Skip to main content
American Journal of Physiology - Heart and Circulatory Physiology logoLink to American Journal of Physiology - Heart and Circulatory Physiology
. 2022 Feb 25;322(5):H702–H724. doi: 10.1152/ajpheart.00021.2022

Mechanisms of pulmonary vascular dysfunction in pulmonary hypertension and implications for novel therapies

Helen Christou 1, Raouf A Khalil 2,
PMCID: PMC8977136  PMID: 35213243

Abstract

Pulmonary hypertension (PH) is a serious disease characterized by various degrees of pulmonary vasoconstriction and progressive fibroproliferative remodeling and inflammation of the pulmonary arterioles that lead to increased pulmonary vascular resistance, right ventricular hypertrophy, and failure. Pulmonary vascular tone is regulated by a balance between vasoconstrictor and vasodilator mediators, and a shift in this balance to vasoconstriction is an important component of PH pathology, Therefore, the mainstay of current pharmacological therapies centers on pulmonary vasodilation methodologies that either enhance vasodilator mechanisms such as the NO-cGMP and prostacyclin-cAMP pathways and/or inhibit vasoconstrictor mechanisms such as the endothelin-1, cytosolic Ca2+, and Rho-kinase pathways. However, in addition to the increased vascular tone, many patients have a “fixed” component in their disease that involves altered biology of various cells in the pulmonary vascular wall, excessive pulmonary artery remodeling, and perivascular fibrosis and inflammation. Pulmonary arterial smooth muscle cell (PASMC) phenotypic switch from a contractile to a synthetic and proliferative phenotype is an important factor in pulmonary artery remodeling. Although current vasodilator therapies also have some antiproliferative effects on PASMCs, they are not universally successful in halting PH progression and increasing survival. Mild acidification and other novel approaches that aim to reverse the resident pulmonary vascular pathology and structural remodeling and restore a contractile PASMC phenotype could ameliorate vascular remodeling and enhance the responsiveness of PH to vasodilator therapies.

Keywords: endothelium, remodeling, smooth muscle, vasoconstriction, vasodilation

INTRODUCTION

Pulmonary hypertension (PH) comprises a heterogeneous group of disorders with diverse etiology and pathogenetic mechanisms. PH affects individuals of all ages and is an expanding public health problem in the United States and globally. In healthy subjects with normal pulmonary circulation, the mean pulmonary artery pressure (mPAP) is 14.0 ± 3 mmHg with an upper limit of 20 mmHg. In all forms of PH there is disruption of pulmonary vascular homeostasis and increased mPAP >20 mmHg (1). In severe PH, mPAP could reach ≥35 mmHg. Clinical symptomatology of PH includes dyspnea on exertion that progressively worsens to stages I–IV as described in the World Health Organization (WHO) functional classification (Table 1) (2). PH progresses rapidly and eventually leads to right ventricular hypertrophy and failure that are ultimately fatal (3). Despite promising therapeutic approaches, the mortality rates from PH remain unacceptably high and could be accelerated by COVID-19-related complications. Prognosis of PH is poor and early detection and management is critical, making it important to understand the underlying factors and mechanisms.

Table 1.

WHO functional classification of pulmonary hypertension

Functional Class Symptomatology
I No limitations of usual physical activity
II No symptoms at rest, but dyspnea, fatigue, or chest pains during usual activity
III No symptoms at rest, but limitations of usual activity due to dyspnea, fatigue, chest pain, or syncope
IV Symptoms at rest

See Galie et al. (2). WHO, World Health Organization.

Several underlying cardiopulmonary and autoimmune disorders as well as genetic, racial, gender-related, and environmental factors have been suggested as predisposing factors in PH. Also, extensive research has helped to identify some of the pathophysiological components of PH including increased pulmonary arteriolar tone, and pulmonary vascular wall thickening, remodeling, fibrosis, and inflammation (2, 4). Specifically, substantial progress has been made in our understanding of the mechanisms of dysregulated pulmonary vascular tone in both human PH and preclinical animal models. This knowledge was successfully leveraged to develop vasodilator therapies that are variably effective in improving symptoms and prolonging survival among patients with PH. For instance, identifying the alterations in endothelium-derived vasoactive mediators such as nitric oxide (NO), prostacyclin (PGI2) and endothelin-1 (ET-1) have led to their therapeutic targeting using phosphodiesterase-5 (PDE5) inhibitors (PDE5i), cGMP, and PGI2 analogs, and ET-1 antagonists (59). Combination treatments targeting complementary endothelium-dependent pathways also aim to enhance the vasodilation responsiveness of the PH circulation to levels better than those achieved by each of the vasodilators when used separately (1012). In addition to endothelial cell dysfunction, enhanced pulmonary arterial smooth muscle cell (PASMC) contraction mechanisms such as Ca2+ and Rho-kinase could enhance pulmonary vascular tone in PH. Aberrant transforming growth factor-β (TGFβ)/bone morphogenetic protein (BMP) signaling, increased proliferation of PASMCs and fibroblasts, excessive extracellular matrix (ECM) deposition, fibrosis, and inflammation also contribute to abnormal pulmonary vascular remodeling in PH. Therefore, many patients with PH do not respond adequately to vasodilators and their quality of life remains markedly compromised due to progressive dyspnea and exertional fatigue that could lead to ultimate immobilization.

Although PH is increasingly recognized as a complex disease with different underlying mechanisms and multiple pulmonary and cardiovascular organs involved, current therapeutic approaches remain primarily focused on correcting the disrupted pulmonary vascular homeostasis and reducing pulmonary vascular resistance (PVR) by counterbalancing the residual vasoconstriction using vasodilation rather than targeting key regulators of the cellular pathology. Although therapeutic efforts should also target other components of the PH disease including the right ventricle response to high PVR, skeletal muscle dysfunction, and other extrapulmonary manifestations, there remains an unmet clinical need to further examine, evaluate, and enhance the effectiveness of current therapeutic approaches for PH. In this respect, it is increasingly appreciated that a strategy to restore the structural and functional integrity of the pulmonary vessel wall could open new avenues for PH therapies.

This review will discuss reports in PubMed and Medline databases and data from our laboratories with emphasis on the current understanding of the cellular and molecular mechanisms of the disrupted pulmonary vascular homeostasis in PH, the current therapeutic approaches, and implications for potential novel targets. Clinical data in human PH and preclinical data in experimental models of PH will be discussed. The review will highlight the various predisposing factors of PH, the pathological components of the disease including pulmonary vascular dysfunction, remodeling, fibrosis, and inflammation, and the underlying vascular and cellular mechanisms. The review will also describe how the interaction between different cell types and overlapping mechanisms could add to the complexity of PH pathology. Further understanding of these mechanisms should help to identify new targets and develop novel therapeutic strategies to restore pulmonary vascular homeostasis in PH.

PH CLASSIFICATION, GROUPS, AND EPIDEMIOLOGY

The WHO guidelines classify patients with PH into five groups (Table 2) (13, 14). Pulmonary arterial hypertension (PAH, group 1 PH) is uncommon and is estimated to affect between 10 and 50 people per million worldwide. PAH is associated with extensive PASMC proliferation, vascular remodeling, and obliteration of the small pulmonary arterioles, leading to progressive increase in PVR (15).

Table 2.

World Health Organization clinical classification of PH

Group Mechanisms Examples
1 Pulmonary arterial hypertension Idiopathic, heritable, drug-induced, viral (HIV) or parasitic disease (schistosomiasis), connective tissue disorders, liver cirrhosis, congenital heart disease, PPHN
2 PH due to left heart failure Left-sided atrial, ventricular, or valvular disease
3 PH due to chronic hypoxemic lung disease COPD, interstitial lung disease, obstructive sleep apnea, high-altitude, developmental lung diseases
4 Chronic thromboembolic PH Pulmonary emboli
5 Miscellaneous or unclear Sickle cell disease, sarcoidosis, metabolic disorders, renal failure

See Gelzinis (13) and Walter (14). COPD, chronic obstructive pulmonary disease; PH, pulmonary hypertension; PPHN, persistent pulmonary hypertension of the newborn.

Although PH is considered a rare disease (fewer than 1 in 2,000 individuals in Europe and fewer than 200,000 in the United States, with 500–1,000 new cases per year), it affects individuals of all ages and is an expanding public health problem in the United States and globally as the population ages (16). It is estimated that 1% of the global population suffers from PH, and this percentage rises to 10% in individuals over the age of 65 yr (16). The prevalence of PH in the United States is estimated at 109/million under the age of 65 and 451/million over the age of 65 (17). Importantly, 80% of the affected global population live in developing countries and their disease is associated with uncorrected congenital heart disease, rheumatic disease, HIV, and parasitic disease (schistosomiasis) (16). The increasing prevalence with advancing age is primarily due to PH in association with left-sided heart disease and lung disease. Among patients with PH in developed countries, the 1-year survival is 83%–91% and the 3-year survival is 58%–72.1% (18, 19). The disease burden in the pediatric population is less well defined with data derived primarily from regional registry cohorts. The estimated prevalence is 20–40 cases per million in Europe (20, 21) and 26–33 per million children in the United States (22). One study in the United States has reported that PH is associated with 0.13% of pediatric hospitalizations and with 5.9% of all-cause mortality (23). In the pediatric population, there is also increasing proportion of patients with PH without associated congenital heart disease especially among surviving preterm infants with or without bronchopulmonary dysplasia (23, 24).

PH PREDISPOSING FACTORS

PH is a complex disorder and often a manifestation of an underlying severe cardiopulmonary, autoimmune, or metabolic disease. There is a diverse group of predisposing genetic and environmental factors that contribute to the various morbidities associated with each group of PH. Bacterial and viral infection, air pollution, and nicotine and E-cigarette smoking are important environmental factors that could predispose to PH.

Specifically, although PAH is uncommon, several predisposing genetic, gender-related, and environmental factors have been suggested. Mutation in the bone morphogenetic protein receptor type 2 (BMPR2) gene could be responsible for 80% of heritable PAH and 20% of sporadic PAH. Disruption of BMPR2 signaling is observed in different cells of the pulmonary vasculature including endothelial cells, PASMCs, and fibroblasts. BMPR2 also regulates the immune system, and disruption of BMPR2 signaling disrupts the immune response and increases inflammation (25). Loss of function KCNK3 gene mutation could also alter the circulating immune cells and the inflammation response and enhance susceptibility to PAH (26). Whole exome sequencing (WES) has identified SRY-related HMG box transcription factor (SOX17) as a risk gene and suggested that its deficiency may predispose to PAH (27). Also, WES in two unrelated families with PAH cases showed that TNIP2 and TRAF2 gene loss of function could promote pulmonary vascular remodeling through exaggerated NF-κB signaling (28). WES in 242 Japanese patients with familial or sporadic PAH identified a heterozygous substitution change involving c.226G>A (p.Gly76Ser) in tumor necrosis factor receptor superfamily 13B gene (TNFRSF13B) in six (2.5%) patients. TNFRSF13B controls the differentiation of B cell and secretion of inflammatory cytokines and may be involved in vascular inflammation (29). Also, in a cohort of 79 patients with PAH, four pathogenic/likely pathogenic variants in four different genes (TBX4 encoding T-box 4-containing protein, ABCC8, KCNA5, and GDF2/BMP9) were identified in nine patients (30). Mutant CPS-I A/C rs4399666 minor variant particularly the homozygous CC genotype is frequently distributed among neonates with persistent pulmonary hypertension of the newborn (PPHN) (31). In a cohort of 60 patients with PAH, single-nucleotide polymorphisms (SNPs) for genes encoding the mitochondrial proteins Sirtuin3 (Sirt3) and uncoupling protein 2 (Ucp2) were associated with disease severity and death or need for lung transplant and 10-year outcomes. Also, double knockout mice for Sirt3 and Ucp2 genes show greater severity of PAH, mPAP, right ventricular hypertrophy/dilatation, vascular remodeling, and inflammatory plexogenic lesions compared with wild-type mice (32). Rare variant analysis of 4,241 PAH cases from an international consortium identified seven genes with rare deleterious variants associated with idiopathic PAH including BMPR2, GDF2, TBX4, SOX17, and KDR and two new candidate genes encoding fibulin 2 (FBLN2) and platelet-derived growth factor D (PDGFD) (33).

Epigenetic mechanisms including DNA methylation and histone posttranslational modifications regulate chromatin structure and lineage-specific gene expression during cardiovascular development and disease. Analysis of cells and tissues isolated from patients with PAH and preclinical animal models of PH showed alterations in the levels of multiple histone deacetylases, SIRT1, SIRT3, and bromodomain-containing protein 4 (BRD4) and their strong association to proliferative, fibrotic, and inflammatory phenotypes that lead to pulmonary artery remodeling (34). Also, histone deacetylase 1 is upregulated in monocrotaline (MCT) model of PH, and its inhibition reduces ECM accumulation, pulmonary arterial remodeling, right ventricular systolic pressure (RVSP), and right ventricle hypertrophy index. The underlying mechanism involves upregulation of miR-34a and modulation of matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases (TIMPS), specifically MMP-9/TIMP-1 and MMP-2/TIMP-2 activity (35).

Regarding gender differences, there is a predominance of young females affected by heritable PAH, but among patients with PAH, women show better prognosis and right ventricular function and higher survival rates than men (36, 37). The increased frequency of PAH among women has been partly related to increased expression/activity of the long-noncoding RNA X-inactive-specific transcript (Xist), which is essential for X-chromosome inactivation and dosage compensation of X-linked genes, leading to sexual dimorphism in female pulmonary artery endothelial cells (38). The increased susceptibility to PAH in women has also been related to estrogen/estrogen receptor signaling. Paradoxically, estrogen may have protective effects in animal models of PH, as the pathology is exaggerated in ovariectomized female animals. In isolated human pulmonary artery and PASMCs from non-PH subjects and group 3 patients with PH, estrogen pretreatment has shown variable effects ranging from no effect to enhancement of PGI2 synthesis and the levels of 6-keto-prostaglandin F1α (PGI2 stable metabolite), the PGI2 IP-receptor, and arachidonic acid-induced relaxation (39). Also, 17β-estradiol acting on estrogen receptor-α attenuates right ventricular failure in MCT rat model of PH by upregulating the procontractile and prosurvival peptide apelin via a BMPR2-dependent pathway (40). In chronic hypoxia (Hx) rat model of PH, treatment with the estrogen metabolite 2-metyhoxyestradiol reduced RVSP and pulmonary vascular remodeling (41). This “estrogen paradox” may be related to differences among species in the synthesis/metabolism of estrogens, the types, distribution and function of estrogen receptor, the post-estrogen receptor genomic and nongenomic signaling pathways, and changes in estrogen signaling during the development and progression of PH (36). These conflicting observations make it important to further examine the sex differences and the potential protective effects of estrogen in PH.

In addition to genetic and gender-related factors, a large number of drugs and toxins can induce PAH in susceptible individuals by known and unknown mechanisms. The first drugs observed to be causally related to PAH were the amphetamine-like appetite suppressants aminorex, fenfluramine, and dexfenfluramine. In addition to the use of anorexigens, there is increasing recognition of PAH susceptibility in association with the use of illicit drugs such as cocaine and amphetamines as well as medically prescribed drugs such as diazoxide and fluoxetine (42). Amphetamine-like appetite suppressants such as fenfluramine and dexfenfluramine could contribute as predisposing factors to PH pathogenesis through displacing norepinephrine and neurotransmitters. Other proposed underlying mechanisms include aberrant serotonin release, K+ channel dysregulation, and genetic predisposition.

