly by small-molecule drugs. Other promising alternatives include artificial TF mimics, which consist of normal TF domains conjugated with synthetic compounds targeting DNA-binding domains or regulatory domains.

In addition, we need to carefully assess the effects of TFs-therapeutic targeting on other tissues. For example, modulating Notch signaling with GSIs in the early GSI trials for Alzheimer’s disease, which led to widespread gastrointestinal toxicity,97,98 underscores that targeting of TFs has to be approached with a great deal of precision. The potential crosstalk of Notch signaling with several other signaling mechanisms, such as TRPC6, opens up interesting possibilities for combining Notch modulation with other therapeutic strategies. Indeed, the combined inhibition of Notch and TRPC6 signaling could be a novel and safe therapeutic approach.

Epigenetics: definition and mechanisms

Epigenetics is generally defined as heritable changes in gene activity and expression that occur without alteration in DNA sequence.99 Epigenetic regulation of chromatin structure is fundamental to establish and maintain cell type–specific gene expression during development and disease states.

Epigenetic mechanisms tightly regulate chromatin accessibility. Double-stranded DNA does not exist naked in the nucleus of the eukaryotic cell but is packaged into a highly organized nucleoprotein complex known as chromatin. The basic unit of chromatin (a protein-DNA complex) is the nucleosome, which comprises 147 base pairs of DNA wrapped around an octameric histone core containing two copies each of the histones H2A, H2B, H3, and H4.100 Overall, the higher-order chromatin structure facilitates the packaging, organization, and distribution of DNA but negatively affects several fundamental biological processes, such as transcription, replication, and DNA repair, by restricting the accessibility for multiprotein complexes.100 Chromatin adopts different structural conformations, depending on the epigenetic modifications that occur in the DNA and histones.100 Changes in the chromatin status of specific genes can lead to their repression or activation. Regulation of chromatin structure involves several interconnected mechanisms, such as (1) DNA methylation, (2) ATP-dependent chromatin remodeling, (3) histone PTMs, (4) ncRNAs, (5) replacement of canonical histones with histone variants, and (6) organization within the three-dimensional nuclear architecture (Fig. 2).101

Figure 2.

Figure 2

Epigenetic mechanisms in gene regulation. (1) DNA methyltransferases (DNMT) and the ten-eleven translocation (TET) methyl cytosine dioxygenase family modulate DNA methylation levels. (2) Chromatin remodeling complexes move, eject, or restructure nucleosomes. (3) Histone modifications are regulated by the counteracting activity of histone acetyltransferases (HATs) and histone deacetylases (HDACs) or lysine methyltransferases (KMTs) and lysine demethylases (KDMs). Acetylation is recognized by bromodomain-containing proteins and methylation by PHD, chromo, WD40, Tudor, double/tandem Tudor, MBT, ankyrin repeats, and PWWP domains. (4) Noncoding RNAs (long noncoding RNAs and microRNAs) affect gene expression by gene silencing or inhibition of protein translation. (5) Histone variants; these mechanisms or processes are the key regulators of genome-environment interactions. K-Ac: lysine acetylation; K-Me: lysine methylation; MBD: methyl CpG (cytosine-phosphate-guanine)-binding domain.

Why epigenetic alterations might be involved in PH

Despite our progressive understanding of the pathogenesis of PH and recent therapeutic advances, PH remains a fatal disease. PH is a complex disease with multifactorial etiology mediated by the interplay of genetic background, epigenetic changes, and exposure to injurious events such as viruses, drugs, toxins, hypoxia, and inflammation, which may explain the great variability in disease susceptibility. In PAH, the key genetic determinants, such as low-penetrance dominant BMPR2 mutations, account for ∼70% in familial and only ∼15% in idiopathic PAH cases.102 In a suitable genetic background, the interplay of epigenetics and injurious events may augment the severity of the disease, aggravate vascular remodeling and worsen clinical outcome.103

We presume that persistence of hypertensive stimuli might disrupt cellular homeostasis and alter the existing epigenetic state (quiescent) of the pulmonary vascular cells and reestablish a new epigenetic signature that favors a disease phenotype exhibiting uncontrolled cell proliferation (Fig. 3). After acquisition of the disease phenotype, the pulmonary vascular cells contributing to the vascular remodeling process exhibit stable proproliferative, promigratory, antiapoptotic, proinflammatory, and profibrotic vascular cell phenotypes (Fig. 3).104 Notably, we and others have observed that vascular cells (PAECs, PASMCs, PAAFs) isolated from hypertensive lung vessels and cultured ex vivo outside their vascular microenvironment maintain a hyperproliferative, apoptosis-resistant phenotype for a longer time than control cells19 and also maintain higher mesenchymal transitional potential,34,35,105 demonstrating the presence of heritable and sustained phenotype.

Figure 3.

Figure 3

Phenotype alterations in pulmonary arterial hypertension (PAH). Pathological stimuli-induced recruitment of transcription factors and alterations in epigenetic mechanisms involves DNA methylation, histone modifications, and microRNAs. These epigenetic alterations in the gene expression programs facilitate the normal vascular cells to exhibit increased proliferation, migration, apoptosis, inflammatory gene expression, and increased mesenchymal transition and collagen extracellular matrix remodeling, as commonly observed in PAH pulmonary arteries. The persistence of pathological stimuli further establishes a phenotypic memory in PAH vascular cells that largely contributes to the development of complex vascular lesions during progressive vascular remodeling process in PAH.

Summary of epigenetics-related findings in PH/PAH

Emerging evidence suggests that the pathogenesis of PH is influenced by abnormalities in epigenetic mechanisms such as DNA methylation, HDAC activity, and miRs.103

MicroRNA dysregulation in the pathogenesis of PH

MicroRNAs are small ncRNAs that negatively regulate gene expression by binding to the 3′ untranslated region of target mRNAs, thereby promoting mRNA degradation or inhibiting translation. Profiling of the miR expression in rodent models of PH and in human pulmonary arteries from PAH patients indicated dysregulation of several miRs, including miR204, the mir17/92 cluster, the miR143/145 cluster, and others.103 To fine-balance these dysregulated miRs, different strategies, such as miR mimics or antagomirs, were employed and demonstrated to reverse the vascular remodeling in PH experimental models. Because of space limitations, we do not provide an extended analysis here, as the topic is covered in greater depth elsewhere.103,106

Altered DNA methylation patterns in PH

DNA methylation is one of the best-studied epigenetic modifications occurring in mammals, almost exclusively on the 5′ position of the pyrimidine ring of cytosines in the context of CpG (5′-cytosine-phosphate-guanine-3′) dinucleotides. This DNA modification is mediated by DNA methyltransferases and generally results in tight packing of DNA and histones (heterochromatin) and in the long-term silencing of transcription. DNA methylation may result in gene silencing that can be propagated to daughter cells.

The influence of nongenetic factors in autoimmune and inflammatory diseases has been demonstrated in studies of genetically identical (monozygotic [MZ]) twins who show a variable degree of discordance with respect to different phenotypic traits, including susceptibility to these diseases. MZ twins show a concordance rate for different autoimmune diseases ranging from 12% for rheumatoid arthritis up to 70% for psoriasis. There is substantial epigenetic variation between MZ twin pairs. This variation increases with age and if twins live in different environments. These environment-host interactions might result in such severe epigenetic changes that the epigenetic homeostasis can no longer be maintained, leading to aberrant gene expression. Aberrant gene expression in specific cells of the immune system most likely contributes to the loss of immune tolerance, inflammation, and autoimmunity, which is characterized by the failure of an organism to recognize self-antigens. While a large number of candidate gene and genome-wide studies analyzing the DNA methylation profiles have been published for inflammatory and autoimmune diseases such as type 1 diabetes, multiple sclerosis, and systemic lupus erythematosus,107 evidence for the implication of DNA methylation modifications in PAH is currently scarce.