PATHOPHYSIOLOGICAL COMPONENTS OF PH

PAH involves residual pulmonary vasoconstriction, progressive small vessel remodeling and obliteration, large vessel thickening and obstruction, and development of plexiform lesions (43). Increased PVR in PH is the result of 1) increased pulmonary arteriolar tone due to vasoconstriction and 2) structural remodeling of the pulmonary arterioles that includes PASMC proliferation perivascular fibrosis and inflammation causing obliteration of the vessel lumen and fixed obstruction (Fig. 1). Increased release of neurovascular mediators from pulmonary nerve endings could contribute to increased pulmonary vasoconstriction. Endothelial cell dysfunction is also a major factor in PAH (44). An imbalance between endothelial-derived vasodilators and vasoconstrictors has long been recognized as a modifiable contributor to the disease process, and consequently has been the target of most of the current vasodilator therapies (45). Changes in PASMC contraction pathways such as cytosolic Ca2+ concentration ([Ca2+]cyt) and Rho-kinase have also been linked to PH, and their targeting is promising, but less well developed (46, 47). The structural remodeling of pulmonary arterioles involves a complex interplay of multiple resident and circulating cells as well as humoral factors that collectively disrupt all vascular layers of the pulmonary arterioles and render them less responsive to current vasodilator therapies (48). Changes in growth factors, inflammatory elements, and the tissue pH could promote PASMC phenotypic switching from contractile to synthetic phenotype and activate multiple mediators of structural pulmonary vascular remodeling in PH (49, 50). This is an area of enormous potential as better mechanistic insights are accumulating.

Figure 1.

Figure 1.

Vascular mechanisms of pulmonary hypertension (PH). Genetic factors or underlying left cardiac, pulmonary hypoxic, thromboembolic, or autoimmune disease led to hypoxia and targeting of various cells in the pulmonary vasculature. In endothelial cells, decreased release of endothelium-derived relaxing factors (EDRFs) reduces pulmonary arterial smooth muscle cell (PASMC) relaxation, whereas increased endothelium-derived constricting factors (EDCFs) increases PASMC contraction, leading to increased pulmonary vascular tone. Increased sympathetic nerve ending activity could also increase pulmonary vasoconstriction. Phenotypic switching of PASMCs from contractile to synthetic phenotype promotes PASMC growth, proliferation, migration, and resistance to apoptosis, and leads to increased pulmonary arteriole medial thickening. Activation of fibroblasts and pericytes causes increases in the release of transforming growth factor-β (TGFβ) and matrix metalloproteinases (MMPs), decreases in tissue inhibitors of metalloproteinases (TIMPs), and increases in extracellular matrix (ECM), collagen deposition, fibrosis, and pulmonary arteriolar remodeling. Inflammatory cell infiltration, increased release of inflammatory cytokines, and macrophage polarization lead to pulmonary arteriole inflammation and further remodeling. The increased pulmonary arteriolar tone, medial thickening, and remodeling lead to increased pulmonary vascular resistance (PVR), pulmonary hypertension (PH), and right ventricular failure. Current vasodilator therapies aim at counteracting the residual pulmonary arteriolar narrowing, whereas mild acidification, antifibrotics, and anti-inflammatory drugs could provide adjuvant therapies to minimize pulmonary arteriolar obliteration and reduce PVR and PH. ECM, extracellular matrix; EDHF, endothelium-derived hyperpolarizing factor; ET-1, endothelin-1; ETAR, endothelin receptor type A; HIF, hypoxia-inducible factor; NO, nitric oxide; PDE5, phosphodiesterase-5 inhibitor; PGI2, prostacyclin.

Rodent models of experimental PH recapitulate to a certain extent the histologic features of human PH and provide a useful tool to study the efficacy of novel therapeutic approaches (5155). The MCT and Hx rat models are particularly useful in the evaluation of antiproliferative and proapoptotic therapies since medial hypertrophy of the pulmonary arterioles is a key feature in these PH models (5659). Of note, although chronic exposure to Hx is widely used as an animal model of PH, Hx may induce different genes in the lungs of rats versus mice (60). The sugen 5416/hypoxia (Su5416/Hx) rat model of PH more closely recapitulates the human pathology due to its progressive nature, formation of plexiform lesions, and ultimate lethality (6164). Sugen 5416 (semaxanib) is an antineoplastic drug with tyrosine kinase inhibitor and vascular endothelial growth factor receptor (VEGFR)-2 inhibitor properties (65). Su5416/Hx in rats and mice sets off a cascade of events that lead to structural remodeling of the pulmonary arterioles and progressively worsening hemodynamics and plexiform lesions that are not seen in any other model of PH (66, 67). As such, this model has been very useful to study the pathogenetic mechanisms of PH and identify novel therapeutic targets.

INCREASED SYMPATHETIC TONE AND NEUROVASCULAR MEDIATORS IN PH

Neurohormonal imbalance is observed in many cardiovascular disorders and has been associated with worse prognosis and survival in patients with PH. Increased neurovascular release of noradrenaline is expected to increase pulmonary vasoconstriction. However, we have shown that phenylephrine was less potent in producing vasoconstriction in pulmonary arteries of Su5416/Hx versus control rats (68), suggesting decreased sensitivity of the α-adrenergic receptors. This could be partly explained by chronic increase in sympathetic discharge from the nerve endings leading to desensitization of the α-adrenergic receptors, making it important to further examine pulmonary sympathetic activity in PH.

Neuropeptides such as calcitonin gene-related peptide (CGRP) and substance P are released from sensory C fiber nerve endings in pulmonary microvessels likely through activation of transient receptor potential vanilloid type-1 channel (TRPV1) (69). CGRP is a vasodilator (via cAMP) and substance P is a vasoconstrictor or vasodilator depending on which cell it acts on, but it also promotes vascular smooth muscle cell (VSMC) proliferation.

Dysregulation of the renin-angiotensin-aldosterone system (RAS) has also been implicated as a causative factor in PH. Angiotensin II (ANG II) can exert deleterious effects on the pulmonary vasculature by promoting vasoconstriction, VSMC proliferation, and vascular inflammation (70). Therapies targeting RAS, such as angiotensin converting enzyme (ACE) inhibitors or angiotensin receptor type 1 (AT1R) blockers, may be useful in PH, but the likelihood of these RAS modulators causing systemic hypotension is a serious consideration. Other studies have shown that activation of angiotensin converting enzyme 2 (ACE2), which converts ANG II to Ang-(1–7), can also induce beneficial outcomes in PH through vasodilation and inhibition of inflammation (7174).

Transcatheter pulmonary artery denervation by applying circumferential radiofrequency ameliorated mPAP and PVR in some patients with left heart failure with reduced ejection fraction and PH (75). Although pulmonary artery denervation could reduce the sympathetic activity in the pulmonary circulation, the sympathetic system is crucial to facilitate bronchodilation and oxygen uptake, making it important to further evaluate the benefits of this approach in large-scale multicenter randomized controlled clinical trials. In effect, some studies suggest potential benefits of parasympathetic stimulation in PH, and show that oral cholinesterase inhibitor donepezil (Aricept), a drug used to treat Alzheimer’s disease, enhances parasympathetic activity, and reduces mPAP, RVSP, and pulmonary arterial remodeling in MCT-treated rat model (76). Although this study did not address potential detrimental effects of parasympathetic activation such as bronchoconstriction, these effects will need to be considered in the evaluation of translatability to clinical application.

ENDOTHELIAL DYSFUNCTION AND ENDOTHELIUM-DERIVED VASOACTIVE MEDIATORS IN PH

PH is associated with dysfunction of different cells of the pulmonary vascular wall, including endothelial cells. Maintenance of pulmonary vascular homeostasis involves complex endocrine, autocrine, and paracrine interactions in the vascular milieu, and the endothelium is a major determinant of vascular homeostasis. The intact endothelium has thrombo-resistant and anti-inflammatory properties, and its barrier function prevents extravasation of fluid and macromolecules and penetration of inflammatory cells such as neutrophils and monocytes into the vessel wall. In response to a variety of injurious stimuli such as Hx, shear stress, or inflammatory cytokines, the endothelium assumes an activated state, loses its barrier function and thrombo-resistant properties, and releases growth factors and adhesion molecules that contribute to vascular wall remodeling, in situ thrombosis, and PH pathology. Endothelial cell dysfunction has been described in both clinical PH and preclinical animal models especially in the context of dysfunctional BMPR2 signaling (77) with resultant disruption of PPARγ and p53 transcriptional regulation of downstream genes involved in endothelial cell survival and angiogenesis (7880). Also, endothelial cell deficiency in the iron-sulfur biogenesis protein frataxin could decrease iron-sulfur clusters and lead to endothelial cell senescence and PH (81). The endothelium is known to release endothelium-derived relaxing factors such as nitric oxide (NO), prostacyclin (PGI2), and endothelium-derived hyperpolarizing factor as well as vasoconstricting factors such as endothelin-1 (ET-1) and thromboxane A2 (TXA2). Endothelial cell dysfunction could lead to imbalance between vasodilator and vasoconstrictor factors and increased pulmonary vasoconstriction and PVR.

Nitric Oxide

NO is a major effector of endothelium-dependent vasodilation (Fig. 2). NO is produced by the enzymatic action of endothelial NO synthase (eNOS or NOS2) on l-arginine and diffuses to the adjacent VSMCs where it activates soluble guanylate cyclase that generates cyclic GMP (cGMP). cGMP activates protein kinase G that induces VSMC relaxation by promoting Ca2+ extrusion and reducing Ca2+ sensitivity of the contractile proteins. Phosphodiesterase-5 (PDE5) breaks down and terminates the vasodilator effects of cGMP. Downregulation of the NO-cGMP relaxation pathway is an important factor in the increased pulmonary vasoconstriction in PH. Expression of endothelial NOS is reduced in patients with PAH (82). Acetylcholine (ACh)-induced relaxation is reduced in isolated lung preparation of Hx rats (83). Also, ACh-induced relaxation and the NO-cGMP pathway are reduced in pulmonary arteries of the MCT, Hx, and Su5416/Hx rat models (68, 84). ACh-induced NO production is reduced in the Su5416/Hx rat model (68), likely due to decreased expression/activity of NOS. The reduced ACh-induced relaxation could involve decreased NO bioavailability due to increased reactive oxygen species during chronic Hx-induced vascular remodeling and PH (85). The reduced ACh relaxation in PH models could also be due to structural changes in the pulmonary vascular wall that decrease the responsiveness of PASMCs to vasodilators. This is supported by our observation that pulmonary artery relaxation to the exogenous NO donor sodium nitroprusside is reduced in the MCT, Hx, and Su5416/Hx rats (68, 84). This is also consistent with reports that both endothelium-dependent and -independent relaxation are reduced in the MCT-induced rat model of progressive lung injury (86). Some studies found that endothelium-independent vasodilation by sodium nitroprusside was fully active in the Hx rat model (83), and the different findings could be related to the vascular preparation used (pulmonary artery vs. isolated perfused lung) or other factors affecting the responsiveness to the exogenous NO donors, including changes in PDE5 or protein kinase G activity.

Figure 2.

Figure 2.

Nitric oxide-cyclic guanosine monophosphate (NO-cGMP) pulmonary arterial relaxation pathway. l-Arginine is taken up by pulmonary arterial endothelial cells to be converted by endothelial NO synthase (eNOS) to NO and l-citrulline. NO diffuses into pulmonary arterial smooth muscle cells (PASMCs), where it activates soluble guanylate cyclase that converts GTP into cGMP. cGMP activates cGMP-dependent protein kinase or protein kinase G (PKG) leading to decreased [Ca2+]cyt, inhibition of myosin light chain kinase (MLCK) activity, and PASMC relaxation. The NO-cGMP pathway could be enhanced by l-arginine supplementation, NOS induction by gene therapy, NO inhalation, NO donors, or guanylate cyclase activators. The NO-cGMP pathway could also be augmented by inhibiting phosphodiesterase-5 (PDE5) and preventing the conversion of cGMP to GMP, or by inhibiting arginase and preventing the conversion of l-arginine to ornithine and urea. cGMP, cyclic guanosine monophosphate; PDE5i, PDE5 inhibitor.

Considering the important role of NO-cGMP in pulmonary vascular relaxation, many of the current therapies involve modulation of this pathway (Table 3). NO inhalation ameliorates PH and improves organ functions in children undergoing corrective surgeries for congenital heart disease and in adults after heart valve surgeries (87). The PDE5i sildenafil may facilitate weaning from inhaled NO in children with PAH following surgery for congenital heart disease (7). Combined use of PDE5i and nitrates has also been considered in ameliorating PAH in patients with heart failure (8). A meta-analysis of clinical trials has shown that sildenafil administration by nasogastric feeding can improve PPHN (88). Sildenafil treatment also improves MCT-induced PH in rats (53). Enhancing the NO-cGMP pathway using l-arginine supplementation, the NO donor molsidomine or PDE5i sildenafil also ameliorated right ventricle hypertrophy and myocardial fibrosis and apoptosis in Hx rat model of PH (89). Antioxidants and modulators of mitochondrial oxidative phosphorylation pathways reduce reactive oxygen species and could increase NO bioavailability and promote vasodilation. In support, dichloroacetate (DCA), an inhibitor of the mitochondrial pyruvate dehydrogenase kinase, the gatekeeping enzyme of glucose oxidation, has been shown to improve hemodynamics and functional capacity in a subset of patients with PAH. Of note, other patients who had inactivating mutations of two genes encoding mitochondrial enzymes (SIRT3 and UCP2) were not responsive to DCA, suggesting a relationship between mitochondrial pyruvate dehydrogenase kinase activity and genetic susceptibility to PAH (90).

Table 3.

Endothelial-derived vasoactive mediators, signaling pathways, and related medications for PH

Mediator Receptor/Target in Smooth Muscle Postreceptor Pathways Related Approved Medications
Vasodilators
 NO Guanylate cyclase cGMP, PKG
-PMCA, ↑Ca2+ extrusion
-Phosphorylation of MLCK, ↓MLCK activity, ↓ Ca2+ sensitivity
-Inhaled NO in acute PH
-Guanylate cyclase stimulators, Riociguat
-PDE5i, Sildenafil, Tadalafil, Vardenafil
 PGI2 IP receptor cAMP, PKA
-PMCA, ↑Ca2+ extrusion
-Phosphorylation of MLCK, ↓MLCK activity, ↓Ca2+ sensitivity
IP receptor agonists,
Epoprostenol, Treprostinil, Beraprost, Iloprost, Selexipag
Vasoconstrictors
 ET-1 ETAR -Ca2+ release from SR
-Ca2+ influx via channels
-Ca2+/CAM, activates MLCK
-Rho-A/ROCK pathway, PKC pathway, inhibit MLC phosphatase
-Nonselective ETR antagonists, Bosentan, Macitentan
-Selective ETAR antagonists. Ambrisentan
 TXA2 TP receptor -Ca2+ release from SR
-Ca2+ influx via channels
-Ca2+/CAM, activates MLCK
-Rho-A/ROCK pathway, PKC pathway, inhibit MLC phosphatase
TXA2 antagonists
Need further evaluation clinically
-Daltroban, Domitroban, Sulotroban, Terutroban
-Vapiprost, Seratrodast

CAM, calmodulin; cAMP, cyclic adenosine monophosphate; cGMP, cyclic guanosine monophosphate; ET-1, endothelin-1; ETAR, endothelin receptor type A; IP, prostacyclin receptor; MLC, myosin light chain; MLCK, MLC kinase; NO, nitric oxide; PDE5, phosphodiesterase-5; PH, pulmonary hypertension; PKA, protein kinase A; PKC, protein kinase C; PKG, protein kinase G; PMCA, plasma membrane Ca2+-ATPase, ROCK, Rho-associated protein kinase; SR, sarcoplasmic reticulum; TXA2, thromboxane A2.

Other approaches that could enhance the NO-cGMP pathway have been studied. Oral 1-nitro-2-phenylethane and trans-4-methoxy-β-nitrostyrene are novel soluble guanylate cyclase stimulators that improve mPAP, pulmonary hemodynamics, and pulmonary vascular function and remodeling, and reduce RVSP and right ventricular hypertrophy in MCT rat model (91, 92). We have shown that ACh relaxation and NO production are improved in pulmonary arteries of Su5416/Hx rats by inducing mild acidification using the carbonic anhydrase inhibitor acetazolamide (ACTZ) (93). The ACTZ beneficial effects could be related to improved NOS activity, increased NO bioavailability (94), or improved PASMC responsiveness to NO and other vasodilators (93).