Archer´s group108 has identified a methylation-induced attenuation of Sod2 (mitochondrial superoxide dismutase 2) expression as an epigenetic mechanism in an animal model of spontaneous and heritable PAH (fawn-hooded rats). This may be relevant for clinical PAH because SOD2 is also downregulated in human PAH. The epigenetic downregulation of SOD2 impairs H2O2-mediated redox signaling, activates HIF-1, and creates a proliferative, apoptosis-resistant state. Both the mechanism of SOD2 downregulation and its consequences parallel those discovered in human cancer.109

Moreover, DNA methylation for the measurement of a PAH marker has a number of attractive features. First, DNA molecules are very stable and, in contrast to mRNA and many proteins, can survive harsh conditions for long periods of time. Second, unlike proteins, nucleic acid can be amplified by polymerase chain reaction and related techniques, thus allowing measurements on small amounts of test sample.110 Moreover, blood levels of most currently used protein biomarkers are rarely increased in the early stage of the disease. Consequently, most existing blood protein biomarkers are of little value in either screening for or aiding the early diagnosis of PAH. On the other hand, aberrant methylation of the promoter regions of multiple genes may exist in both early and advanced PAH. As proof of principle, FP’s group111,112 published an epigenetic study showing that DNA methylation alterations in the granulysin (GNLY) gene in explanted lungs and in peripheral blood mononuclear cells allow discrimination between pulmonary veno-occlusive disease (PVOD) patients (PVOD is a subgroup of PH with worse prognosis and risk of developing severe pulmonary edema with specific PAH therapy) and PAH patients.

At the 2015 Grover Conference, FP presented his ongoing work about the identification of a DNA methylation-based PAH signature. His group analyzed the promoters of protein-coding sequences, miRs, and long noncoding RNA in the genomic DNA from cultured PAECs isolated from IPAH and heritable PAH (BMPR2 mutation carriers) patients and controls. Enrichment analyses demonstrated that the differentially methylated promoters clustering in controls and PAH groups were enriched in pathways, networks, and functions related to metabolism, cardiovascular system development and function, cellular development, skeletal and muscular system development and function, cell morphology, cellular function and maintenance, cell death and survival, cell-mediated immune response, cellular development, cancer, and organismal injury and abnormalities (F. Perros, unpublished data). Further analysis of these data will enhance the discovery of novel regulatory pathways in PAH that require innovative therapy.

Alterations in histone levels in PAH

Linker histones (H1) have been found to play an important role in transcription, gene regulation, protein-protein interactions, and innate immunity and are associated with complexes to downregulate proinflammatory genes. Talati et al.113 employed histology-based matrix-assisted laser desorption ionization–mass spectrometry (MALDI-MS) and identified significant increases, compared to controls, in two fragments of histone H1 in the pulmonary arteries from the explanted lungs of the IPAH patients. However, a reduction in the nuclear histone H1 and an increase in the cytoplasmic H1 were observed in IPAH cells. The decreased nuclear H1 contributes to the less compact nucleosomal pattern in IPAH, and this, in turn, contributes to the increase in nucleosomal repeat length and ultimately to changes in transcription.113

Role of HDACs in PAH

Two different families of enzymes mediate the balance between the acetylated and deacetylated states of histone or nonhistone proteins: histone acetyltransferases and HDACs. The role of aberrant HDAC activity and histone acetylation has been strongly implicated in PAH pathogenesis on the basis of the promising therapeutic benefits observed upon the application of small-molecule HDAC inhibitors in different animal models of PH. Treatment with pan-HDAC inhibitors (suberoylanilide hydroxamic acid [vorinostat]) and class I selective inhibitors (MGCD0103, valproic acid) both attenuated and reversed the development of hypoxia- and MCT-induced PH in a rodent model.114,115 Table 2 summarizes all research findings that reported the aberrant expression and activity of HDACs and the therapeutic effects of pan-HDAC inhibition. Moreover, Li et al.83 demonstrated that class I HDAC inhibition reverses stable proinflammatory phenotype of PH. This study suggests that the persistent proinflammatory phenotype exhibited by activated PAAFs was associated with abnormal activity of HDACs, specifically, class I HDACs.83 Importantly, catalytic inhibition of class I HDACs using the selective class I HDAC inhibitor apicidin significantly suppressed production of proinflammatory mediators by PH-fibs and caused a marked reduction in the ability of PH-fibs to induce monocyte migration and activation.83 Using Bmpr2+/− mice, Soon and coworkers122 recently demonstrated that BMPR2 deficiency promotes an exaggerated inflammatory response. In their study, BMPR2 deficiency in mouse and human PASMCs was associated with increased phospho-STAT3 and loss of extracellular superoxide dismutase (SOD3). Interestingly, the proinflammatory effect of constitutive BMPR2 haploinsufficiency could not be mimicked by short-term BMPR2 deficiency. The authors suggested that a potential mechanism is epigenetic modification of the promoter regions of genes as a consequence of chronic loss of BMPR2. Accordingly, they demonstrated that the loss of SOD3 expression in Bmpr2+/− cells was reversed by incubation with trichostatin A.122,123

Table 2.

Histone deacetylases in PH: published evidence

Species Model/tissues/cells Summary Reference
Rat PAB; RV Suppression of HDACs (TSA) worsens RV dysfunction after PAB Bogaard et al.116
Bovine Hypoxia; adventitial fibroblasts Alterations in class (I) HDAC1/2/3 expression and activity contribute to a proinflammatory phenotype of PH-fibroblasts and was attenuated by application of HDAC inhibitors (SAHA, apicidin) Li et al.83
Human/bovine/rat Increased expression of HDAC1/4/5 in IPAH lung homogenates, HI PH lungs and RV; HDAC inhibition (SAHA, VPA) attenuated the development of HI PH and hyperproliferative phenotype of PH-fibroblasts and R-cells from hypertensive bovine pulmonary arteries Zhao et al.115
 Human IPAH; lungs
 Bovine Adventitial fibroblasts, R-cells
 Rat Hypoxia; lungs, RV
Rat Hypoxia; RV, PASMCs Selective class I HDAC inhibitors (MGCD0103 and MS-275) suppresses HI PH and RVH in a preclinical model of hypoxia-mediated PH Cavasin et al.114
Ovine (sheep) Serum; PASMCs Inhibition of class I and II HDACs by apicidin and HDACi VIII suppressed serum-induced proliferation and migration of newborn PASMCs Yang et al.117
Rat PDGF; PASMCs Sodium butyrate inhibits PDGF-induced proliferation and migration in PASMCs through Akt inhibition Cantoni et al.118
Rat PDGF; PASMCs Class I HDAC inhibitor (MC1855), but not class II HDAC inhibitor (MC1575), counteracts PDGF-induced proliferation and migration of PAH PASMCs Galletti et al.119
Bovine Hypoxia; adventitial fibroblasts Pan-HDAC inhibitors (SAHA, apicidin) rescued the HI downregulation of antiproliferative miR-124 expression in hypoxic adventitial fibroblasts Wang et al.120
Rat MCT, SuHx-PH, PAB Pan-HDAC inhibitor TSA has no therapeutic or beneficial effects in SuHx-PH De Raaf et al.121
Human IPAH PAECs, MCT, SuHx-PH Increased nuclear accumulation of class IIa HDACs HDAC4 and HDAC5 impaired MEF2 activity in PAH PAECs; pharmacological inhibition of class IIa HDACs using MC1568 rescued the impaired MEF2 transcriptional activity in PAH PAECs and also reduced PAH PAEC proliferation and migration; no evidence of myocardial fibrosis in MCT and SuHx groups upon HDAC inhibition Kim et al.33
Note

HDAC: histone deacetylase; HI: hypoxia-induced; IPAH: idiopathic PAH; MCT: monocrotaline; MEF2: myocyte enhancer factor; PAB: pulmonary artery banding; PAECs: pulmonary artery endothelial cells; PAH: pulmonary arterial hypertension; PASMCs: pulmonary artery smooth muscle cells; PDGF: platelet-derived growth factor; RV: right ventricle/ventricular; RVH: RV hypertrophy; SAHA: suberoylanilide hydroxamic acid; SuHX-PH: hypoxia+SU5416–induced PAH (SU5416 is a vascular endothelial growth factor receptor [VEGFR-2] inhibitor); TSA: trichostatin A; VPA: valproic acid.

Collectively, these findings also highlight the need for identification of deregulated isoforms in the PAH setting and to dissect the “histone code” that can mediate a unique cellular response, such as the proinflammatory/proproliferative PAAF phenotype, the hyperproliferative PASMC phenotype, and the angioproliferative PAEC phenotype. In order to elucidate the histone acetylation code underlying the PAH phenotype in human setting, SSP’s group employed three approaches: (1) investigate the expression, regulation, and functional role of histone acetylation modifiers, (2) explore the therapeutic potential of isoform-selective modulation of deregulated HDAC isoforms in PAH disease phenotype reversal ex vivo and in vivo, and (3) employ an unbiased next-generation sequencing–based approach to identify the disease-specific epigenetic signatures (histone modification patterns and transcriptome profiles) and the regulatory network that facilitates the vascular cells to acquire and sustain the disease phenotype. Interestingly, aberrant gene and protein expression profiles of class I HDAC isoforms across a variety of cardiopulmonary samples from human PAH was documented. The screening also revealed an organ- and cell-specific expression pattern of these isoforms between IPAH patients and donors. The stable disease phenotype exhibited by ex vivo isolated IPAH PAAFs was associated with aberrant expression and activity of class I HDACs isoforms. Importantly, integrative analysis of vascular cell–specific epigenomics data provides insights into the mechanistic link between recruitment of histone modifiers and the regulatory network of histone modifications that cooperatively regulates aberrant transcriptional responses in PAH (S. S. Pullamsetti and P. Chelladurai, unpublished data).

Epigenetic modulation: promising new strategies for PH therapy?

As miR inhibitors are currently tested for different indications in clinical trials, their clinical development as a new therapy for PH might therefore be imminent. It will be important to define the most suitable miR(s) to target and the optimum delivery strategy to address the pulmonary vascular compartment. A systematic evaluation of compartment- and cell-specific HDACs in experimental and human PAH and of the isoform-selective functions/role of HDACs in pulmonary vascular and RV remodeling is missing. In addition, detailed study of HDAC-mediated versus non-HDAC-mediated effects of pan-HDAC inhibitors is needed. Isoform-selective HDAC inhibitors should be evaluated for preclinical and clinical efficacy. In addition, we have to understand how the combination of different histone modifications (acetylation, methylation) appearing on different histones at different times (the “histone code”) can mediate a unique cellular response, such as the hyperproliferative PASMC phenotype.

For epigenetic modulation therapies, the lack of specificity of any interference is an important issue. For example, manipulation of global DNA demethylation via the use of nonselective agents can contribute to increased chromosomal instability and may even be transmitted between generations.124 Therefore, interference has to be fine-tuned to focus on specific epigenetic regulators/modifiers linked with the genes most relevant for PH development.

Source of Support: Nil.

Conflict of Interest: None declared.