Prostacyclin

Prostacyclin (PGI2) is a member of the prostanoid family of eicosanoids derived from arachidonic acid by the sequential actions of phospholipase A2, cyclooxygenase (COX), and prostaglandin (PG) synthases in endothelial cells. Endothelial-derived PGI2 binds to the prostacyclin receptor (IP) in VSMCs (95) and stimulates a G protein-coupled increase in cAMP that activates protein kinase A, resulting in decreased [Ca2+]cyt and enhanced vasorelaxation. PGI2 may also reduce VSMC contraction and PVR by inhibiting Rho-kinase. PGI2 through intracrine signaling may also target nuclear peroxisome proliferator-activated receptor (PPAR) and in turn regulate gene transcription, VSMC proliferation, and pulmonary vascular remodeling (96, 97).

Studies in patients with PH and preclinical animal models have identified deficiencies in different components of the PGI2 pathway, and demonstrated therapeutic benefits of enhancing PGI2 signaling, and consequently led to clinical use of PGI2 analogs in PH (97104). Patients with severe PAH have reduced IP receptor expression in their remodeled PASMCs (97). Studies in Hx mouse model have shown that IP receptor KO mice develop more severe PH and vascular remodeling compared with WT mice, and suggested that modulation of PGI2 synthesis and IP receptor activity could reduce pulmonary vascular remodeling and PH (97). In support, overexpression of pulmonary PGI2 synthase in transgenic mice protects against the development of Hx-induced PH (99).

Different parenteral, inhalation (epoprostenol), and oral prostanoids are currently available as PH therapies. Oral IP receptor agonist selexipag reduces disease progression and morbidity/mortality in patients with PAH (105). Treprostinil palmitil is a long-acting inhaled pulmonary vasodilator prodrug of treprostinil. Early initiation and higher doses of intraperitoneal (IV) or subcutaneous (SC) prostanoids and combination with other PAH-targeted therapy showed favorable outcome, and transition from parenteral to oral or inhaled prostanoid therapies was safe on the long-term in children with PAH (106). Inhaled iloprost may also be better than inhaled NO in offering an effective, safe, and pulmonary-selective vasodilation strategy for the treatment of group 2 patients with PH and left heart disease (9).

Endothelium-Dependent Hyperpolarization

Endothelium-dependent hyperpolarization involves activation of K+ channels in vascular cells (101, 107). In pulmonary artery endothelial cells, K+ channel activity controls resting membrane potential, Ca2+ entry, and the release of different vasoactive factors. Transient receptor potential vanilloid-4 (TRPV4) channels activity in endothelial cell caveolae plays an important role in maintaining normal mPAP. Dysregulation of K+ channel expression/activity by Hx or other vasoactive factors could contribute to the pathogenesis of PH. Pulmonary arteries from patients with PH and mouse models show upregulation of iNOS and NOX1 enzymes and increased peroxynitrite in endothelial cell caveolae that could affect caveolin-1 and reduce TRPV4 channel activity in PH (108).

Endothelium-dependent hyperpolarization spreads through myoendothelial junctions to underlying VSMCs. In PASMCs, K+ channels determine membrane potential, with increased activity leading to membrane hyperpolarization and pulmonary vasodilation whereas decreased activity leads to membrane depolarization, Ca2+ entry, and pulmonary vasoconstriction. K+ channel activity could also affect PASMC phenotype, proliferation, and survival/apoptosis.

Enhancing K+ channel expression/activity could be an attractive strategy to reduce PVR in patients with PH (109). Many of the existing vasodilator treatments indirectly increase K+ channel activity (110112), and direct modulators of channel K+ channel expression/activity have also been proposed as potential therapeutic approaches in PH. For instance, genetic approaches such as gene therapy or administration of antago-miRs have been considered to increase K+ channel expression in PH (113, 114). Selective and nonselective K+ channel activators have also shown beneficial effects ex vivo (115, 116) and in preclinical models (117, 118). In addition, nutritional factors, antioxidants, and other compounds have shown K+ channel activating effects that could be applied therapeutically (119122). Further understanding of the diversity of K+ channel types and the complexity of mechanisms by which their dysregulation contributes to PH pathogenesis as well as consideration of potential off-target effects of these K+ channel modulators are needed before clinical translation.

Hydrogen Sulfide

Hydrogen sulfide (H2S) is a major transmitter with regulatory effects in the cardiovascular system and the pulmonary circulation. H2S is involved in pulmonary vascular cell homeostasis and could counterbalance the Hx response and the disruption in mitochondrial bioenergetics induced during PH development. For instance, H2S modulates ATP-sensitive K+ channel (KATP) activity and in turn promotes pulmonary artery relaxation. H2S also reduces oxidative stress, the inflammatory response, and pulmonary artery remodeling. Alteration in H2S biogenesis could contribute to the pathogenesis of PH. H2S may promote protective effects against PH, and H2S-releasing agents could be a new therapeutic strategy in PH (123).

Endothelin-1

ET-1 is a potent endothelium-derived vasoconstrictor and mitogen for VSMCs. ET-1 binds to G protein-coupled endothelin receptor type A and type B on the adjacent VSMCs causing increases in [Ca2+]cyt, myosin light chain (MLC) phosphorylation, and vasoconstriction. ET-1 also activates Rho-Kinase and protein kinase C leading to inhibition of MLC phosphatase, increased myofilament sensitivity to Ca2+ and further vasoconstriction (124126).

Studies have shown elevated ET-1 levels in patients with PAH (127) and animal models of PH (128) and suggested a causal relationship with disease pathogenesis, and this led to the development of endothelin receptor antagonists as therapeutic agents for PAH (129132). Bosentan, macitentan, and aprocitentan are dual ET-1 receptor antagonists to both endothelin receptor type A (ETAR) and endothelin receptor type B (ETBR). Importantly, ETBRs are also expressed in endothelial cells and mediate endothelin clearance and NO and PGI2 release. ETBR-deficient mice show exaggerated response to Hx and PH (133). To avoid interference with the beneficial vasodilator endothelial ETBRs, selective ETAR blockers have been developed. ETAR blockade partially reverses neonatal hypoxic pulmonary vascular remodeling (134) and reduces contraction in pulmonary arteries from Hx rats (128). However, clinically, selective ETAR antagonist has not been proven to be superior to nonselective ET antagonists in ameliorating PAH (135).

Analysis of blood samples from the pulmonary artery and systemic vessels of children with PAH showed a wide range of plasma levels of ET-1 and big ET-1, suggesting possible changes in the net release or lung clearance of ET-1. Also, in neonates with PPHN, we found increased levels of circulating ET-1 that decreased in response to treatment with inhaled NO (136). However, in other studies, ET-1 and big ET-1 levels showed no correlation with the clinical and hemodynamic parameters or the severity of PAH and were not able to predict outcome (137), supporting the involvement of other factors in the disease process.

Thromboxane A2

Increased TXA2 activity may play a role in the pathogenesis of cardiopulmonary disorders including atherosclerosis, myocardial infarction, stroke, bronchial asthma, and PH. Studies have empirically determined a dose of the TXA2 analog U46619 that could induce PH in pigs without causing systemic hemodynamic disturbances, making it an attractive model to evaluate the efficiency of different therapeutic approaches of PH in large animals (138). Several TXA2 antagonists have been developed, but their effectiveness and safety need to be further evaluated clinically in PH.

Heme Oxygenase-1

Heme oxygenase-1 (HO-1) is a rate-limiting enzyme responsible for degradation of heme to biliverdin, free iron, and carbon monoxide. VSMCs through increased HO-1 activity could release carbon monoxide that exerts paracrine effects on the adjacent endothelial cells. Carbon monoxide is a potent vasodilator. HO-1 via its products inhibits oxidative stress and inflammation (139). HO-1 deficiency results in endothelial damage with elevation of thrombomodulin and von Willebrand factor (140).

Stimuli that cause oxidative stress such as peroxynitrite and Hx modulate HO-1 expression (141). HO-1 enzymatic products have shown beneficial cardiopulmonary actions in chronic Hx (142). Also, enhancement of endogenous HO-1 activity improved the hemodynamics and mPAP in Hx rat model (143).

IMPAIRED VSMC CONTRACTION PATHWAYS IN PH

Changes in VSMC contraction mechanisms could also contribute to increased pulmonary vascular tone and PVR. Studies have shown enhanced ET-1-induced pulmonary vasoconstriction in the Fawn-Hooded rat model of spontaneous PAH (144). Also, pulmonary vessels are hyperresponsive in perfused lungs from MCT-treated rats (145), and pulmonary vasomotor tone is increased in Hx rat model (146, 147). Chronic Hx is thought to increase the production/activity of ET-1 and ANG II in the lung leading to increased vasoconstriction (146). On the contrary, we have found that the contraction of pulmonary artery, but not the aorta and systemic mesenteric vessels, to phenylephrine and high KCl depolarizing solution is reduced in the MCT, Hx, and Su5416/Hx rat models (68, 84), indicating specific changes in the pulmonary circulation. The reduced pulmonary artery contraction in MCT and Hx rats may not be due to decreased sensitivity/number of α-adrenergic receptors because phenylephrine was equally potent in control, MCT, and Hx rats, and the contraction to high KCl, a receptor-independent response, was also reduced. Other studies have shown that ET-1-induced pulmonary artery contraction is also reduced in the Hx rat model (128), suggesting a reduction in a common postreceptor contraction pathway or a switch in PASMCs from a contractile to synthetic phenotype. The differences in the pulmonary vascular contractile response in different studies could be due to differences in the animal model, the vasoconstrictor studied (phenylephrine and KCl vs. ET-1 and ANG II) or the vascular preparation used (pulmonary artery versus isolated perfused lung), making it important to further examine the molecular mechanisms of VSMC contraction and their potential changes in PH.

Ca2+-Dependent Contraction Pathways in PH

Ca2+ is a major determinant of VSMC contraction (Fig. 3). In PASMCs, vasoconstrictor agonists such as ET-1 and TXA2 activate G protein-coupled receptor (GPCR) leading to activation of plasma membrane phospholipase C and increased generation of inositol trisphosphate (IP3). IP3 releases Ca2+ from the sarcoplasmic reticulum (SR). Vasoconstrictor agonists also activate Ca2+ influx though plasma membrane Ca2+ channels. Increased [Ca2+]cyt binds calmodulin and activates MLC kinase (MLCK) leading to increased MLC phosphorylation, actin-myosin interaction, and VSMC contraction. Hx could alter the synthesis of or responsiveness to vasoactive factors and in turn causing changes in PASMC electrophysiological properties and signaling pathways (148). Hx could also affect PASMC function by modulating the receptor population, ion channel activity, K+ current, and membrane depolarization leading to elevation of [Ca2+]cyt and increased vascular contraction, PVR, and mPAP (146). Ca2+-dependent activation of PI3K/AKT/mTOR pathway could also promote gene expression, PASMC proliferation, and pulmonary artery remodeling (149, 150).

Figure 3.

Figure 3.

Interaction between pulmonary arterial smooth muscle cell (PASMC) contraction and relaxation pathways. In PASMCs, vasoconstrictors such as endothelin-1 (ET-1) and thromboxane A2 (TXA2) activate their corresponding surface membrane G protein-coupled receptor (GPCR) and stimulate the GTP-binding protein Gq-11 leading to activation of phospholipase C (PLC) and increased generation of inositol-trisphosphate (IP3) and diacylglycerol (DAG). IP3 causes Ca2+ release from the sarcoplasmic reticulum (SR), whereas activation of surface membrane Ca2+ channels leads to Ca2+ influx and increased [Ca2+]cyt. Ca2+ binds calmodulin to activate myosin light chain kinase (MLCK), leading to MLC phosphorylation, increased myosin ATPase activity, actin-myosin interaction, and Ca2+-dependent contraction. DAG with or without Ca2+ activates different protein kinase C (PKC) isoforms. Activation of GPCRs could also stimulate the GTP-binding protein G12-13 and guanine nucleotide exchange factor (GEF) leading to stimulation of Rho-A that activates Rho-kinase (ROCK). PKC and ROCK directly or indirectly through phosphorylation of PKC-potentiated phosphatase inhibitor protein-17 (CPI-17) leading to inhibition of MLC phosphatase, thus augmenting MLC phosphorylation and Ca2+ sensitization of the contractile proteins. Prostacyclin (PGI2) is a pulmonary vasodilator acting through prostacyclin receptor (IP) to stimulate adenylate cyclase (AC) and convert ATP to cAMP, which activates protein kinase A (PKA). Also, nitric oxide (NO) diffuses into PASMCs to activate guanylate cyclase (GC) and convert GTP to cGMP, which activates protein kinase G (PKG). Both PKA and PKG stimulate Ca2+ extrusion and decrease [Ca2+]cyt, leading to activation of MLC phosphatase, MLC dephosphorylation, and PASMC relaxation. PKA and PKG could also phosphorylate MLCK and decrease its activity, further enhancing PASMC relaxation. PKA may also inhibit ROCK and prevent its Ca2+ sensitization mechanisms. Interrupted lines indicate inhibition.

Transient receptor potential channels (TRPCs) are stretch-activated nonselective Ca2+-permeable cation channels. TRPC6 channels are upregulated in PASMCs from patients with PAH. Also, 2-aminoethyl diphenyl borniate (2-APB), a nonselective cation channel blocker, and BI-749237, a selective TRPC6 blocker, inhibited acute Hx-induced pulmonary vasoconstriction, and intraperitoneal injection of 2-APB ameliorated PH in animal model, suggesting TRPC6 channel modulation as a novel therapeutic target in PH (151).

Piezo channel superfamily is also a mechanosensitive cation channel responsible for stretch-mediated Ca2+ and Na+ influx in multiple cell types. Upregulation of Piezo1 has been associated with increased [Ca2+]cyt in PASMCs from patients with idiopathic PAH (152). Interestingly, Piezo1 is upregulated in pulmonary arterial endothelial cells from patients with idiopathic PAH, and Piezo1 mRNA expression and protein levels are increased in the pulmonary artery and PASMCs from animal models of severe PH, supporting a role of mechanosensitive stretch-activated channels and Ca2+ influx in the increased vasoconstriction and pulmonary vascular remodeling in PH (153).

Hx may also promote polymerization and activation of the extracellular Ca2+-sensing receptor, and a peptide blocking the receptor polymerization was found to reduce its Hx-induced activation and downstream pathways leading to PH (154).

Rho-Kinase Pathway in PH

Vasoconstrictor agonists could activate the RhoA/Rho-associated protein kinase (ROCK) pathway and increase the sensitivity of the contractile apparatus to [Ca2+]cyt (155). ROCK2 levels are greater in PASMCs from patients with idiopathic PAH than normal PASMCs (156). Enhanced ROCK activity and Ca2+ sensitization of the contractile proteins has also been demonstrated in preclinical PH models. In Fawn-Hooded rat model of spontaneous PAH, ET-1 promotes vasoconstriction partly by increasing ROCK activity (144). Hx could increase pulmonary vasoconstriction by enhancing ROCK activity in PASMCs. Both ET-1 and serotonin could be involved in enhancing RhoA/ROCK signaling in the pulmonary circulation of Hx rat model (157). Studies have also suggested an important role of ROCK-mediated vasoconstriction in rat models of severe occlusive PAH (147). MCT-treated PH rats also show increased ROCK2 expression in PASMCs (156).

ROCK inhibitors such as fasudil may have the potential to reduce vasoconstriction and vascular remodeling in PH (Table 4). ROCK2 inhibition using specific ROCK2 inhibitor KD025 or its knockdown using siRNA reduced the exaggerated proliferation of PASMCs from patients with PAH. KD025 also attenuated the elevated RVSP in MCT rats, supporting a role of ROCK2 in the pathogenesis of PAH (156). Based on the therapeutic potential of fasudil (ROCK inhibition) and dichloroacetate (inhibition of mitochondrial pyruvate dehydrogenase kinase and oxidative stress) on PAH, studies have explored the effects of fasudil dichloroacetate and found that it attenuated Su5416/Hx-induced PAH, and reduced RVSP, pulmonary artery wall thickness, pulmonary vessel muscularization, perivascular fibrosis, and right ventricular hypertrophy and fibrosis. In PASMCs, fasudil dichloroacetate inhibited Hx-induced proliferation, migration, and contraction to a greater extent than fasudil or dichloroacetate alone, and reduced [Ca2+]cyt, Ca2+/calmodulin-dependent MLC phosphorylation and Rho-kinase activity, and restored mitochondrial function (158).