References

  • 1.Lelli KM, Slattery M, Mann RS. Disentangling the many layers of eukaryotic transcriptional regulation. Annu Rev Genet 2012;46:43–68. doi:10.1146/annurev-genet-110711-155437. [DOI] [PMC free article] [PubMed]
  • 2.Ong CT, Corces VG. Enhancers: emerging roles in cell fate specification. EMBO Rep 2012;13(5):423–430. doi:10.1038/embor.2012.52. [DOI] [PMC free article] [PubMed]
  • 3.Spitz F, Furlong EE. Transcription factors: from enhancer binding to developmental control. Nat Rev Genet 2012;13(9):613–626. doi:10.1038/nrg3207. [DOI] [PubMed]
  • 4.Goodrich JA, Tjian R. Unexpected roles for core promoter recognition factors in cell-type-specific transcription and gene regulation. Nat Rev Genet 2010;11(8):549–558. doi:10.1038/nrg2847. [DOI] [PMC free article] [PubMed]
  • 5.Sadhale P, Verma J, Naorem A. Basal transcription machinery: role in regulation of stress response in eukaryotes. J Biosci 2007;32(3):569–578. [DOI] [PubMed]
  • 6.Lee TI, Young RA. Transcriptional regulation and its misregulation in disease. Cell 2013;152(6):1237–1251. doi:10.1016/j.cell.2013.02.014. [DOI] [PMC free article] [PubMed]
  • 7.Yu S, Reddy JK. Transcription coactivators for peroxisome proliferator-activated receptors. Biochim Biophys Acta 2007;1771(8):936–951. doi:10.1016/j.bbalip.2007.01.008. [DOI] [PubMed]
  • 8.Kim J, Choi M, Kim JR, Jin H, Kim VN, Cho KH. The co-regulation mechanism of transcription factors in the human gene regulatory network. Nucleic Acids Res 2012;40(18):8849–8861. doi:10.1093/nar/gks664. [DOI] [PMC free article] [PubMed]
  • 9.Rosenfeld MG, Lunyak VV, Glass CK. Sensors and signals: a coactivator/corepressor/epigenetic code for integrating signal-dependent programs of transcriptional response. Genes Dev 2006;20(11):1405–1428. doi:10.1101/gad.1424806. [DOI] [PubMed]
  • 10.Hoeper MM, Bogaard HJ, Condliffe R, Frantz R, Khanna D, Kurzyna M, Langleben D, et al. Definitions and diagnosis of pulmonary hypertension. J Am Coll Cardiol 2013;62(25 suppl):D42–D50. doi:10.1016/j.jacc.2013.10.032. [DOI] [PubMed]
  • 11.Pullamsetti SS, Schermuly R, Ghofrani A, Weissmann N, Grimminger F, Seeger W. Novel and emerging therapies for pulmonary hypertension. Am J Respir Crit Care Med 2014;189(4):394–400. doi:10.1164/rccm.201308-1543PP. [DOI] [PubMed]
  • 12.Tuder RM, Archer SL, Dorfmüller 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(25 suppl):D4–D12. doi:10.1016/j.jacc.2013.10.025. [DOI] [PMC free article] [PubMed]
  • 13.Paulin R, Michelakis ED. Addressing complexity in pulmonary hypertension: the FoxO1 case. Circ Res 2015;116(11):1732–1735. doi:10.1161/CIRCRESAHA.115.305773. [DOI] [PubMed]
  • 14.Ameshima S, Golpon H, Cool CD, Chan D, Vandivier RW, Gardai SJ, Wick M, Nemenoff RA, Geraci MW, Voelkel NF. Peroxisome proliferator-activated receptor gamma (PPARγ) expression is decreased in pulmonary hypertension and affects endothelial cell growth. Circ Res 2003;92(10):1162–1169. [DOI] [PubMed]
  • 15.Hansmann G, de Jesus Perez VA, Alastalo TP, Alvira CM, Guignabert C, Bekker JM, Schellong S, et al. An antiproliferative BMP-2/PPARγ/apoE axis in human and murine SMCs and its role in pulmonary hypertension. J Clin Investig 2008;118(5):1846–1857. [DOI] [PMC free article] [PubMed]
  • 16.Brusselmans K, Compernolle V, Tjwa M, Wiesener MS, Maxwell PH, Collen D, Carmeliet P. Heterozygous deficiency of hypoxia-inducible factor-2α protects mice against pulmonary hypertension and right ventricular dysfunction during prolonged hypoxia. J Clin Investig 2003;111(10):1519–1527. doi:10.1172/JCI15496. [DOI] [PMC free article] [PubMed]
  • 17.Bonnet S, Michelakis ED, Porter CJ, Andrade-Navarro MA, Thébaud B, Bonnet S, Haromy A, et al. An abnormal mitochondrial–hypoxia inducible factor-1α–Kv channel pathway disrupts oxygen sensing and triggers pulmonary arterial hypertension in fawn hooded rats: similarities to human pulmonary arterial hypertension. Circulation 2006;113(22):2630–2641. [DOI] [PubMed]
  • 18.Bonnet S, Rochefort G, Sutendra G, Archer SL, Haromy A, Webster L, Hashimoto K, Bonnet SN, Michelakis ED. The nuclear factor of activated T cells in pulmonary arterial hypertension can be therapeutically targeted. Proc Natl Acad Sci USA 2007;104(27):11418–11423. [DOI] [PMC free article] [PubMed]
  • 19.Masri FA, Xu W, Comhair SA, Asosingh K, Koo M, Vasanji A, Drazba J, Anand-Apte B, Erzurum SC. Hyperproliferative apoptosis-resistant endothelial cells in idiopathic pulmonary arterial hypertension. Am J Physiol Lung Cell Mol Physiol 2007;293(3):L548–L554. doi:10.1152/ajplung.00428.2006. [DOI] [PubMed]
  • 20.Li X, Zhang X, Leathers R, Makino A, Huang C, Parsa P, Macias J, Yuan JX, Jamieson SW, Thistlethwaite PA. Notch3 signaling promotes the development of pulmonary arterial hypertension. Nat Med 2009;15(11):1289–1297. doi:10.1038/nm.2021. [DOI] [PMC free article] [PubMed]
  • 21.Huang Z, Wang D, Ihida-Stansbury K, Jones PL, Martin JF. Defective pulmonary vascular remodeling in Smad8 mutant mice. Hum Mol Genet 2009;18(15):2791–2801. [DOI] [PMC free article] [PubMed]
  • 22.Firth AL, Yao W, Remillard CV, Ogawa A, Yuan JX. Upregulation of Oct-4 isoforms in pulmonary artery smooth muscle cells from patients with pulmonary arterial hypertension. Am J Physiol Lung Cell Mol Physiol 2010;298(4):L548–L557. [DOI] [PMC free article] [PubMed]
  • 23.Bierer R, Nitta CH, Friedman J, Codianni S, de Frutos S, Dominguez-Bautista JA, Howard TA, Resta TC, Bosc LV. NFATc3 is required for chronic hypoxia-induced pulmonary hypertension in adult and neonatal mice. Am J Physiol Lung Cell Mol Physiol 2011;301(6):L872–L880. [DOI] [PMC free article] [PubMed]
  • 24.Courboulin A, Tremblay VL, Barrier M, Meloche J, Jacob MH, Chapolard M, Bisserier M, et al. Krüppel-like factor 5 contributes to pulmonary artery smooth muscle proliferation and resistance to apoptosis in human pulmonary arterial hypertension. Respir Res 2011;12:128. doi:10.1186/1465-9921-12-128. [DOI] [PMC free article] [PubMed]
  • 25.Mizuno S, Bogaard HJ, Kraskauskas D, Alhussaini A, Gomez-Arroyo J, Voelkel NF, Ishizaki T. p53 gene deficiency promotes hypoxia-induced pulmonary hypertension and vascular remodeling in mice. Am J Physiol Lung Cell Mol Physiol 2011;300(5):L753–L761. [DOI] [PubMed]
  • 26.Xu YP, Zhu JJ, Cheng F, Jiang KW, Gu WZ, Shen Z, Wu YD, Liang L, Du LZ. Ghrelin ameliorates hypoxia-induced pulmonary hypertension via phospho-GSK3 β/β-catenin signaling in neonatal rats. J Mol Endocrinol 2011;47(1):33–43. [DOI] [PubMed]
  • 27.Mata M, Sarrion I, Milian L, Juan G, Ramon M, Naufal D, Gil J, Ridocci F, Fabregat-Andres O, Cortijo J. PGC-1α induction in pulmonary arterial hypertension. Oxid Med Cell Longev 2012;2012:236572. doi:10.1155/2012/236572. [DOI] [PMC free article] [PubMed]
  • 28.Price LC, Caramori G, Perros F, Meng C, Gambaryan N, Dorfmüller P, Montani D, et al. Nuclear factor κ-B is activated in the pulmonary vessels of patients with end-stage idiopathic pulmonary arterial hypertension. PLoS ONE 2013;8(10):e75415. doi:10.1371/journal.pone.0075415. [DOI] [PMC free article] [PubMed]
  • 29.Ghatnekar A, Chrobak I, Reese C, Stawski L, Seta F, Wirrig E, Paez-Cortez J, et al. Endothelial GATA-6 deficiency promotes pulmonary arterial hypertension. Am J Pathol 2013;182(6):2391–2406. [DOI] [PMC free article] [PubMed]
  • 30.Biasin V, Chwalek K, Wilhelm J, Best J, Marsh LM, Ghanim B, Klepetko W, et al. Endothelin-1 driven proliferation of pulmonary arterial smooth muscle cells is c-fos dependent. Int J Biochem Cell Biol 2014;54:137–148. [DOI] [PubMed]
  • 31.Savai R, Al-Tamari HM, Sedding D, Kojonazarov B, Muecke C, Teske R, Capecchi MR, et al. Pro-proliferative and inflammatory signaling converge on FoxO1 transcription factor in pulmonary hypertension. Nat Med 2014;20(11):1289–1300. doi:10.1038/nm.3695. [DOI] [PubMed]
  • 32.Shatat MA, Tian H, Zhang R, Tandon G, Hale A, Fritz JS, Zhou G, et al. Endothelial Krüppel-like factor 4 modulates pulmonary arterial hypertension. Am J Respir Cell Mol Biol 2014;50(3):647–653. [DOI] [PMC free article] [PubMed]
  • 33.Kim J, Hwangbo C, Hu X, Kang Y, Papangeli I, Mehrotra D, Park H, et al. Restoration of impaired endothelial myocyte enhancer factor 2 function rescues pulmonary arterial hypertension. Circulation 2015;131(2):190–199. [DOI] [PMC free article] [PubMed]
  • 34.Ranchoux B, Antigny F, Rucker-Martin C, Hautefort A, Péchoux C, Bogaard HJ, Dorfmüller P, et al. Endothelial-to-mesenchymal transition in pulmonary hypertension. Circulation 2015;131(11):1006–1018. doi:10.1161/CIRCULATIONAHA.114.008750. [DOI] [PubMed]
  • 35.Hopper RK, Moonen JA, 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(18):1783–1794. doi:10.1161/CIRCULATIONAHA.115.020617. [DOI] [PMC free article] [PubMed]
  • 36.Kaestner KH, Knöchel W, Martínez DE. Unified nomenclature for the winged helix/forkhead transcription factors. Genes Dev 2000;14(2):142–146. [PubMed]
  • 37.Calnan DR, Brunet A. The FoxO code. Oncogene 2008;27(16):2276–2288. doi:10.1038/onc.2008.21. [DOI] [PubMed]
  • 38.Furuyama T, Nakazawa T, Nakano I, Mori N. Identification of the differential distribution patterns of mRNAs and consensus binding sequences for mouse DAF-16 homologues. Biochem J 2000;349(2):629–634. [DOI] [PMC free article] [PubMed]
  • 39.Hosaka T, Biggs WH III, Tieu D, Boyer AD, Varki NM, Cavenee WK, Arden KC. Disruption of forkhead transcription factor (FOXO) family members in mice reveals their functional diversification. Proc Natl Acad Sci USA 2004;101(9):2975–2980. doi:10.1073/pnas.0400093101. [DOI] [PMC free article] [PubMed]
  • 40.Paik JH. FOXOs in the maintenance of vascular homoeostasis. Biochem Soc Trans 2006;34(5):731–734. doi:10.1042/BST0340731. [DOI] [PubMed]
  • 41.Wilhelm K, Happel K, Eelen G, Schoors S, Oellerich MF, Lim R, Zimmermann B, et al. FOXO1 couples metabolic activity and growth state in the vascular endothelium. Nature 2016;529(7585):216–220. doi:10.1038/nature16498. [DOI] [PMC free article] [PubMed]
  • 42.Zhao Y, Wang Y, Zhu WG. Applications of post-translational modifications of FoxO family proteins in biological functions. J Mol Cell Biol 2011;3(5):276–282. doi:10.1093/jmcb/mjr013. [DOI] [PubMed]
  • 43.Piao L, Sidhu VK, Fang YH, Ryan JJ, Parikh KS, Hong Z, Toth PT, et al. FOXO1-mediated upregulation of pyruvate dehydrogenase kinase-4 (PDK4) decreases glucose oxidation and impairs right ventricular function in pulmonary hypertension: therapeutic benefits of dichloroacetate. J Mol Med (Berl) 2013;91(3):333–346. doi:10.1007/s00109-012-0982-0. [DOI] [PMC free article] [PubMed]
  • 44.Hatta M, Cirillo LA. Chromatin opening and stable perturbation of core histone:DNA contacts by FoxO1. J Biol Chem 2007;282(49):35583–35593. doi:10.1074/jbc.M704735200. [DOI] [PubMed]
  • 45.Semenza GL. Regulation of vascularization by hypoxia-inducible factor 1. Ann NY Acad Sci 2009;1177:2–8. doi:10.1111/j.1749-6632.2009.05032.x. [DOI] [PubMed]
  • 46.Hubbi ME, Gilkes DM, Hu H, Kshitiz, Ahmed I, Semenza GL. Cyclin-dependent kinases regulate lysosomal degradation of hypoxia-inducible factor 1α to promote cell-cycle progression. Proc Natl Acad Sci USA 2014;111(32):E3325–E3334. doi:10.1073/pnas.1412840111. [DOI] [PMC free article] [PubMed]
  • 47.Shimoda LA, Manalo DJ, Sham JS, Semenza GL, Sylvester JT. Partial HIF-1α deficiency impairs pulmonary arterial myocyte electrophysiological responses to hypoxia. Am J Physiol Lung Cell Mol Physiol 2001;281(1):L202–L208. [DOI] [PubMed]
  • 48.Ball MK, Waypa GB, Mungai PT, Nielsen JM, Czech L, Dudley VJ, Beussink L, et al. Regulation of hypoxia-induced pulmonary hypertension by vascular smooth muscle hypoxia-inducible factor-1α. Am J Respir Crit Care Med 2014;189(3):314–324. doi:10.1164/rccm.201302-0302OC. [DOI] [PMC free article] [PubMed]
  • 49.Hickey MM, Lam JC, Bezman NA, Rathmell WK, Simon MC. von Hippel-Lindau mutation in mice recapitulates Chuvash polycythemia via hypoxia-inducible factor-2α signaling and splenic erythropoiesis. J Clin Investig 2007;117(12):3879–3889. doi:10.1172/JCI32614. [DOI] [PMC free article] [PubMed]
  • 50.Kapitsinou PP, Rajendran G, Astleford L, Michael M, Schonfeld MP, Fields T, Shay S, French JL, West J, Haase VH. The endothelial prolyl-4-hydroxylase domain 2/hypoxia-inducible factor 2 axis regulates pulmonary artery pressure in mice. Mol Cell Biol 2016;36(10):1584–1594. doi:10.1128/MCB.01055-15. [DOI] [PMC free article] [PubMed]
  • 51.Pawlus MR, Hu CJ. Enhanceosomes as integrators of hypoxia inducible factor (HIF) and other transcription factors in the hypoxic transcriptional response. Cell Signal 2013;25(9):1895–1903. doi:10.1016/j.cellsig.2013.05.018. [DOI] [PMC free article] [PubMed]