Table 4.

VSMC contraction mechanisms and potential therapies for PH

Mechanism Sources/Stimulators Target Potential Therapies for PH
[Ca2+]cyt Ca2+ release from SR
Ca2+ entry through channels
Ca2+/calmodulin, MLCK, MLC phosphorylation Ca2+ channel blockers,
Nifedipine
ROCK Rho-A Phosphorylates CPI-17, inhibits MLC phosphatase, ↑ Ca2+ sensitivity ROCK inhibitors, Fasudil
ROCK2 inhibitors, KD025
PKC Diacylglycerol, Ca2+ Phosphorylates CPI-17, inhibits MLC phosphatase, ↑ Ca2+ sensitivity PKC inhibitors, GF 109203X (Bisindolylmaleimide)

CPI-17, protein kinase C-potentiated phosphatase inhibitor protein-17; MLC, myosin light chain; MLCK, MLC kinase; PH, pulmonary hypertension; PKC, protein kinase C; ROCK, Rho-associated protein kinase; SR, sarcoplasmic reticulum.

Protein Kinase C Signaling in PH

Vasoconstrictor agonists also increase diacylglycerol production and activate protein kinase C (PKC), which in turn enhances the Ca2+ sensitivity of the contractile proteins (159). PKC activation may be involved in the increased vasoconstriction and vascular remodeling in PH. In Fawn-Hooded rat model of spontaneous PAH, ET-1-induced vasoconstriction could partly involve PKC (144). In cultured mouse PASMCs, exposure to Hx promoted cell proliferation and caused upregulation of PKCδ and PKCε expression and increased phosphorylation of AKT and extracellular signal-regulated kinase (ERK), suggesting a role of novel PKCs and downstream AKT and ERK signaling in Hx-induced PASMC proliferation, and offering new target for molecular therapy of PH (160).

COMBINATION VASODILATOR TREATMENT STRATEGIES IN PH

Because of the complexity of PH pathobiology, a widely adopted treatment strategy is to use a combination vasodilator therapy rather than a monotherapy approach. Much of the current treatment strategies has centered on initial combination therapy with PDE5i tadalafil and ETR antagonist ambrisentan. Sildenafil/ambrisentan combination therapy is more effective than sildenafil monotherapy in improving mPAP in patients with PH (161). Oral PGI2 analog selexipag may be added as a triple oral combination therapy in case of insufficient response to oral combination therapy with PDE5i and ETR antagonist. However, more is not always better. In a multicenter, double-blind, randomized phase 3b study in newly diagnosed patients with PAH, both initial triple (tadalafil, macitentan, and selexipag) versus initial double (tadalafil, macitentan, and placebo) oral therapy markedly reduced PVR by week 26, with no significant difference between groups (162).

VSMC PHENOTYPIC SWITCHING AND MEDIAL THICKENING IN PH

Although increased vasoconstriction is recognized as a pathophysiological component of PH, only 10%–20% of patients have an adequate vasodilator response when examined by a standardized procedure (163, 164). This suggests that either vasoconstriction is of much lower magnitude than what can be detected by the vasodilator test, or that a fixed component in the pulmonary vascular wall plays a more prominent role in the pathogenesis of PH. One of the hallmarks of PAH is a progressive obliteration of small pulmonary arteries due to exaggerated PASMC proliferation and resistance to apoptosis. This opens new research avenues regarding changes in PASMCs from a contractile to proliferative phenotype and potential loss of the ability of dedifferentiated VSMCs to contract and relax (165). This may also lead to the evolution of PH therapy from vasodilators to antiproliferative drugs (166).

Studies have shown that PAH is associated with remodeling of the small pulmonary arteries, exaggerated vascular cell proliferation, and obliteration of the pulmonary microvasculature (15, 167, 168). We have shown that phenylephrine-induced contraction, and ACh- and sodium nitroprusside-induced relaxation are reduced in pulmonary arteries of MCT, Hx, and Su5416/Hx rat models, suggesting decreased VSMC contractility and reduced responsiveness to vasodilators in PH (68, 84). This could be related to extensive remodeling of the pulmonary arterioles and increased PASMC proliferation. Hx induces VSMC proliferation and migration and promotes synthetic PASMC phenotypic switching (169171). Loss of PASMC differentiation leads to excessive proliferation and migration and resistance to apoptosis, all of which lead to medial hypertrophy in the pulmonary arterioles. In Hx rat model, hypoxia-inducible factor-1α (HIF-1α) promotes the expression of Cx43, and the HIF-1α/Cx43 axis regulates the proliferation and migration of PASMCs (172). Augmented total and phosphorylated transcription factor CREB and reduced CREB phosphatases are associated with increased PASMC proliferation in MCT and Hx models (173). Increased release of growth factors such as VEGF, platelet-derived growth factor (PDGF), and TGFβ can activate the JAK/STAT pathway and further induce PASMC proliferation, migration, and transformation to synthetic phenotype. The “proliferative” or “synthetic” VSMCs do not contract because they lack the contractile protein myosin and other contractile markers. Cartilage oligomeric matrix protein (COMP) is believed to have protective effects in the cardiovascular system, and serum COMP levels are decreased in patients with PH, especially in the female subgroup, as well as in rat models of PH (174). We have shown increases in wall thickness of pulmonary arterioles of MCT, Hx, and Su5416/Hx models of PH (68, 84), supporting the contention that the decreased pulmonary artery responsiveness to vasoconstrictor and vasodilator stimuli is related to pulmonary vascular remodeling and PASMC switching from a contractile to synthetic/proliferative phenotype.

It is increasingly appreciated that alternative approaches such as antiproliferative, proapoptotic agents could hold promise in further improving the outcome of PH (5658, 175, 176). ANG II is a major promoter of VSMC proliferation. In a large retrospective cohort of patients with PH, the use of angiotensin-converting enzyme (ACE) inhibitors and ANG II receptor blockers was associated with improved survival. Also, the ANG II receptor blocker losartan reversed PH and right ventricular remodeling induced by chronic inhaled nicotine in rats and mice (177). Sotatercept is a fusion protein that binds activins and growth differentiation factors and could correct the imbalance between growth-promoting and growth-inhibiting pathways in PH (178). In preclinical models of PH, increased PASMC proliferation and pulmonary arterial stiffness are associated with activation of the Hippo pathway and concomitant increase in the expression of STAT-3, Yes-associated protein (YAP), and Notch-3 (179, 180). The prospect of interrupting this mechanobiological feedback loop to restore pulmonary vascular homeostasis offers promise as a novel strategy for treatment of PH.

In addition to their role in vascular relaxation, K+ channels are important in many aspects of VSMC homeostasis including the phenotypic switching from a contractile to a synthetic type seen in PH (181). K+ channels regulate key cellular processes such as VSMC differentiation, cell cycle control, proliferation, migration, and apoptosis through multiple mechanisms (182, 183). In general, upregulation of K+ channel activity leads to antiproliferative and proapoptotic effects, and these effects are often channel isoform-specific (111, 184). Given that different K+ channel types have been identified in VSMCs and that they regulate VSMC homeostasis through multiple mechanisms, careful consideration of the channel isoforms, expression levels, and stage of differentiation and activation is needed to optimally leverage this knowledge therapeutically.

We have shown that mild acidification by treating rats with NH4Cl (1.5%) in the drinking water improved pulmonary hemodynamics, reduced RVSP, right ventricular weight, and pulmonary arteriolar hypertrophic remodeling, and improved PASMC contraction and responsiveness to vasodilators in pulmonary arteries of MCT and Hx models (Fig. 4) (185). Extracellular acidosis inhibits proliferation and migration of cultured rat and mouse VSMCs (186, 187), likely by inhibiting their proliferative and migratory response to growth factors and by increasing their susceptibility to apoptosis (188). Carbonic anhydrases are ubiquitously expressed enzymes that catalyze the hydration of carbon dioxide to bicarbonate and induce mild acidosis. Ongoing studies are evaluating the therapeutic potential of acetazolamide (ACTZ) and other carbonic anhydrase inhibitors in cancer, obesity, allergy, and inflammation (189191). Interestingly, ACTZ shows antiproliferative properties in cancer cells (192). ACTZ also improves the pulmonary hemodynamics in Hx rat model (193). We have shown that treatment of Su5416/Hx rat model with ACTZ or NH4Cl induces metabolic acidosis, decreases RVSP and mPAP without affecting left ventricular systolic pressure, and reduces right ventricular hypertrophy (93, 194). Also, tissue histology of peripheral lung sections showed ameliorated pulmonary vascular remodeling and reduced arteriolar wall thickness in ACTZ-treated versus nontreated Su5416/Hx rats. Treatment of Su5416/Hx rats with ACTZ also improved pulmonary artery contraction and ACh- and sodium nitroprusside-induced relaxation, supporting that mild acidification improves PASMC contractility and responsiveness to vasodilators (93). In support, pulmonary arteries harvested from Su5416/Hx rats show a PASMC dedifferentiation pattern with decreased mRNA levels of the contractile markers Myocd (myocardin), Tagln (transgelin), Smtn (smoothelin), and Acta2 and increased proliferative marker Ccnd1 (cyclin D1), and treatment of Su6516/Hx rats with ACTZ partially restored the contractile markers and reduced the proliferative marker (194).

Figure 4.

Figure 4.

Potential benefits of mild acidification in pulmonary hypertension (PH). Mild acidification could affect pulmonary endothelial cells leading to increased release of vasodilators and improved relaxation, as well as pulmonary arterial smooth muscle cells (PASMCs) leading to restoration of contractile phenotype, improved response to vasodilators, decreased proliferation and migration. and reduced medial thickening. Mild acidification could also affect components of the extracellular matrix (ECM) leading to decreased fibrosis and hypertrophic remodeling, as well as inflammatory cells leading to decreased macrophage infiltration and reduced release of cytokines. These beneficial effects of mild acidification have improved the hemodynamics and reduced mean pulmonary arterial pressure (mPAP), right ventricular systolic pressure (RVSP), and right ventricular hypertrophy in rat models of PH. NO, nitric oxide; NOS, NO synthase.

PULMONARY ARTERIOLAR FIBROSIS AND REMODELING IN PH

The extracellular matrix (ECM) determines the structural architecture of the pulmonary vasculature and the myocardium and provides mechanical stability necessary for tissue homeostasis. PH is associated with pathological remodeling of the pulmonary vasculature and right ventricle, excessive deposition of structural and nonstructural proteins, and abnormal expression of TGFβ, fibroblast growth factor (FGF), MMPs, and other proteases in ECM. Aberrant TGFβ signaling is linked to PH. BMPR2 mutations perturb the balance between BMP and TGFβ pathways, leading to vascular remodeling and narrowing of the lumen of the pulmonary vasculature (195). Reduced BMP and BMPR2 have also been linked to reduced sphingosine-1-phosphate and zinc transporter ZIP12 signaling in MCT rat model of PH (196). Also, FGF receptors 1 and 2 (FGFR1/2) are elevated in patients with PH and in Hx mouse model (197).

Changes in the amount and content of ECM have been observed in patients with PH and animal models, supporting a role of ECM in disease progression. Accumulation of ECM proteins could also impact vascular cell biology, proliferation, and migration (198). Cardiac magnetic resonance imaging has shown increased pulmonary artery stiffness in PAH (199). Imbalance between MMPs and TIMPs could be an important pathobiological component of PH. MMP-8 is an interstitial collagenase involved in regulating fibrosis and inflammation in the lung and vasculature. MMP-8 expression is increased in plasma and pulmonary arteries of patients with PH compared with controls and in the Hx mouse model. MMP-8−/− mice show high mortality, increased RVSP, severe right ventricular dysfunction, and exaggerated vascular remodeling, compared with WT mice. MMP-8−/− PASMCs also show increased proliferation, suggesting protective role of MMP-8 by altering ECM composition in PH (200).

Serum levels of procollagen III NH2-terminal peptide, carboxyterminal propeptide of type I procollagen, MMP-2, and MMP-9 are decreased in patients with chronic thromboembolic PH compared with healthy subjects. Also, MMP-2-to-TIMP-1 ratio is lower in patients with PH than controls, and correlates negatively with PVR, suggesting that decreased MMP-2 or increased TIMP-1 could be an important factor in pulmonary vascular remodeling (201). Changes in the structure and function of elastin may also have an impact on the proximal pulmonary arterial mechanics in hypertensive calves (202).

Antifibrotics and other molecular and cellular approaches to reduce pulmonary artery remodeling could be useful in PH therapy. Emergent targeted approaches for PH include the development of a TGFβ ligand trap, and upregulation of BMPR2 (196). Angiotensin-converting enzyme 2 (ACE2) catalyzes the hydrolysis of the vasoconstrictor ANG II into the vasodilator Ang-(1–7), and is an important regulator of blood pressure and cardiovascular function. In MCT + Hx rat model of PH, the ACE2 activator diminazene aceturate reduced serum and perivascular focal adhesion kinase (FAK) and improved pulmonary arteriolar remodeling (203). Also, galectin-3 is a carbohydrate-binding lectin that affects reactive oxygen species production, NOX enzyme expression, inflammation, and fibrosis, and in turn contributes to pulmonary artery remodeling and PAH, and downregulating galectin-3 could be useful in PH (204).

Lineage tracing studies have provided important insights into the role of other cell types in the pulmonary vascular wall in the pathogenesis of PH including pericytes and VSM progenitors. Pericytes are mesenchymal-derived mural cells localized in the basement membrane of pulmonary capillaries. Besides structural support, pericytes regulate pulmonary vascular tone and produce ECM components and factors that control vascular homeostasis and angiogenesis. Pericytes may also produce profibrotic and proinflammatory cytokines and in turn contribute to vascular pathology. In the lung, loss of pericyte communication with alveolar capillaries and a switch to a profibrotic/proinflammatory phenotype would promote vascular remodeling, fibrosis, and inflammation in PH (205). Further studies to dissect the complex interactions of pericytes with other pulmonary and vascular cells would help to understand the mechanisms of PH and offer opportunities to develop novel therapeutics.

Resident VSM progenitors at the junction of the muscular to nonmuscular part of the pulmonary arterioles may contribute to pulmonary vascular remodeling in Hx-induced PH via clonal expansion mediated by the pluripotency transcription factor KLF4 (206). Studies in a chronic inflammation model of experimental PH showed that a subpopulation of VSMCs with high levels of Notch-3 expression is the major cellular origin of neointima formation (207).

In addition to resident cells, circulating vascular progenitors and stem cells including endothelial progenitor cells (EPCs), mesenchymal stromal cells (MSCs), and fibrocytes may contribute to vascular remodeling in PH (208). CXCR4 is a stem cell surface receptor of the cytokine CXCL12 that regulates homing of hematopoietic progenitor cells and their mobilization. Bone marrow-derived CXCR4 and proangiogenic cell accumulation could play a role in the development of PAH. CXCR4 expression is increased in pulmonary arteries of patients with PAH and Hx rat model. Downregulation of CXCR4 expression reduced β-catenin levels and PASMC proliferation, and ameliorated pulmonary vascular remodeling in Hx rats, suggesting that CXCL12/CXCR4 is an important factor in PAH (209).