  • 52.Merika M, Williams AJ, Chen G, Collins T, Thanos D. Recruitment of CBP/p300 by the IFNβ enhanceosome is required for synergistic activation of transcription. Mol Cell 1998;1(2):277–287. [DOI] [PubMed]
  • 53.Carey M. The enhanceosome and transcriptional synergy. Cell 1998;92(1):5–8. [DOI] [PubMed]
  • 54.Freedman SJ, Sun ZY, Poy F, Kung AL, Livingston DM, Wagner G, Eck MJ. Structural basis for recruitment of CBP/p300 by hypoxia-inducible factor-1α. Proc Natl Acad Sci USA 2002;99(8):5367–5372. doi:10.1073/pnas.082117899. [DOI] [PMC free article] [PubMed]
  • 55.Kopan R, Ilagan MX. The canonical Notch signaling pathway: unfolding the activation mechanism. Cell 2009;137(2):216–233. doi:10.1016/j.cell.2009.03.045. [DOI] [PMC free article] [PubMed]
  • 56.Zhou S, Hayward SD. Nuclear localization of CBF1 is regulated by interactions with the SMRT corepressor complex. Mol Cell Biol 2001;21(18):6222–6232. [DOI] [PMC free article] [PubMed]
  • 57.Ranganathan P, Weaver KL, Capobianco AJ. Notch signalling in solid tumours: a little bit of everything but not all the time. Nat Rev Cancer 2011;11(5):338–351. doi:10.1038/nrc3035. [DOI] [PubMed]
  • 58.Miele L. Transcription factor RBPJ/CSL: a genome-wide look at transcriptional regulation. Proc Natl Acad Sci USA 2011;108(36):14715–14716. doi:10.1073/pnas.1110570108. [DOI] [PMC free article] [PubMed]
  • 59.Yamamura H, Yamamura A, Ko EA, Pohl NM, Smith KA, Zeifman A, Powell FL, Thistlethwaite PA, Yuan JX. Activation of Notch signaling by short-term treatment with Jagged-1 enhances store-operated Ca2+ entry in human pulmonary arterial smooth muscle cells. Am J Physiol Cell Physiol 2014;306(9):C871–C878. doi:10.1152/ajpcell.00221.2013. [DOI] [PMC free article] [PubMed]
  • 60.Smith KA, Voiriot G, Tang H, Fraidenburg DR, Song S, Yamamura H, Yamamura A, et al. Notch activation of Ca2+ signaling in the development of hypoxic pulmonary vasoconstriction and pulmonary hypertension. Am J Respir Cell Mol Biol 2015;53(3):355–367. doi:10.1165/rcmb.2014-0235OC. [DOI] [PMC free article] [PubMed]
  • 61.Firth AL, Remillard CV, Yuan JX. TRP channels in hypertension. Biochim Biophys Acta 2007;1772(8):895–906. doi:10.1016/j.bbadis.2007.02.009. [DOI] [PMC free article] [PubMed]
  • 62.Song MY, Makino A, Yuan JX. STIM2 contributes to enhanced store-operated Ca2+ entry in pulmonary artery smooth muscle cells from patients with idiopathic pulmonary arterial hypertension. Pulm Circ 2011;1(1):84–94. doi:10.4103/2045-8932.78106. [DOI] [PMC free article] [PubMed]
  • 63.Fuchs B, Dietrich A, Gudermann T, Kalwa H, Grimminger F, Weissmann N. The role of classical transient receptor potential channels in the regulation of hypoxic pulmonary vasoconstriction. Adv Exp Med Biol 2010;661:187–200. doi:10.1007/978-1-60761-500-2_12. [DOI] [PubMed]
  • 64.Lin MJ, Leung GP, Zhang WM, Yang XR, Yip KP, Tse CM, Sham JS. Chronic hypoxia-induced upregulation of store-operated and receptor-operated Ca2+ channels in pulmonary arterial smooth muscle cells: a novel mechanism of hypoxic pulmonary hypertension. Circ Res 2004;95(5):496–505. doi:10.1161/01.RES.0000138952.16382.ad. [DOI] [PubMed]
  • 65.Yamamura A, Guo Q, Yamamura H, Zimnicka AM, Pohl NM, Smith KA, Fernandez RA, et al. Enhanced Ca2+-sensing receptor function in idiopathic pulmonary arterial hypertension. Circ Res 2012;111(4):469–481. doi:10.1161/CIRCRESAHA.112.266361. [DOI] [PMC free article] [PubMed]
  • 66.Wang J, Weigand L, Lu W, Sylvester JT, Semenza GL, Shimoda LA. Hypoxia inducible factor 1 mediates hypoxia-induced TRPC expression and elevated intracellular Ca2+ in pulmonary arterial smooth muscle cells. Circ Res 2006;98(12):1528–1537. [DOI] [PubMed]
  • 67.Yu Y, Keller SH, Remillard CV, Safrina O, Nicholson A, Zhang SL, Jiang W, et al. A functional single-nucleotide polymorphism in the TRPC6 gene promoter associated with idiopathic pulmonary arterial hypertension. Circulation 2009;119(17):2313–2322. doi:10.1161/CIRCULATIONAHA.108.782458. [DOI] [PMC free article] [PubMed]
  • 68.Golovina VA, Platoshyn O, Bailey CL, Wang J, Limsuwan A, Sweeney M, Rubin LJ, Yuan JX. Upregulated TRP and enhanced capacitative Ca2+ entry in human pulmonary artery myocytes during proliferation. Am J Physiol Heart Circ Physiol 2001;280(2):H746–H755. [DOI] [PubMed]
  • 69.Sylvester JT, Shimoda LA, Aaronson PI, Ward JP. Hypoxic pulmonary vasoconstriction. Physiol Rev 2012;92(1):367–520. doi:10.1152/physrev.00041.2010. [DOI] [PMC free article] [PubMed]
  • 70.Stenmark KR, Fagan KA, Frid MG. Hypoxia-induced pulmonary vascular remodeling: cellular and molecular mechanisms. Circ Res 2006;99(7):675–691. doi:10.1161/01.RES.0000243584.45145.3f. [DOI] [PubMed]
  • 71.Altenberg B, Greulich KO. Genes of glycolysis are ubiquitously overexpressed in 24 cancer classes. Genomics 2004;84(6):1014–1020. doi:10.1016/j.ygeno.2004.08.010. [DOI] [PubMed]
  • 72.Yang W, Zheng Y, Xia Y, Ji H, Chen X, Guo F, Lyssiotis CA, Aldape K, Cantley LC, Lu Z. ERK1/2-dependent phosphorylation and nuclear translocation of PKM2 promotes the Warburg effect. Nat Cell Biol 2012;14(12):1295–1304. doi:10.1038/ncb2629. [DOI] [PMC free article] [PubMed]
  • 73.Lv L, Xu YP, Zhao D, Li FL, Wang W, Sasaki N, Jiang Y, et al. Mitogenic and oncogenic stimulation of K433 acetylation promotes PKM2 protein kinase activity and nuclear localization. Mol Cell 2013;52(3):340–352. doi:10.1016/j.molcel.2013.09.004. [DOI] [PMC free article] [PubMed]
  • 74.Wang R, Asangani IA, Chakravarthi BV, Ateeq B, Lonigro RJ, Cao Q, Mani RS, et al. Role of transcriptional corepressor CtBP1 in prostate cancer progression. Neoplasia 2012;14(10):905–914. [DOI] [PMC free article] [PubMed]
  • 75.Yang W, Xia Y, Hawke D, Li X, Liang J, Xing D, Aldape K, Hunter T, Yung WK, Lu Z. PKM2 phosphorylates histone H3 and promotes gene transcription and tumorigenesis. Cell 2012;150(4):685–696. doi:10.1016/j.cell.2012.07.018. [DOI] [PMC free article] [PubMed]
  • 76.Luo W, Hu H, Chang R, Zhong J, Knabel M, O’Meally R, Cole RN, Pandey A, Semenza GL. Pyruvate kinase M2 is a PHD3-stimulated coactivator for hypoxia-inducible factor 1. Cell 2011;145(5):732–744. doi:10.1016/j.cell.2011.03.054. [DOI] [PMC free article] [PubMed]