ROLE OF INFLAMMATION IN PH

Irrespective of pathogenic origin, dysregulated immune and inflammatory responses underlie the pathobiology of PH. Human and preclinical studies have suggested a role of inflammation in PH pathogenesis (210215). A skewed immune response favoring a proinflammatory environment leads to infiltration of inflammatory cells such as lymphocytes, macrophages, and neutrophils. Elevated circulating levels of the proinflammatory cytokines tumor necrosis factor-α (TNFα) and interleukin-6 (IL-6) predicted poor outcomes in idiopathic and familial patients with PAH (213, 216). Also, PAH lungs show characteristic perivascular inflammatory infiltrates comprising lymphocytes, mast cells, dendritic cells, and macrophages (212, 217). Our group has shown induction of IL-6 and CCL2 and upregulation of IL-1β mRNA in lung specimens from human idiopathic PAH (194). Recruitment and activation of proinflammatory M1 and alternatively activated M2 macrophages have been demonstrated in the Hx mouse model and the Su5416 athymic rat model of PAH (214, 218). Studies in MCT rat model showed temporal change of macrophage M1/M2 polarization status in the development of PH, with CD68+NOS2+ M1-like macrophages participating in the initial stage of inflammation by accelerating apoptosis of endothelial cell, while CD68+CD206+ M2-like macrophages predominating in the reparative stage of inflammation and the following stage of vascular remodeling (219). The plasma levels of osteopontin, a pleiotropic protein involved in inflammation and fibrogenesis, are elevated and associated with PAH among patients with connective tissue diseases, and may serve as a biomarker for early diagnosis and intervention in PAH (220). TNFα may drive PAH by suppressing tBMPR2 and altering Notch signaling (221). Other studies suggested a role of IL-6/IL-21 signaling in the pathogenesis of PAH (222). IL-6, IL-13, and IL-11 activate the JAK/STAT pathway and induce pulmonary artery remodeling and fibrosis. We and others have shown increased alveolar macrophages and pulmonary expression of proinflammatory mediators Tnf, Il-6, and Ccl2 in Su5416/Hx model of PAH (194, 223). Of note, treatment of VSMCs with TNFα or IL-1β alters cell morphology, increases cell proliferation in response to 20% fetal bovine serum, and results in downregulation of the transcriptional coactivator myocardin (Myocd) and markers of contractile VSMC phenotype including Tagln (transgelin), Smtn (smoothelin), Acta2, Cnn1 (calponin 1), and Myh11 (myosin heavy chain 11), while upregulating markers of dedifferentiation such as Rbp1 (retinol-binding protein 1) and proliferation such as Ccnd1 (cyclin D1) (194, 221, 224, 225).

The identification of inflammation as a key pathogenetic factor in PH has directed research into specific therapies that could ameliorate the inflammatory process. Several proinflammatory pathways have been proposed as novel therapeutic targets for PH. Preclinical studies using anti-inflammatory approaches have shown promise. For instance, pharmacological targeting of TNFα or the IL-6 receptor ameliorates experimental PAH (221, 222). There are also compelling preclinical data to support the benefits of IL-6 blockade using tocilizumab, but the clinical evidence is not as robust. Blocking macrophage leukotriene B4 could prevent endothelial injury and ameliorates experimental PH (226). Immunomodulatory therapies have also been sought to improve the outcome of PAH (59, 175, 227).

Regulatory T cells (Tregs) are important contributors to autoimmunity, and their activity may be compromised during the inflammatory process in PAH. In support, athymic Treg-depleted rats treated with Su5416 develop PAH, which is prevented by infusing missing CD4+CD25highFOXP3+ Tregs (228), and these effects are exaggerated in female compared with male animals (229).

Immunosuppressive therapy may have some benefits in PH. However, clinical trials of immune modulators in PAH have yielded conflicting results (230), highlighting the need for other approaches. New avenues for targeting inflammation in PH include more targeted anti-inflammatory and proresolution strategies. Targeted expression of HO-1 has been shown to ameliorate the pulmonary inflammatory and vascular responses to Hx (215). In MCT model, BI113823, a selective kinin B1 receptor antagonist reverses experimental PH, decreases macrophage infiltration and TNFα and IL-1β cytokine production, and downregulates MMPs in the lungs (231). Resolvin E1 is a proresolving lipid mediator that could have protective effects in inflammatory diseases. Resolvin E1 levels are reduced in the plasma of patients with idiopathic PAH and in lungs from MCT rat model and Su5416/Hx mouse model of PH. Also, resolvin E1 treatment ameliorated experimental PH and reduced Hx-induced Wnt7a/β-catenin signaling and PASMC proliferation (232).

Mild acidosis could suppress inflammation through different metabolites such as lactic acid, ketone bodies, or protons (H+) (233235). Also, carbonic anhydrase inhibition has anti-inflammatory effects by inhibiting macrophage polarization (194). Treatment with ACTZ or NH4Cl reduces macrophage infiltration and expression of proinflammatory mediators in Su5416/Hx rat model, and inhibits cytokine-induced dedifferentiation in PASMCs (194).

Studies have shown increases in ACE2 and transmembrane serine protease-2 expression in lung tissue and a decrease in protective sACE2 in patients with chronic obstructive pulmonary disease (COPD), which may represent the possible risk factors for an increased susceptibility of patients with COPD to severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection (236). COPD and severe PH have been observed as major sequelae after severe postacute COVID-19 pneumonia (237). Also, histological analysis revealed that patients who died of COVID-19 exhibited thickened pulmonary vascular walls, an important feature of PAH (238). Pulmonary thrombosis is common in association with COVID-19, and the risk exists before and during hospitalization, and continues at least up to 4 wk after discharge (239), and its long-term effect on thromboembolic PH needs to be assessed. Interestingly, patients with COVID-19 infection may show a dysregulated acid-base status influenced by carbonic anhydrase activity, and ACTZ may be useful as a pharmacological approach for ameliorating COVID-19 symptoms (240).

CONCLUSIONS AND FUTURE DIRECTIONS

PH is characterized by various degrees of pulmonary vasoconstriction and progressive fibroproliferative obliteration of pulmonary arterioles, pulmonary vascular remodeling, fibrosis, and inflammation, leading to progressive increase in PVR and right ventricular hypertrophy and failure. An imbalance between pulmonary vasodilators including NO and PGI2 and vasoconstrictor factors such as ET-1 is recognized as an important factor in PH. Therefore, current therapies for PH are focused on vasodilators such as PDE5i, ETAR antagonists, PGI2 analogs, and Ca2+ channel blockers. To enhance their effectiveness, vasodilators are often used in combination therapy of PH. However, current vasodilator therapies are not universally successful in altering PH progression and increasing survival. Many patients do not respond adequately to vasodilators, largely due to a phenotypic switch in PASMCs from a contractile to a synthetic phenotype, with increased PASMC proliferation, excessive pulmonary vascular remodeling, fibrosis, and inflammation. Interventions to improve the responsiveness of the remodeled pulmonary arteries to vasodilators are emerging as novel strategies in PH. These novel strategies should target pulmonary artery remodeling, fibrosis, and inflammation using new approaches that restore the pulmonary vascular cell phenotype and reduce PASMC proliferation and ECM deposition. Ample preclinical evidence supports that antiproliferative, proapoptotic, immunomodulatory, and cell-based therapies could be effective in PH (5659, 175, 176, 227, 241); however, translation to clinical application is lagging behind experimental evidence. Induction of mild acidosis could reverse PASMC phenotypic switching, improve the pulmonary hemodynamics and vascular function, and reduce pulmonary artery remodeling, and therefore may provide a complimentary approach to enhance the effectiveness of vasodilator therapy in PH. ACTZ is a well-studied carbonic anhydrase inhibitor and inducer of mild acidosis with multiple clinical applications. Further studies using suitable experimental models of PH and additional methods of controlling and monitoring acidosis in vivo are needed to define the underlying mechanisms and translational potential of these new approaches.

GRANTS

This work was supported by Brigham Research Institute (BRI) Fund to Sustain Research Excellence from BRI (to R. A. Khalil) and National Heart, Lung, and Blood Institute Grants HL111775, R56HL147889, and R01HL147889-A1 (to R. A. Khalil) and R01HL116573 (to H. Christou).

DISCLOSURES

No conflicts of interest, financial or otherwise, are declared by the authors.

AUTHOR CONTRIBUTIONS

H.C. and R.A.K. conceived and designed research; H.C. interpreted results of experiments; H.C. and R.A.K. prepared figures; H.C. and R.A.K. drafted manuscript; H.C. and R.A.K. edited and revised manuscript; H.C. and R.A.K. approved final version of manuscript.

REFERENCES

  • 1.Simonneau G, Montani D, Celermajer DS, Denton CP, Gatzoulis MA, Krowka M, Williams PG, Souza R. Haemodynamic definitions and updated clinical classification of pulmonary hypertension. Eur Respir J 53: 1801913, 2019. doi: 10.1183/13993003.01913-2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Galie N, Humbert M, Vachiery JL, Gibbs S, Lang I, Torbicki A, Simonneau G, Peacock A, Vonk Noordegraaf A, Beghetti M, Ghofrani A, Gomez Sanchez MA, Hansmann G, Klepetko W, Lancellotti P, Matucci M, McDonagh T, Pierard LA, Trindade PT, Zompatori M, Hoeper M. 2015 ESC/ERS guidelines for the diagnosis and treatment of pulmonary hypertension: The Joint Task Force for the Diagnosis and Treatment of Pulmonary Hypertension of the European Society of Cardiology (ESC) and the European Respiratory Society (ERS): endorsed by: Association for European Paediatric and Congenital Cardiology (AEPC), International Society for Heart and Lung Transplantation (ISHLT). Eur Heart J 37: 67–119, 2016. doi: 10.1093/eurheartj/ehv317. [DOI] [PubMed] [Google Scholar]
  • 3.Philip JL, Murphy TM, Schreier DA, Stevens S, Tabima DM, Albrecht M, Frump AL, Hacker TA, Lahm T, Chesler NC. Pulmonary vascular mechanical consequences of ischemic heart failure and implications for right ventricular function. Am J Physiol Heart Circ Physiol 316: H1167–H1177, 2019. doi: 10.1152/ajpheart.00319.2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Omura J, Bonnet S, Kutty S. Right ventricular and pulmonary vascular changes in pulmonary hypertension associated with left heart disease. Am J Physiol Heart Circ Physiol 316: H1144–H1145, 2019. doi: 10.1152/ajpheart.00159.2019. [DOI] [PubMed] [Google Scholar]
  • 5.Fattouch K, Sbraga F, Bianco G, Speziale G, Gucciardo M, Sampognaro R, Ruvolo G. Inhaled prostacyclin, nitric oxide, and nitroprusside in pulmonary hypertension after mitral valve replacement. J Card Surg 20: 171–176, 2005. doi: 10.1111/j.0886-0440.2005.200383w.x. [DOI] [PubMed] [Google Scholar]
  • 6.Burger CD. Pulmonary hypertension in COPD: a review and consideration of the role of arterial vasodilators. COPD 6: 137–144, 2009. doi: 10.1080/15412550902754252. [DOI] [PubMed] [Google Scholar]
  • 7.Lee JE, Hillier SC, Knoderer CA. Use of sildenafil to facilitate weaning from inhaled nitric oxide in children with pulmonary hypertension following surgery for congenital heart disease. J Intensive Care Med 23: 329–334, 2008. doi: 10.1177/0885066608321389. [DOI] [PubMed] [Google Scholar]
  • 8.Stehlik J, Movsesian MA. Combined use of PDE5 inhibitors and nitrates in the treatment of pulmonary arterial hypertension in patients with heart failure. J Card Fail 15: 31–34, 2009. doi: 10.1016/j.cardfail.2008.09.005. [DOI] [PubMed] [Google Scholar]
  • 9.Yin N, Kaestle S, Yin J, Hentschel T, Pries AR, Kuppe H, Kuebler WM. Inhaled nitric oxide versus aerosolized iloprost for the treatment of pulmonary hypertension with left heart disease. Crit Care Med 37: 980–986, 2009. doi: 10.1097/CCM.0b013e3181962ce6. [DOI] [PubMed] [Google Scholar]
  • 10.McLaughlin VV, Benza RL, Rubin LJ, Channick RN, Voswinckel R, Tapson VF, Robbins IM, Olschewski H, Rubenfire M, Seeger W. Addition of inhaled treprostinil to oral therapy for pulmonary arterial hypertension: a randomized controlled clinical trial. J Am Coll Cardiol 55: 1915–1922, 2010. doi: 10.1016/j.jacc.2010.01.027. [DOI] [PubMed] [Google Scholar]
  • 11.Hoeper MM, Markevych I, Spiekerkoetter E, Welte T, Niedermeyer J. Goal-oriented treatment and combination therapy for pulmonary arterial hypertension. Eur Respir J 26: 858–863, 2005. doi: 10.1183/09031936.05.00075305. [DOI] [PubMed] [Google Scholar]
  • 12.Boucly A, Savale L, Jaïs X, Bauer F, Bergot E, Bertoletti L, et al. Association between initial treatment strategy and long-term survival in pulmonary arterial hypertension. Am J Respir Crit Care Med 204: 842–854, 2021. doi: 10.1164/rccm.202009-3698OC. [DOI] [PubMed] [Google Scholar]
  • 13.Gelzinis TA. Pulmonary hypertension in 2021. I. Definition, classification, pathophysiology, and presentation. J Cardiothorac Vasc Anesth. 2021 Jul 8:S1053-0770(21)00572-3. doi: 10.1053/j.jvca.2021.06.036. Epub ahead of print. [DOI] [PubMed] [Google Scholar]
  • 14.Walter K. Pulmonary hypertension. JAMA 326: 1116, 2021. doi: 10.1001/jama.2021.11054. [DOI] [PubMed] [Google Scholar]
  • 15.Farber HW, Loscalzo J. Pulmonary arterial hypertension. N Engl J Med 351: 1655–1665, 2004. doi: 10.1056/NEJMra035488. [DOI] [PubMed] [Google Scholar]
  • 16.Hoeper MM, Humbert M, Souza R, Idrees M, Kawut SM, Sliwa-Hahnle K, Jing ZC, Gibbs JS. A global view of pulmonary hypertension. Lancet Respir Med 4: 306–322, 2016. doi: 10.1016/S2213-2600(15)00543-3. [DOI] [PubMed] [Google Scholar]
  • 17.Kirson NY, Birnbaum HG, Ivanova JI, Waldman T, Joish V, Williamson T. Prevalence of pulmonary arterial hypertension and chronic thromboembolic pulmonary hypertension in the United States. Curr Med Res Opin 27: 1763–1768, 2011. doi: 10.1185/03007995.2011.604310. [DOI] [PubMed] [Google Scholar]
  • 18.Humbert M, Sitbon O, Chaouat A, Bertocchi M, Habib G, Gressin V, Yaïci A, Weitzenblum E, Cordier JF, Chabot F, Dromer C, Pison C, Reynaud-Gaubert M, Haloun A, Laurent M, Hachulla E, Cottin V, Degano B, Jaïs X, Montani D, Souza R, Simonneau G. Survival in patients with idiopathic, familial, and anorexigen-associated pulmonary arterial hypertension in the modern management era. Circulation 122: 156–163, 2010. doi: 10.1161/CIRCULATIONAHA.109.911818. [DOI] [PubMed] [Google Scholar]
  • 19.Benza RL, Miller DP, Gomberg-Maitland M, Frantz RP, Foreman AJ, Coffey CS, Frost A, Barst RJ, Badesch DB, Elliott CG, Liou TG, McGoon MD. Predicting survival in pulmonary arterial hypertension: insights from the Registry to Evaluate Early and Long-Term Pulmonary Arterial Hypertension Disease Management (REVEAL). Circulation 122: 164–172, 2010. doi: 10.1161/CIRCULATIONAHA.109.898122. [DOI] [PubMed] [Google Scholar]
  • 20.del Cerro Marín MJ, Sabaté Rotés A, Rodriguez Ogando A, Mendoza Soto A, Quero Jiménez M, Gavilán Camacho JL, Raposo Sonnenfeld I, Moya Bonora A, Albert Brotons DC, Moreno Galdó A. Assessing pulmonary hypertensive vascular disease in childhood. Data from the Spanish registry. Am J Respir Crit Care Med 190: 1421–1429, 2014. doi: 10.1164/rccm.201406-1052OC. [DOI] [PubMed] [Google Scholar]
  • 21.van Loon RL, Roofthooft MT, Hillege HL, ten Harkel AD, van Osch-Gevers M, Delhaas T, Kapusta L, Strengers JL, Rammeloo L, Clur SA, Mulder BJ, Berger RM. Pediatric pulmonary hypertension in the Netherlands: epidemiology and characterization during the period 1991 to 2005. Circulation 124: 1755–1764, 2011. doi: 10.1161/CIRCULATIONAHA.110.969584. [DOI] [PubMed] [Google Scholar]
  • 22.Li L, Jick S, Breitenstein S, Hernandez G, Michel A, Vizcaya D. Pulmonary arterial hypertension in the USA: an epidemiological study in a large insured pediatric population. Pulm Circ 7: 126–136, 2017. doi: 10.1086/690007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Maxwell BG, Nies MK, Ajuba-Iwuji CC, Coulson JD, Romer LH. Trends in hospitalization for pediatric pulmonary hypertension. Pediatrics 136: 241–250, 2015. doi: 10.1542/peds.2014-3834. [DOI] [PubMed] [Google Scholar]
  • 24.Arattu Thodika FMS, Nanjundappa M, Dassios T, Bell A, Greenough A. Pulmonary hypertension in infants with bronchopulmonary dysplasia: risk factors, mortality and duration of hospitalisation. J Perinat Med 50: 327–333, 2021. doi: 10.1515/jpm-2021-0366. [DOI] [PubMed] [Google Scholar]
  • 25.Tatius B, Wasityastuti W, Astarini FD, Nugrahaningsih DAA. Significance of BMPR2 mutations in pulmonary arterial hypertension. Respir Investig 59: 397–407, 2021. doi: 10.1016/j.resinv.2021.03.011. [DOI] [PubMed] [Google Scholar]
  • 26.West JD, Austin ED, Rizzi EM, Yan L, Tanjore H, Crabtree AL, Moore CS, Muthian G, Carrier EJ, Jacobson DA, Hamid R, Kendall PL, Majka S, Rathinasabapathy A. KCNK3 mutation causes altered immune function in pulmonary arterial hypertension patients and mouse models. Int J Mol Sci 22: 5014, 2021. doi: 10.3390/ijms22095014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Wu Y, Wharton J, Walters R, Vasilaki E, Aman J, Zhao L, Wilkins MR, Rhodes CJ. The pathophysiological role of novel pulmonary arterial hypertension gene SOX17. Eur Respir J 58: 2004172, 2021. doi: 10.1183/13993003.04172-2020. [DOI] [PubMed] [Google Scholar]
  • 28.Pienkos S, Gallego N, Condon DF, Cruz-Utrilla A, Ochoa N, Nevado J, Arias P, Agarwal S, Patel H, Chakraborty A, Lapunzina P, Escribano P, Tenorio-Castaño J, de Jesús Perez VA. Novel TNIP2 and TRAF2 variants are implicated in the pathogenesis of pulmonary arterial hypertension. Front Med (Lausanne) 8: 625763, 2021. doi: 10.3389/fmed.2021.625763. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Shinya Y, Hiraide T, Momoi M, Goto S, Suzuki H, Katsumata Y, Kurebayashi Y, Endo J, Sano M, Fukuda K, Kosaki K, Kataoka M. TNFRSF13B c.226G>A (p.Gly76Ser) as a novel causative mutation for pulmonary arterial hypertension. J Am Heart Assoc 10: e019245, 2021. doi: 10.1161/JAHA.120.019245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Hernandez-Gonzalez I, Tenorio-Castano J, Ochoa-Parra N, Gallego N, Pérez-Olivares C, Lago-Docampo M, Palomino Doza J, Valverde D, Lapunzina P, Escribano-Subias P. Novel genetic and molecular pathways in pulmonary arterial hypertension associated with connective tissue disease. Cells 10: 1488, 2021. doi: 10.3390/cells10061488. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.El-Khazragy N, El Barbary M, Fouad H, Abdelgawad A, Rabie D. Association between genetic variations in carbamoyl-phosphate synthetase gene and persistent neonatal pulmonary hypertension. Eur J Pediatr 180: 2831–2838, 2021. doi: 10.1007/s00431-021-04053-8. [DOI] [PubMed] [Google Scholar]
  • 32.Zhang Y, Zervopoulos SD, Boukouris AE, Lorenzana-Carrillo MA, Saleme B, Webster L, Liu Y, Haromy A, Tabatabaei Dakhili SA, Ussher JR, Sutendra G, Michelakis ED. SNPs for genes encoding the mitochondrial proteins Sirtuin3 and uncoupling protein 2 are associated with disease severity, type 2 diabetes, and outcomes in patients with pulmonary arterial hypertension and this is recapitulated in a new mouse model lacking both genes. J Am Heart Assoc 10: e020451, 2021. doi: 10.1161/JAHA.120.020451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Zhu N, Swietlik EM, Welch CL, Pauciulo MW, Hagen JJ, Zhou X, Guo Y, Karten J, Pandya D, Tilly T, Lutz KA, Martin JM, Treacy CM, Rosenzweig EB, Krishnan U, Coleman AW, Gonzaga-Jauregui C, Lawrie A, Trembath RC, Wilkins MR; Regeneron Genetics Center, PAH Biobank Enrolling Centers’ Investigators, NIHR BioResource for Translational Research - Rare Diseases, National Cohort Study of Idiopathic and Heritable PAH, Morrell NW, Shen Y, Gräf S, Nichols WC, Chung WK; Regeneron Genetics Center. Rare variant analysis of 4241 pulmonary arterial hypertension cases from an international consortium implicates FBLN2, PDGFD, and rare de novo variants in PAH. Genome Med 13: 80, 2021. [Erratum in Genome Med 13: 106, 2021] doi: 10.1186/s13073-021-00891-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Chelladurai P, Boucherat O, Stenmark K, Kracht M, Seeger W, Bauer UM, Bonnet S, Pullamsetti SS. Targeting histone acetylation in pulmonary hypertension and right ventricular hypertrophy. Br J Pharmacol 178: 54–71, 2021. doi: 10.1111/bph.14932. [DOI] [PubMed] [Google Scholar]
  • 35.Li F, Wang D, Wang H, Chen L, Sun X, Wan Y. Inhibition of HDAC1 alleviates monocrotaline-induced pulmonary arterial remodeling through up-regulation of miR-34a. Respir Res 22: 239, 2021. doi: 10.1186/s12931-021-01832-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Sun Y, Sangam S, Guo Q, Wang J, Tang H, Black SM, Desai AA. Sex differences, estrogen metabolism and signaling in the development of pulmonary arterial hypertension. Front Cardiovasc Med 8: 719058, 2021. doi: 10.3389/fcvm.2021.719058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Oakland HT, Joseph P. Sex differences and the role of sex hormones in pulmonary hypertension. Clin Chest Med 42: 457–465, 2021. doi: 10.1016/j.ccm.2021.04.006. [DOI] [PubMed] [Google Scholar]
  • 38.Qin S, Predescu D, Carman B, Patel P, Chen J, Kim M, Lahm T, Geraci M, Predescu SA. Up-regulation of the long noncoding RNA X-inactive-specific transcript and the sex bias in pulmonary arterial hypertension. Am J Pathol 191: 1135–1150, 2021. doi: 10.1016/j.ajpath.2021.03.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Amgoud Y, Senbel A, Bouhadoun A, Abdelazeem H, Ozen G, Savané I, Manikpurage HD, Mani S, Tran-Dinh A, Castier Y, Guyard A, Longrois D, Silverstein AM, Norel X. In search of pulmonary hypertension treatments: effect of 17β-estradiol on PGI2 pathway in human pulmonary artery. Prostaglandins Leukot Essent Fatty Acids 172: 102321, 2021. doi: 10.1016/j.plefa.2021.102321. [DOI] [PubMed] [Google Scholar]
  • 40.Frump AL, Albrecht M, Yakubov B, Breuils-Bonnet S, Nadeau V, Tremblay E, Potus F, Omura J, Cook T, Fisher A, Rodriguez B, Brown RD, Stenmark KR, Rubinstein CD, Krentz K, Tabima DM, Li R, Sun X, Chesler NC, Provencher S, Bonnet S, Lahm T. 17β-Estradiol and estrogen receptor alpha protect right ventricular function in pulmonary hypertension via BMPR2 and apelin. J Clin Invest 131: e129433, 2021. doi: 10.1172/JCI129433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Tofovic SP, Zhang X, Jones TJ, Petruševska G. 2-Methoxyestradiol attenuates the development and retards the progression of hypoxia-and α-naphthylthiourea-induced pulmonary hypertension. Pril (Makedon Akad Nauk Umet Odd Med Nauki) 42: 41–51, 2021. doi: 10.2478/prilozi-2021-0003. [DOI] [PubMed] [Google Scholar]
  • 42.Ramirez RL 3rd, Pienkos SM, de Jesus Perez V, Zamanian RT. Pulmonary arterial hypertension secondary to drugs and toxins. Clin Chest Med 42: 19–38, 2021. doi: 10.1016/j.ccm.2020.11.008. [DOI] [PubMed] [Google Scholar]
  • 43.Evans CE, Cober ND, Dai Z, Stewart DJ, Zhao YY. Endothelial cells in the pathogenesis of pulmonary arterial hypertension. Eur Respir J 58: 2003957, 2021. doi: 10.1183/13993003.03957-2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Budhiraja R, Tuder RM, Hassoun PM. Endothelial dysfunction in pulmonary hypertension. Circulation 109: 159–165, 2004. doi: 10.1161/01.CIR.0000102381.57477.50. [DOI] [PubMed] [Google Scholar]
  • 45.Rich S. The effects of vasodilators in pulmonary hypertension: pulmonary vascular or peripheral vascular? Circ Heart Fail 2: 145–150, 2009. doi: 10.1161/CIRCHEARTFAILURE.108.805374. [DOI] [PubMed] [Google Scholar]
  • 46.Tang H, Yamamura A, Yamamura H, Song S, Fraidenburg DR, Chen J, Gu Y, Pohl NM, Zhou T, Jimenez-Perez L, Ayon RJ, Desai AA, Goltzman D, Rischard F, Khalpey Z, Black SM, Garcia JG, Makino A, Yuan JX. Pathogenic role of calcium-sensing receptors in the development and progression of pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 310: L846–L859, 2016. doi: 10.1152/ajplung.00050.2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Doggrell SA. Rho-kinase inhibitors show promise in pulmonary hypertension. Expert Opin Investig Drugs 14: 1157–1159, 2005. doi: 10.1517/13543784.14.9.1157. [DOI] [PubMed] [Google Scholar]
  • 48.Durmowicz AG, Stenmark KR. Mechanisms of structural remodeling in chronic pulmonary hypertension. Pediatr Rev 20: e91–e102, 1999. [PubMed] [Google Scholar]