  • 77.Jiang Y, Wang Y, Wang T, Hawke DH, Zheng Y, Li X, Zhou Q, et al. PKM2 phosphorylates MLC2 and regulates cytokinesis of tumour cells. Nat Commun 2014;5:5566. doi:10.1038/ncomms6566. [DOI] [PMC free article] [PubMed]
  • 78.Bluemlein K, Grüning NM, Feichtinger RG, Lehrach H, Kofler B, Ralser M. No evidence for a shift in pyruvate kinase PKM1 to PKM2 expression during tumorigenesis. Oncotarget 2011;2(5):393–400. doi:10.18632/oncotarget.278. [DOI] [PMC free article] [PubMed]
  • 79.Desai S, Ding M, Wang B, Lu Z, Zhao Q, Shaw K, Yung WK, Weinstein JN, Tan M, Yao J. Tissue-specific isoform switch and DNA hypomethylation of the pyruvate kinase PKM gene in human cancers. Oncotarget 2014;5(18):8202–8210. doi:10.18632/oncotarget.1159. [DOI] [PMC free article] [PubMed]
  • 80.Das M, Burns N, Wilson SJ, Zawada WM, Stenmark KR. Hypoxia exposure induces the emergence of fibroblasts lacking replication repressor signals of PKCζ in the pulmonary artery adventitia. Cardiovasc Res 2008;78(3):440–448. doi:10.1093/cvr/cvn014. [DOI] [PubMed]
  • 81.El Kasmi KC, Pugliese SC, Riddle SR, Poth JM, Anderson AL, Frid MG, Li M, et al. Adventitial fibroblasts induce a distinct proinflammatory/profibrotic macrophage phenotype in pulmonary hypertension. J Immunol 2014;193(2):597–609. doi:10.4049/jimmunol.1303048. [DOI] [PMC free article] [PubMed]
  • 82.Krick S, Hänze J, Eul B, Savai R, Seay U, Grimminger F, Lohmeyer J, Klepetko W, Seeger W, Rose F. Hypoxia-driven proliferation of human pulmonary artery fibroblasts: cross-talk between HIF-1α and an autocrine angiotensin system. FASEB J 2005;19(7):857–859. doi:10.1096/fj.04-2890fje. [DOI] [PubMed]
  • 83.Li M, Riddle SR, Frid MG, El Kasmi KC, McKinsey TA, Sokol RJ, Strassheim D, et al. Emergence of fibroblasts with a proinflammatory epigenetically altered phenotype in severe hypoxic pulmonary hypertension. J Immunol 2011;187(5):2711–2722. doi:10.4049/jimmunol.1100479. [DOI] [PMC free article] [PubMed]
  • 84.Paulin R, Michelakis ED. The metabolic theory of pulmonary arterial hypertension. Circ Res 2014;115(1):148–164. doi:10.1161/CIRCRESAHA.115.301130. [DOI] [PubMed]
  • 85.Zhao L, Ashek A, Wang L, Fang W, Dabral S, Dubois O, Cupitt J, et al. Heterogeneity in lung 18FDG uptake in pulmonary arterial hypertension: potential of dynamic 18FDG positron emission tomography with kinetic analysis as a bridging biomarker for pulmonary vascular remodeling targeted treatments. Circulation 2013;128(11):1214–1224. doi:10.1161/CIRCULATIONAHA.113.004136. [DOI] [PubMed]
  • 86.Chinnadurai G. The transcriptional corepressor CtBP: a foe of multiple tumor suppressors. Cancer Res 2009;69(3):731–734. doi:10.1158/0008-5472.CAN-08-3349. [DOI] [PMC free article] [PubMed]
  • 87.Peña C, García JM, García V, Silva J, Domínguez G, Rodríguez R, Maximiano C, García de Herreros A, Muñoz A, Bonilla F. The expression levels of the transcriptional regulators p300 and CtBP modulate the correlations between SNAIL, ZEB1, E-cadherin and vitamin D receptor in human colon carcinomas. Int J Cancer 2006;119(9):2098–2104. doi:10.1002/ijc.22083. [DOI] [PubMed]
  • 88.Di LJ, Byun JS, Wong MM, Wakano C, Taylor T, Bilke S, Baek S, et al. Genome-wide profiles of CtBP link metabolism with genome stability and epithelial reprogramming in breast cancer. Nat Commun 2013;4:1449. doi:10.1038/ncomms2438. [DOI] [PMC free article] [PubMed]
  • 89.Grooteclaes M, Deveraux Q, Hildebrand J, Zhang Q, Goodman RH, Frisch SM. C-terminal-binding protein corepresses epithelial and proapoptotic gene expression programs. Proc Natl Acad Sci USA 2003;100(8):4568–4573. doi:10.1073/pnas.0830998100. [DOI] [PMC free article] [PubMed]
  • 90.Zhang G, Yan Y, Kang L, Cao Q, Ke K, Wu X, Gao, Y, et al. Involvement of CtBP2 in LPS-induced microglial activation. J Mol Histol 2012;43(3):327–334. doi:10.1007/s10735-012-9399-x. [DOI] [PubMed]
  • 91.Byun JS, Gardner K. C-terminal binding protein: a molecular link between metabolic imbalance and epigenetic regulation in breast cancer. Int J Cell Biol 2013;2013:647975. doi:10.1155/2013/647975. [DOI] [PMC free article] [PubMed]
  • 92.Stenmark KR, Frid M, Perros F. Endothelial-to-mesenchymal transition: an evolving paradigm and a promising therapeutic target in PAH. Circulation 2016;133(18):1734–1737. doi:10.1161/CIRCULATIONAHA.116.022479. [DOI] [PMC free article] [PubMed]
  • 93.Hayashi M, Nimura K, Kashiwagi K, Harada T, Takaoka K, Kato H, Tamai K, Kaneda Y. Comparative roles of Twist-1 and Id1 in transcriptional regulation by BMP signaling. J Cell Sci 2007;120(8):1350–1357. doi:10.1242/jcs.000067. [DOI] [PubMed]
  • 94.Dobrian AD. A tale with a Twist: a developmental gene with potential relevance for metabolic dysfunction and inflammation in adipose tissue. Front Endocrinol (Lausanne) 2012;3:108. doi:10.3389/fendo.2012.00108. [DOI] [PMC free article] [PubMed]
  • 95.Lu S, Nie J, Luan Q, Feng Q, Xiao Q, Chang Z, Shan C, Hess D, Hemmings BA, Yang Z. Phosphorylation of the Twist1-family basic helix-loop-helix transcription factors is involved in pathological cardiac remodeling. PLoS ONE 2011;6(4):e19251. doi:10.1371/journal.pone.0019251. [DOI] [PMC free article] [PubMed]
  • 96.Mastroyiannopoulos NP, Antoniou AA, Koutsoulidou A, Uney JB, Phylactou LA. Twist reverses muscle cell differentiation through transcriptional down-regulation of myogenin. Biosci Rep 2013;33(6):e00083. doi:10.1042/BSR20130068. [DOI] [PMC free article] [PubMed]
  • 97.Doody RS, Raman R, Farlow M, Iwatsubo T, Vellas B, Joffe S, Kieburtz K, et al. A phase 3 trial of semagacestat for treatment of Alzheimer’s disease. N Engl J Med 2013;369(4):341–350. doi:10.1056/NEJMoa1210951. [DOI] [PubMed]
  • 98.Siemers ER, Quinn JF, Kaye J, Farlow MR, Porsteinsson A, Tariot P, Zoulnouni P, et al. Effects of a γ-secretase inhibitor in a randomized study of patients with Alzheimer disease. Neurology 2006;66(4):602–604. doi:10.1212/01.WNL.0000198762.41312.E1. [DOI] [PubMed]
  • 99.Goldberg AD, Allis CD, Bernstein E. Epigenetics: a landscape takes shape. Cell 2007;128(4):635–638. doi:10.1016/j.cell.2007.02.006. [DOI] [PubMed]
  • 100.Luo RX, Dean DC. Chromatin remodeling and transcriptional regulation. J Natl Cancer Inst 1999;91(15):1288–1294. [DOI] [PubMed]
  • 101.Bönisch C, Hake SB. Histone H2A variants in nucleosomes and chromatin: more or less stable? Nucleic Acids Res 2012;40(21):10719–10741. doi:10.1093/nar/gks865. [DOI] [PMC free article] [PubMed]