  • 49.Rensen SS, Doevendans PA, van Eys GJ. Regulation and characteristics of vascular smooth muscle cell phenotypic diversity. Neth Heart J 15: 100–108, 2007. doi: 10.1007/BF03085963. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Undem C, Luke T, Shimoda LA. Contribution of elevated intracellular calcium to pulmonary arterial myocyte alkalinization during chronic hypoxia. Pulm Circ 6: 93–102, 2016. doi: 10.1086/685053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Laudi S, Trump S, Schmitz V, West J, McMurtry IF, Mutlak H, Christians U, Weimann J, Kaisers U, Steudel W. Serotonin transporter protein in pulmonary hypertensive rats treated with atorvastatin. Am J Physiol Lung Cell Mol Physiol 293: L630–L638, 2007. doi: 10.1152/ajplung.00110.2006. [DOI] [PubMed] [Google Scholar]
  • 52.Crossno JT Jr, Garat CV, Reusch JE, Morris KG, Dempsey EC, McMurtry IF, Stenmark KR, Klemm DJ. Rosiglitazone attenuates hypoxia-induced pulmonary arterial remodeling. Am J Physiol Lung Cell Mol Physiol 292: L885–L897, 2007. doi: 10.1152/ajplung.00258.2006. [DOI] [PubMed] [Google Scholar]
  • 53.Schermuly RT, Kreisselmeier KP, Ghofrani HA, Yilmaz H, Butrous G, Ermert L, Ermert M, Weissmann N, Rose F, Guenther A, Walmrath D, Seeger W, Grimminger F. Chronic sildenafil treatment inhibits monocrotaline-induced pulmonary hypertension in rats. Am J Respir Crit Care Med 169: 39–45, 2004. doi: 10.1164/rccm.200302-282OC. [DOI] [PubMed] [Google Scholar]
  • 54.Guilluy C, Sauzeau V, Rolli-Derkinderen M, Guérin P, Sagan C, Pacaud P, Loirand G. Inhibition of RhoA/Rho kinase pathway is involved in the beneficial effect of sildenafil on pulmonary hypertension. Br J Pharmacol 146: 1010–1018, 2005. doi: 10.1038/sj.bjp.0706408. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Inagaki T, Pearson JT, Tsuchimochi H, Schwenke DO, Saito S, Higuchi T, Masaki T, Umetani K, Shirai M, Nakaoka Y. Evaluation of right coronary vascular dysfunction in severe pulmonary hypertensive rats using synchrotron radiation microangiography. Am J Physiol Heart Circ Physiol 320: H1021–H1036, 2021. doi: 10.1152/ajpheart.00327.2020. [DOI] [PubMed] [Google Scholar]
  • 56.Schermuly RT, Dony E, Ghofrani HA, Pullamsetti S, Savai R, Roth M, Sydykov A, Lai YJ, Weissmann N, Seeger W, Grimminger F. Reversal of experimental pulmonary hypertension by PDGF inhibition. J Clin Invest 115: 2811–2821, 2005. doi: 10.1172/JCI24838. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Nishimura T, Vaszar LT, Faul JL, Zhao G, Berry GJ, Shi L, Qiu D, Benson G, Pearl RG, Kao PN. Simvastatin rescues rats from fatal pulmonary hypertension by inducing apoptosis of neointimal smooth muscle cells. Circulation 108: 1640–1645, 2003. doi: 10.1161/01.CIR.0000087592.47401.37. [DOI] [PubMed] [Google Scholar]
  • 58.McMurtry MS, Archer SL, Altieri DC, Bonnet S, Haromy A, Harry G, Bonnet S, Puttagunta L, Michelakis ED. Gene therapy targeting survivin selectively induces pulmonary vascular apoptosis and reverses pulmonary arterial hypertension. J Clin Invest 115: 1479–1491, 2005. doi: 10.1172/JCI23203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Ito T, Okada T, Miyashita H, Nomoto T, Nonaka-Sarukawa M, Uchibori R, Maeda Y, Urabe M, Mizukami H, Kume A, Takahashi M, Ikeda U, Shimada K, Ozawa K. Interleukin-10 expression mediated by an adeno-associated virus vector prevents monocrotaline-induced pulmonary arterial hypertension in rats. Circ Res 101: 734–741, 2007. doi: 10.1161/CIRCRESAHA.107.153023. [DOI] [PubMed] [Google Scholar]
  • 60.Hoshikawa Y, Nana-Sinkam P, Moore MD, Sotto-Santiago S, Phang T, Keith RL, Morris KG, Kondo T, Tuder RM, Voelkel NF, Geraci MW. Hypoxia induces different genes in the lungs of rats compared with mice. Physiol Genomics 12: 209–219, 2003. [Erratum in Physiol Genomics 13: 79, 2003]. doi: 10.1152/physiolgenomics.00081.2001. [DOI] [PubMed] [Google Scholar]
  • 61.Abe K, Toba M, Alzoubi A, Ito M, Fagan KA, Cool CD, Voelkel NF, McMurtry IF, Oka M. Formation of plexiform lesions in experimental severe pulmonary arterial hypertension. Circulation 121: 2747–2754, 2010. doi: 10.1161/CIRCULATIONAHA.109.927681. [DOI] [PubMed] [Google Scholar]
  • 62.Taraseviciene-Stewart L, Kasahara Y, Alger L, Hirth P, Mc Mahon G, Waltenberger J, Voelkel NF, Tuder RM. Inhibition of the VEGF receptor 2 combined with chronic hypoxia causes cell death-dependent pulmonary endothelial cell proliferation and severe pulmonary hypertension. FASEB J 15: 427–438, 2001. doi: 10.1096/fj.00-0343com. [DOI] [PubMed] [Google Scholar]
  • 63.de Raaf MA, Schalij I, Gomez-Arroyo J, Rol N, Happé C, de Man FS, Vonk-Noordegraaf A, Westerhof N, Voelkel NF, Bogaard HJ. SuHx rat model: partly reversible pulmonary hypertension and progressive intima obstruction. Eur Respir J 44: 160–168, 2014. doi: 10.1183/09031936.00204813. [DOI] [PubMed] [Google Scholar]
  • 64.Zhou C, Crockett ES, Batten L, McMurtry IF, Stevens T. Pulmonary vascular dysfunction secondary to pulmonary arterial hypertension: insights gained through retrograde perfusion. Am J Physiol Lung Cell Mol Physiol 314: L835–L845, 2018. doi: 10.1152/ajplung.00201.2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.O'Farrell AM, Yuen HA, Smolich B, Hannah AL, Louie SG, Hong W, Stopeck AT, Silverman LR, Lancet JE, Karp JE, Albitar M, Cherrington JM, Giles FJ. Effects of SU5416, a small molecule tyrosine kinase receptor inhibitor, on FLT3 expression and phosphorylation in patients with refractory acute myeloid leukemia. Leuk Res 28: 679–689, 2004. doi: 10.1016/j.leukres.2003.11.004. [DOI] [PubMed] [Google Scholar]
  • 66.Zhou C, Townsley MI, Alexeyev M, Voelkel NF, Stevens T. Endothelial hyperpermeability in severe pulmonary arterial hypertension: role of store-operated calcium entry. Am J Physiol Lung Cell Mol Physiol 311: L560–L569, 2016. doi: 10.1152/ajplung.00057.2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Aiello RJ, Bourassa PA, Zhang Q, Dubins J, Goldberg DR, De Lombaert S, Humbert M, Guignabert C, Cavasin MA, McKinsey TA, Paralkar V. Tryptophan hydroxylase 1 inhibition impacts pulmonary vascular remodeling in two rat models of pulmonary hypertension. J Pharmacol Exp Ther 360: 267–279, 2017. doi: 10.1124/jpet.116.237933. [DOI] [PubMed] [Google Scholar]
  • 68.Christou H, Hudalla H, Michael Z, Filatava EJ, Li J, Zhu M, Possomato-Vieira JS, Dias-Junior C, Kourembanas S, Khalil RA. Impaired pulmonary arterial vasoconstriction and nitric oxide-mediated relaxation underlie severe pulmonary hypertension in the sugen-hypoxia rat model. J Pharmacol Exp Ther 364: 258–274, 2018. doi: 10.1124/jpet.117.244798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Lo CCW, Moosavi SM, Bubb KJ. The regulation of pulmonary vascular tone by neuropeptides and the implications for pulmonary hypertension. Front Physiol 9: 1167, 2018. doi: 10.3389/fphys.2018.01167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Maron BA, Leopold JA. The role of the renin-angiotensin-aldosterone system in the pathobiology of pulmonary arterial hypertension (2013 Grover Conference series). Pulm Circ 4: 200–210, 2014. doi: 10.1086/675984. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Hemnes AR, Rathinasabapathy A, Austin EA, Brittain EL, Carrier EJ, Chen X, Fessel JP, Fike CD, Fong P, Fortune N, Gerszten RE, Johnson JA, Kaplowitz M, Newman JH, Piana R, Pugh ME, Rice TW, Robbins IM, Wheeler L, Yu C, Loyd JE, West J. A potential therapeutic role for angiotensin-converting enzyme 2 in human pulmonary arterial hypertension. Eur Respir J 51: 1702638, 2018. doi: 10.1183/13993003.02638-2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Chen IC, Lin JY, Liu YC, Chai CY, Yeh JL, Hsu JH, Wu BN, Dai ZK. Angiotensin-converting enzyme 2 activator ameliorates severe pulmonary hypertension in a rat model of left pneumonectomy combined with VEGF inhibition. Front Med (Lausanne) 8: 619133, 2021. doi: 10.3389/fmed.2021.619133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Chen IC, Lin JY, Liu YC, Chai CY, Yeh JL, Hsu JH, Wu BN, Dai ZK. The beneficial effects of angiotensin-converting enzyme II (ACE2) activator in pulmonary hypertension secondary to left ventricular dysfunction. Int J Med Sci 17: 2594–2602, 2020. doi: 10.7150/ijms.48096. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Shenoy V, Gjymishka A, Jarajapu YP, Qi Y, Afzal A, Rigatto K, Ferreira AJ, Fraga-Silva RA, Kearns P, Douglas JY, Agarwal D, Mubarak KK, Bradford C, Kennedy WR, Jun JY, Rathinasabapathy A, Bruce E, Gupta D, Cardounel AJ, Mocco J, Patel JM, Francis J, Grant MB, Katovich MJ, Raizada MK. Diminazene attenuates pulmonary hypertension and improves angiogenic progenitor cell functions in experimental models. Am J Respir Crit Care Med 187: 648–657, 2013. doi: 10.1164/rccm.201205-0880OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Witkowski A, Szumowski Ł, Urbanek P, Jastrzębski J, Skowroński J, Sobieszczańska-Małek M, Hoffman P, Kowalik E, Wiśniewska J, Banasiak A, Parulski A, Zieliński T. Transcatheter pulmonary denervation in patients with left heart failure with reduced ejection fraction and combined precapillary and postcapillary pulmonary hypertension: a prospective single center experience. Catheter Cardiovasc Interv 98: 588–594, 2021. doi: 10.1002/ccd.29526. [DOI] [PubMed] [Google Scholar]
  • 76.Qiu H, Zhang Y, Li Z, Jiang P, Guo S, He Y, Guo Y. Donepezil ameliorates pulmonary arterial hypertension by inhibiting M2-macrophage activation. Front Cardiovasc Med 8: 639541, 2021. doi: 10.3389/fcvm.2021.639541. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Humbert M, Guignabert C, Bonnet S, Dorfmuller P, Klinger JR, Nicolls MR, Olschewski AJ, Pullamsetti SS, Schermuly RT, Stenmark KR, Rabinovitch M. Pathology and pathobiology of pulmonary hypertension: state of the art and research perspectives. Eur Respir J 53: 1801887, 2019. doi: 10.1183/13993003.01887-2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Diebold I, Hennigs JK, Miyagawa K, Li CG, Nickel NP, Kaschwich M, Cao A, Wang L, Reddy S, Chen PI, Nakahira K, Alcazar MA, Hopper RK, Ji L, Feldman BJ, Rabinovitch M. BMPR2 preserves mitochondrial function and DNA during reoxygenation to promote endothelial cell survival and reverse pulmonary hypertension. Cell Metab 21: 596–608, 2015. doi: 10.1016/j.cmet.2015.03.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Alastalo T-P, Li M, Perez VdJ, Pham D, Sawada H, Wang JK, Koskenvuo M, Wang L, Freeman BA, Chang HY, Rabinovitch M. Disruption of PPARγ/β-catenin-mediated regulation of apelin impairs BMP-induced mouse and human pulmonary arterial EC survival. J Clin Invest 121: 3735–3746, 2011. doi: 10.1172/JCI43382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Hennigs JK, Cao A, Li CG, Shi M, Mienert J, Miyagawa K, Korbelin J, Marciano DP, Chen PI, Roughley M, Elliott MV, Harper RL, Bill MA, Chappell J, Moonen JR, Diebold I, Wang L, Snyder MP, Rabinovitch M. PPARγ-p53-mediated vasculoregenerative program to reverse pulmonary hypertension. Circ Res 128: 401–418, 2021. doi: 10.1161/CIRCRESAHA.119.316339. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Culley MK, Zhao J, Tai YY, Tang Y, Perk D, Negi V, Yu Q, Woodcock CC, Handen A, Speyer G, Kim S, Lai YC, Satoh T, Watson AM, Aaraj YA, Sembrat J, Rojas M, Goncharov D, Goncharova EA, Khan OF, Anderson DG, Dahlman JE, Gurkar AU, Lafyatis R, Fayyaz AU, Redfield MM, Gladwin MT, Rabinovitch M, Gu M, Bertero T, Chan SY. Frataxin deficiency promotes endothelial senescence in pulmonary hypertension. J Clin Invest 131: e136459, 2021. doi: 10.1172/JCI136459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Giaid A, Saleh D. Reduced expression of endothelial nitric oxide synthase in the lungs of patients with pulmonary hypertension. N Engl J Med 333: 214–221, 1995. doi: 10.1056/NEJM199507273330403. [DOI] [PubMed] [Google Scholar]
  • 83.Adnot S, Raffestin B, Eddahibi S, Braquet P, Chabrier PE. Loss of endothelium-dependent relaxant activity in the pulmonary circulation of rats exposed to chronic hypoxia. J Clin Invest 87: 155–162, 1991. doi: 10.1172/JCI114965. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Mam V, Tanbe AF, Vitali SH, Arons E, Christou HA, Khalil RA. Impaired vasoconstriction and nitric oxide-mediated relaxation in pulmonary arteries of hypoxia- and monocrotaline-induced pulmonary hypertensive rats. J Pharmacol Exp Ther 332: 455–462, 2010. doi: 10.1124/jpet.109.160119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Nozik-Grayck E, Stenmark KR. Role of reactive oxygen species in chronic hypoxia-induced pulmonary hypertension and vascular remodeling. Adv Exp Med Biol 618: 101–112, 2007. doi: 10.1007/978-0-387-75434-5_8. [DOI] [PubMed] [Google Scholar]
  • 86.Fullerton DA, Hahn AR, McIntyre RC Jr.. Mechanistic imbalance of pulmonary vasomotor control in progressive lung injury. Surgery 119: 98–103, 1996. doi: 10.1016/S0039-6060(96)80220-0. [DOI] [PubMed] [Google Scholar]
  • 87.Nakane T, Esaki J, Ueda R, Honda M, Okabayashi H. Inhaled nitric oxide improves pulmonary hypertension and organ functions after adult heart valve surgeries. Gen Thorac Cardiovasc Surg 69: 1519–1526, 2021. doi: 10.1007/s11748-021-01651-z. [DOI] [PubMed] [Google Scholar]
  • 88.Sun L, Wang C, Zhou Y, Sun W. Clinical efficacy and safety of different doses of sildenafil in the treatment of persistent pulmonary hypertension of the newborn: a network meta-analysis. Front Pharmacol 12: 697287, 2021. doi: 10.3389/fphar.2021.697287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Reinero M, Beghetti M, Tozzi P, Segesser LKV, Samaja M, Milano G. Nitric oxide-cGMP pathway modulation in an experimental model of hypoxic pulmonary hypertension. J Cardiovasc Pharmacol Ther 26: 665–676, 2021. doi: 10.1177/10742484211014162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Michelakis ED, Gurtu V, Webster L, Barnes G, Watson G, Howard L, Cupitt J, Paterson I, Thompson RB, Chow K, O'Regan DP, Zhao L, Wharton J, Kiely DG, Kinnaird A, Boukouris AE, White C, Nagendran J, Freed DH, Wort SJ, Gibbs JSR, Wilkins MR. Inhibition of pyruvate dehydrogenase kinase improves pulmonary arterial hypertension in genetically susceptible patients. Sci Transl Med 9: eaao4583, 2017. doi: 10.1126/scitranslmed.aao4583. [DOI] [PubMed] [Google Scholar]
  • 91.Gonzaga-Costa K, Roque CR, Vasconcelos-Silva AA, Sousa-Brito HL, Martins CS, Caetano-Souza MM, Duarte GP, da Silva JKR, Borges RS, Dos Santos AA, Magalhães PJC, Lahlou S. The soluble guanylate cyclase stimulator, 1-nitro-2-phenylethane, reverses monocrotaline-induced pulmonary arterial hypertension in rats. Life Sci 275: 119334, 2021. doi: 10.1016/j.lfs.2021.119334. [DOI] [PubMed] [Google Scholar]
  • 92.Gonzaga-Costa K, Vasconcelos-Silva AA, Rodrigues-Silva MJ, Rebouça C, Duarte GP, Borges RS, Magãlhaes PJC, Lahlou S. Soluble guanylate cyclase stimulator, trans-4-methoxy-β-nitrostyrene, has a beneficial effect in monocrotaline-induced pulmonary arterial hypertension in rats. Eur J Pharmacol 897: 173948, 2021. doi: 10.1016/j.ejphar.2021.173948. [DOI] [PubMed] [Google Scholar]
  • 93.Christou H, Michael Z, Spyropoulos F, Chen Y, Rong D, Khalil RA. Carbonic anhydrase inhibition improves pulmonary artery reactivity and nitric oxide-mediated relaxation in sugen-hypoxia model of pulmonary hypertension. Am J Physiol Regul Integr Comp Physiol 320: R835–R850, 2021. doi: 10.1152/ajpregu.00362.2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Aamand R, Dalsgaard T, Jensen FB, Simonsen U, Roepstorff A, Fago A. Generation of nitric oxide from nitrite by carbonic anhydrase: a possible link between metabolic activity and vasodilation. Am J Physiol Heart Circ Physiol 297: H2068–H2074, 2009. doi: 10.1152/ajpheart.00525.2009. [DOI] [PubMed] [Google Scholar]