  • 102.Germain M, Eyries M, Montani D, Poirier O, Girerd B, Dorfmüller P, Coulet F, et al. Genome-wide association analysis identifies a susceptibility locus for pulmonary arterial hypertension. Nat Genet 2013;45(5):518–521. doi:10.1038/ng.2581. [DOI] [PMC free article] [PubMed]
  • 103.Kim JD, Lee A, Choi J, Park Y, Kang H, Chang W, Lee MS, Kim J. Epigenetic modulation as a therapeutic approach for pulmonary arterial hypertension. Exp Mol Med 2015;47:e175. doi:10.1038/emm.2015.45. [DOI] [PMC free article] [PubMed]
  • 104.Stenmark KR, Frid MG, Yeager M, Li M, Riddle S, McKinsey T, El Kasmi KC. Targeting the adventitial microenvironment in pulmonary hypertension: a potential approach to therapy that considers epigenetic change. Pulm Circ 2012;2(1):3–14. doi:10.4103/2045-8932.94817. [DOI] [PMC free article] [PubMed]
  • 105.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(1):L1–L8. doi:10.1152/ajplung.00378.2006. [DOI] [PubMed]
  • 106.Gupta S, Li L. Modulation of miRNAs in pulmonary hypertension. Int J Hypertens 2015;2015:169069. doi:10.1155/2015/169069. [DOI] [PMC free article] [PubMed]
  • 107.Nielsen HM, Tost J. Epigenetic changes in inflammatory and autoimmune diseases. Subcell Biochem 2013;61:455–478. doi:10.1007/978-94-007-4525-4_20. [DOI] [PubMed]
  • 108.Archer SL, Marsboom G, Kim GH, Zhang HJ, Toth PT, Svensson EC, Dyck JR, et al. Epigenetic attenuation of mitochondrial superoxide dismutase 2 in pulmonary arterial hypertension: a basis for excessive cell proliferation and a new therapeutic target. Circulation 2010;121(24):2661–2671. doi:10.1161/CIRCULATIONAHA.109.916098. [DOI] [PMC free article] [PubMed]
  • 109.Hurt EM, Thomas SB, Peng B, Farrar WL. Molecular consequences of SOD2 expression in epigenetically silenced pancreatic carcinoma cell lines. Br J Cancer 2007;97(8):1116–1123. doi:10.1038/sj.bjc.6604000. [DOI] [PMC free article] [PubMed]
  • 110.Duffy MJ, Napieralski R, Martens JW, Span PN, Spyratos F, Sweep FC, Brunner N, Foekens JA, Schmitt M. Methylated genes as new cancer biomarkers. Eur J Cancer 2009;45(3):335–346. doi:10.1016/j.ejca.2008.12.008. [DOI] [PubMed]
  • 111.Montani D, Lau EM, Dorfmüller P, Girerd B, Jaïs X, Savale L, Perros F, et al. Pulmonary veno-occlusive disease. Eur Respir J 2016;47(5):1518–1534. doi:10.1183/13993003.00026-2016. [DOI] [PubMed]
  • 112.Perros F, Cohen-Kaminsky S, Gambaryan N, Girerd B, Raymond N, Klingelschmitt I, Huertas A, et al. Cytotoxic cells and granulysin in pulmonary arterial hypertension and pulmonary veno-occlusive disease. Am J Respir Crit Care Med 2013;187(2):189–196. doi:10.1164/rccm.201208-1364OC. [DOI] [PubMed]
  • 113.Talati M, Seeley E, Ihida-Stansbury K, Delisser H, McDonald H, Ye F, Zhang X, Shyr Y, Caprioli R, Meyrick B. Altered expression of nuclear and cytoplasmic histone H1 in pulmonary artery and pulmonary artery smooth muscle cells in patients with IPAH. Pulm Circ 2012;2(3):340–351. doi:10.4103/2045-8932.101645. [DOI] [PMC free article] [PubMed]
  • 114.Cavasin MA, Demos-Davies K, Horn TR, Walker LA, Lemon DD, Birdsey N, Weiser-Evans MC, et al. Selective class I histone deacetylase inhibition suppresses hypoxia-induced cardiopulmonary remodeling through an antiproliferative mechanism. Circ Res 2012;110(5):739–748. doi:10.1161/CIRCRESAHA.111.258426. [DOI] [PMC free article] [PubMed]
  • 115.Zhao L, Chen CN, Hajji N, Oliver E, Cotroneo E, Wharton J, Wang D, et al. Histone deacetylation inhibition in pulmonary hypertension: therapeutic potential of valproic acid and suberoylanilide hydroxamic acid. Circulation 2012;126(4):455–467. doi:10.1161/CIRCULATIONAHA.112.103176. [DOI] [PMC free article] [PubMed]
  • 116.Bogaard HJ, Mizuno S, Hussaini AA, Toldo S, Abbate A, Kraskauskas D, Kasper M, Natarajan R, Voelkel NF. Suppression of histone deacetylases worsens right ventricular dysfunction after pulmonary artery banding in rats. Am J Respir Crit Care Med 2011;183(10):1402–1410. [DOI] [PubMed]
  • 117.Yang Q, Dahl MJ, Albertine KH, Ramchandran R, Sun M, Raj JU. Role of histone deacetylases in regulation of phenotype of ovine newborn pulmonary arterial smooth muscle cells. Cell Prolif 2013;46(6):654–664. [DOI] [PMC free article] [PubMed]
  • 118.Cantoni S, Galletti M, Zambelli F, Valente S, Ponti F, Tassinari R, Pasquinelli G, Galiè N, Ventura C. Sodium butyrate inhibits platelet-derived growth factor-induced proliferation and migration in pulmonary artery smooth muscle cells through Akt inhibition. FEBS J 2013;280(9):2042–2055. [DOI] [PubMed]
  • 119.Galletti M, Cantoni S, Zambelli F, Valente S, Palazzini M, Manes A, Pasquinelli G, Mai A, Galiè N, Ventura C. Dissecting histone deacetylase role in pulmonary arterial smooth muscle cell proliferation and migration. Biochem Pharmacol 2014;91(2):181–190. [DOI] [PubMed]
  • 120.Wang D, Zhang H, Li M, Frid MG, Flockton AR, McKeon BA, Yeager ME, et al. MicroRNA-124 controls the proliferative, migratory, and inflammatory phenotype of pulmonary vascular fibroblasts. Circ Res 2014;114(1):67–78. [DOI] [PMC free article] [PubMed]
  • 121.De Raaf MA, Hussaini AA, Gomez-Arroyo J, Kraskaukas D, Farkas D, Happé C, Voelkel NF, Bogaard HJ. Histone deacetylase inhibition with trichostatin A does not reverse severe angioproliferative pulmonary hypertension in rats (2013 Grover Conference series). Pulm Circ 2014;4(2):237–243. [DOI] [PMC free article] [PubMed]
  • 122.Soon E, Crosby A, Southwood M, Yang P, Tajsic T, Toshner M, Appleby S, et al. Bone morphogenetic protein receptor type II deficiency and increased inflammatory cytokine production: a gateway to pulmonary arterial hypertension. Am J Respir Crit Care Med 2015;192(7):859–872. doi:10.1164/rccm.201408-1509OC. [DOI] [PMC free article] [PubMed]
  • 123.Perros F, Bonnet S. Bone morphogenetic protein receptor type II and inflammation are bringing old concepts into the new pulmonary arterial hypertension world. Am J Respir Crit Care Med 2015;192(7):777–779. doi:10.1164/rccm.201506-1115ED. [DOI] [PubMed]
  • 124.Xu XF, Cheng F, Du LZ. Epigenetic regulation of pulmonary arterial hypertension. Hypertens Res 2011;34(9):981–986. doi:10.1038/hr.2011.79. [DOI] [PubMed]

Articles from Pulmonary Circulation are provided here courtesy of Wiley

RESOURCES