  • 95.Whittle BJ, Silverstein AM, Mottola DM, Clapp LH. Binding and activity of the prostacyclin receptor (IP) agonists, treprostinil and iloprost, at human prostanoid receptors: treprostinil is a potent DP1 and EP2 agonist. Biochem Pharmacol 84: 68–75, 2012. doi: 10.1016/j.bcp.2012.03.012. [DOI] [PubMed] [Google Scholar]
  • 96.Majed BH, Khalil RA. Molecular mechanisms regulating the vascular prostacyclin pathways and their adaptation during pregnancy and in the newborn. Pharmacol Rev 64: 540–582, 2012. doi: 10.1124/pr.111.004770. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Hoshikawa Y, Voelkel NF, Gesell TL, Moore MD, Morris KG, Alger LA, Narumiya S, Geraci MW. Prostacyclin receptor-dependent modulation of pulmonary vascular remodeling. Am J Respir Crit Care Med 164: 314–318, 2001. doi: 10.1164/ajrccm.164.2.2010150. [DOI] [PubMed] [Google Scholar]
  • 98.Miyata M, Ueno Y, Sekine H, Ito O, Sakuma F, Koike H, Nishio S, Nishimaki T, Kasukawa R. Protective effect of beraprost sodium, a stable prostacyclin analogue, in development of monocrotaline-induced pulmonary hypertension. J Cardiovasc Pharmacol 27: 20–26, 1996. doi: 10.1097/00005344-199601000-00004. [DOI] [PubMed] [Google Scholar]
  • 99.Geraci MW, Gao B, Shepherd DC, Moore MD, Westcott JY, Fagan KA, Alger LA, Tuder RM, Voelkel NF. Pulmonary prostacyclin synthase overexpression in transgenic mice protects against development of hypoxic pulmonary hypertension. J Clin Invest 103: 1509–1515, 1999. doi: 10.1172/JCI5911. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Hill LL, Pearl RG. Combined inhaled nitric oxide and inhaled prostacyclin during experimental chronic pulmonary hypertension. J Appl Physiol (1985) 86: 1160–1164, 1999. doi: 10.1152/jappl.1999.86.4.1160. [DOI] [PubMed] [Google Scholar]
  • 101.Parkington HC, Coleman HA, Tare M. Prostacyclin and endothelium-dependent hyperpolarization. Pharmacol Res 49: 509–514, 2004. doi: 10.1016/j.phrs.2003.11.012. [DOI] [PubMed] [Google Scholar]
  • 102.Ozen G, Amgoud Y, Abdelazeem H, Mani S, Benyahia C, Bouhadoun A, Tran-Dinh A, Castier Y, Guyard A, Longrois D, Silverstein AM, Norel X. Downregulation of PGI2 pathway in pulmonary hypertension group-III patients. Prostaglandins Leukot Essent Fatty Acids 160: 102158, 2020. doi: 10.1016/j.plefa.2020.102158. [DOI] [PubMed] [Google Scholar]
  • 103.Christman BW, McPherson CD, Newman JH, King GA, Bernard GR, Groves BM, Loyd JE. An imbalance between the excretion of thromboxane and prostacyclin metabolites in pulmonary hypertension. N Engl J Med 327: 70–75, 1992. doi: 10.1056/NEJM199207093270202. [DOI] [PubMed] [Google Scholar]
  • 104.Tuder RM, Cool CD, Geraci MW, Wang J, Abman SH, Wright L, Badesch D, Voelkel NF. Prostacyclin synthase expression is decreased in lungs from patients with severe pulmonary hypertension. Am J Respir Crit Care Med 159: 1925–1932, 1999. doi: 10.1164/ajrccm.159.6.9804054. [DOI] [PubMed] [Google Scholar]
  • 105.Galiè N, Gaine S, Channick R, Coghlan JG, Hoeper MM, Lang IM, McLaughlin VV, Lassen C, Rubin LJ, Hsu Schmitz SF, Sitbon O, Tapson VF, Chin KM. Long-term survival, safety and tolerability with selexipag in patients with pulmonary arterial hypertension: results from GRIPHON and its open-label extension. Adv Ther 39: 796–810, 2022. doi: 10.1007/s12325-021-01898-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Douwes JM, Zijlstra WM, Rosenzweig EB, Ploegstra MJ, Krishnan US, Haarman MG, Roofthooft MTR, Postmus D, Hillege HL, Ivy DD, Berger RMF. Parenteral prostanoids in pediatric pulmonary arterial hypertension: start early, dose high, combine. Ann Am Thorac Soc 19: 227–237, 2022. doi: 10.1513/AnnalsATS.202012-1563OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Félétou M, Vanhoutte PM. Endothelium-derived hyperpolarizing factor: where are we now? Arterioscler Thromb Vasc Biol 26: 1215–1225, 2006. doi: 10.1161/01.ATV.0000217611.81085.c5. [DOI] [PubMed] [Google Scholar]
  • 108.Daneva Z, Marziano C, Ottolini M, Chen YL, Baker TM, Kuppusamy M, Zhang A, Ta HQ, Reagan CE, Mihalek AD, Kasetti RB, Shen Y, Isakson BE, Minshall RD, Zode GS, Goncharova EA, Laubach VE, Sonkusare SK. Caveolar peroxynitrite formation impairs endothelial TRPV4 channels and elevates pulmonary arterial pressure in pulmonary hypertension. Proc Natl Acad Sci USA 118: e2023130118, 2021. doi: 10.1073/pnas.2023130118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Mondéjar-Parreno G, Cogolludo A, Perez-Vizcaino F. Potassium (K+) channels in the pulmonary vasculature: implications in pulmonary hypertension physiological, pathophysiological and pharmacological regulation. Pharmacol Ther 225: 107835, 2021. doi: 10.1016/j.pharmthera.2021.107835. [DOI] [PubMed] [Google Scholar]
  • 110.Jackson WF, Konig A, Dambacher T, Busse R. Prostacyclin-induced vasodilation in rabbit heart is mediated by ATP-sensitive potassium channels. Am J Physiol Heart Circ Physiol 264: H238–H243, 1993. doi: 10.1152/ajpheart.1993.264.1.H238. [DOI] [PubMed] [Google Scholar]
  • 111.Michelakis ED, McMurtry MS, Sonnenberg B, Archer SL. The NO - K+ channel axis in pulmonary arterial hypertension. Activation by experimental oral therapies. Adv Exp Med Biol 543: 293–322, 2003. doi: 10.1007/978-1-4419-8997-0_21. [DOI] [PubMed] [Google Scholar]
  • 112.Tang B, Li Y, Nagaraj C, Morty RE, Gabor S, Stacher E, Voswinckel R, Weissmann N, Leithner K, Olschewski H, Olschewski A. Endothelin-1 inhibits background two-pore domain channel TASK-1 in primary human pulmonary artery smooth muscle cells. Am J Respir Cell Mol Biol 41: 476–483, 2009. doi: 10.1165/rcmb.2008-0412OC. [DOI] [PubMed] [Google Scholar]
  • 113.Pozeg ZI, Michelakis ED, McMurtry MS, Thébaud B, Wu XC, Dyck JR, Hashimoto K, Wang S, Moudgil R, Harry G, Sultanian R, Koshal A, Archer SL. In vivo gene transfer of the O2-sensitive potassium channel Kv1.5 reduces pulmonary hypertension and restores hypoxic pulmonary vasoconstriction in chronically hypoxic rats. Circulation 107: 2037–2044, 2003. doi: 10.1161/01.CIR.0000062688.76508.B3. [DOI] [PubMed] [Google Scholar]
  • 114.Mondejar-Parreño G, Callejo M, Barreira B, Morales-Cano D, Esquivel-Ruiz S, Moreno L, Cogolludo A, Perez-Vizcaino F. miR-1 is increased in pulmonary hypertension and downregulates Kv1.5 channels in rat pulmonary arteries. J Physiol 597: 1185–1197, 2019. doi: 10.1113/JP276054. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Ma L, Roman-Campos D, Austin ED, Eyries M, Sampson KS, Soubrier F, Germain M, Trégouët DA, Borczuk A, Rosenzweig EB, Girerd B, Montani D, Humbert M, Loyd JE, Kass RS, Chung WK. A novel channelopathy in pulmonary arterial hypertension. N Engl J Med 369: 351–361, 2013. doi: 10.1056/NEJMoa1211097. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Bohnen MS, Roman-Campos D, Terrenoire C, Jnani J, Sampson KJ, Chung WK, Kass RS. The impact of heterozygous KCNK3 mutations associated with pulmonary arterial hypertension on channel function and pharmacological recovery. J Am Heart Assoc 6: e006465, 2017. doi: 10.1161/JAHA.117.006465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Morecroft I, Murray A, Nilsen M, Gurney AM, MacLean MR. Treatment with the Kv7 potassium channel activator flupirtine is beneficial in two independent mouse models of pulmonary hypertension. Br J Pharmacol 157: 1241–1249, 2009. doi: 10.1111/j.1476-5381.2009.00283.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Revermann M, Schloss M, Mieth A, Babelova A, Schröder K, Neofitidou S, Buerkl J, Kirschning T, Schermuly RT, Hofstetter C, Brandes RP. Levosimendan attenuates pulmonary vascular remodeling. Intensive Care Med 37: 1368–1377, 2011. doi: 10.1007/s00134-011-2254-9. [DOI] [PubMed] [Google Scholar]
  • 119.Nagaraj C, Tang B, Nagy BM, Papp R, Jain PP, Marsh LM, Meredith AL, Ghanim B, Klepetko W, Kwapiszewska G, Weir EK, Olschewski H, Olschewski A. Docosahexaenoic acid causes rapid pulmonary arterial relaxation via KCa channel-mediated hyperpolarisation in pulmonary hypertension. Eur Respir J 48: 1127–1136, 2016. doi: 10.1183/13993003.01814-2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Callejo M, Mondejar-Parreño G, Morales-Cano D, Barreira B, Esquivel-Ruiz S, Olivencia MA, Manaud G, Perros F, Duarte J, Moreno L, Cogolludo A, Perez-Vizcaíno F. Vitamin D deficiency downregulates TASK-1 channels and induces pulmonary vascular dysfunction. Am J Physiol Lung Cell Mol Physiol 319: L627–L640, 2020. doi: 10.1152/ajplung.00475.2019. [DOI] [PubMed] [Google Scholar]
  • 121.Wong CM, Bansal G, Pavlickova L, Marcocci L, Suzuki YJ. Reactive oxygen species and antioxidants in pulmonary hypertension. Antioxid Redox Signal 18: 1789–1796, 2013. doi: 10.1089/ars.2012.4568. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Jain PP, Zhao T, Xiong M, Song S, Lai N, Zheng Q, Chen J, Carr SG, Babicheva A, Izadi A, Rodriguez M, Rahimi S, Balistrieri F, Rahimi S, Simonson T, Valdez-Jasso D, Thistlethwaite PA, Shyy JY-J, Wang J, Makino A, Yuan JX-J. Halofuginone, a promising drug for treatment of pulmonary hypertension. Br J Pharmacol 178: 3373–3394, 2021. doi: 10.1111/bph.15442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Roubenne L, Marthan R, Le Grand B, Guibert C. Hydrogen sulfide metabolism and pulmonary hypertension. Cells 10: 1477, 2021. doi: 10.3390/cells10061477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Schiffrin EL, Touyz RM. Vascular biology of endothelin. J Cardiovasc Pharmacol 32, Suppl 3: S2–S13, 1998. [PubMed] [Google Scholar]
  • 125.Smith L, Payne JA, Sedeek MH, Granger JP, Khalil RA. Endothelin-induced increases in Ca2+ entry mechanisms of vascular contraction are enhanced during high-salt diet. Hypertension 41: 787–793, 2003. doi: 10.1161/01.HYP.0000051643.05700.56. [DOI] [PubMed] [Google Scholar]
  • 126.McNair LL, Salamanca DA, Khalil RA. Endothelin-1 promotes Ca2+ antagonist-insensitive coronary smooth muscle contraction via activation of epsilon-protein kinase C. Hypertension 43: 897–904, 2004. doi: 10.1161/01.HYP.0000118520.92686.3b. [DOI] [PubMed] [Google Scholar]
  • 127.Stewart DJ, Levy RD, Cernacek P, Langleben D. Increased plasma endothelin-1 in pulmonary hypertension: marker or mediator of disease? Ann Intern Med 114: 464–469, 1991. doi: 10.7326/0003-4819-114-6-464. [DOI] [PubMed] [Google Scholar]
  • 128.Itoh H, Yokochi A, Yamauchi-Kohno R, Maruyama K. Effects of the endothelin ET(A) receptor antagonist, TA-0201, on pulmonary arteries isolated from hypoxic rats. Eur J Pharmacol 376: 233–238, 1999. doi: 10.1016/s0014-2999(99)00380-5. [DOI] [PubMed] [Google Scholar]
  • 129.Apostolopoulou SC, Rammos S, Kyriakides ZS, Webb DJ, Johnston NR, Cokkinos DV, Kremastinos DT. Acute endothelin A receptor antagonism improves pulmonary and systemic haemodynamics in patients with pulmonary arterial hypertension that is primary or autoimmune and related to congenital heart disease. Heart 89: 1221–1226, 2003. doi: 10.1136/heart.89.10.1221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Williamson DJ, Wallman LL, Jones R, Keogh AM, Scroope F, Penny R, Weber C, Macdonald PS. Hemodynamic effects of Bosentan, an endothelin receptor antagonist, in patients with pulmonary hypertension. Circulation 102: 411–418, 2000. doi: 10.1161/01.CIR.102.4.411. [DOI] [PubMed] [Google Scholar]
  • 131.Rubin LJ, Badesch DB, Barst RJ, Galie N, Black CM, Keogh A, Pulido T, Frost A, Roux S, Leconte I, Landzberg M, Simonneau G. Bosentan therapy for pulmonary arterial hypertension. N Engl J Med 346: 896–903, 2002. doi: 10.1056/NEJMoa012212. [DOI] [PubMed] [Google Scholar]
  • 132.Barst RJ, Rich S, Widlitz A, Horn EM, McLaughlin V, McFarlin J. Clinical efficacy of sitaxsentan, an endothelin-A receptor antagonist, in patients with pulmonary arterial hypertension: open-label pilot study. Chest 121: 1860–1868, 2002. doi: 10.1378/chest.121.6.1860. [DOI] [PubMed] [Google Scholar]
  • 133.Ivy DD, Yanagisawa M, Gariepy CE, Gebb SA, Colvin KL, McMurtry IF. Exaggerated hypoxic pulmonary hypertension in endothelin B receptor-deficient rats. Am J Physiol Lung Cell Mol Physiol 282: L703–L712, 2002. doi: 10.1152/ajplung.00272.2001. [DOI] [PubMed] [Google Scholar]
  • 134.Ambalavanan N, Bulger A, Murphy-Ullrich J, Oparil S, Chen YF. Endothelin-A receptor blockade prevents and partially reverses neonatal hypoxic pulmonary vascular remodeling. Pediatr Res 57: 631–636, 2005. doi: 10.1203/01.PDR.0000159512.55862.69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Jacobs A, Preston IR, Gomberg-Maitland M. Endothelin receptor antagonism in pulmonary arterial hypertension—a role for selective ET(A) inhibition? Curr Med Res Opin 22: 2567–2574, 2006. doi: 10.1185/030079906X158020. [DOI] [PubMed] [Google Scholar]
  • 136.Christou H, Adatia I, Van Marter LJ, Kane JW, Thompson JE, Stark AR, Wessel DL, Kourembanas S. Effect of inhaled nitric oxide on endothelin-1 and cyclic guanosine 5'-monophosphate plasma concentrations in newborn infants with persistent pulmonary hypertension. J Pediatr 130: 603–611, 1997. doi: 10.1016/s0022-3476(97)70245-2. [DOI] [PubMed] [Google Scholar]
  • 137.Latus H, Karanatsios G, Basan U, Salser K, Müller S, Khalil M, Kreuder J, Schranz D, Apitz C. Clinical and prognostic value of endothelin-1 and big endothelin-1 expression in children with pulmonary hypertension. Heart 102: 1052–1058, 2016. doi: 10.1136/heartjnl-2015-308743. [DOI] [PubMed] [Google Scholar]
  • 138.Goncharova NS, Andreeva EM, Vakhrushev AD, Leandro HIC, Murashova LA, Voronin SE, Korobchenko LE, Mitrofanova LB, Skorik YA, Galagudza MM, Moiseeva OM, Mikhaylov EN. Modeling of acute pulmonary arterial hypertension in pigs using a stable thromboxane A2 analogue (U46619): dose adjustment and assessment of hemodynamic reactions. Bull Exp Biol Med 170: 729–733, 2021. doi: 10.1007/s10517-021-05142-2. [DOI] [PubMed] [Google Scholar]
  • 139.Dulak J, Deshane J, Jozkowicz A, Agarwal A. Heme oxygenase-1 and carbon monoxide in vascular pathobiology: focus on angiogenesis. Circulation 117: 231–241, 2008. doi: 10.1161/CIRCULATIONAHA.107.698316. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Yachie A, Niida Y, Wada T, Igarashi N, Kaneda H, Toma T, Ohta K, Kasahara Y, Koizumi S. Oxidative stress causes enhanced endothelial cell injury in human heme oxygenase-1 deficiency. J Clin Invest 103: 129–135, 1999. doi: 10.1172/JCI4165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Sikorski EM, Hock T, Hill-Kapturczak N, Agarwal A. The story so far: molecular regulation of the heme oxygenase-1 gene in renal injury. Am J Physiol Renal Physiol 286: F425–F441, 2004. doi: 10.1152/ajprenal.00297.2003. [DOI] [PubMed] [Google Scholar]
  • 142.Vitali SH, Mitsialis SA, Liang OD, Liu X, Fernandez-Gonzalez A, Christou H, Wu X, McGowan FX, Kourembanas S. Divergent cardiopulmonary actions of heme oxygenase enzymatic products in chronic hypoxia. PLoS One 4: e5978, 2009. doi: 10.1371/journal.pone.0005978. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Christou H, Morita T, Hsieh CM, Koike H, Arkonac B, Perrella MA, Kourembanas S. Prevention of hypoxia-induced pulmonary hypertension by enhancement of endogenous heme oxygenase-1 in the rat. Circ Res 86: 1224–1229, 2000. [Erratum in Circ Res 90: e66-a, 2002]. doi: 10.1161/01.res.86.12.1224. [DOI] [PubMed] [Google Scholar]
  • 144.Barman SA. Vasoconstrictor effect of endothelin-1 on hypertensive pulmonary arterial smooth muscle involves Rho-kinase and protein kinase C. Am J Physiol Lung Cell Mol Physiol 293: L472–L479, 2007. doi: 10.1152/ajplung.00101.2006. [DOI] [PubMed] [Google Scholar]
  • 145.Gillespie MN, Olson JW, Reinsel CN, O'Connor WN, Altiere RJ. Vascular hyperresponsiveness in perfused lungs from monocrotaline-treated rats. Am J Physiol Heart Circ Physiol 251: H109–H114, 1986. doi: 10.1152/ajpheart.1986.251.1.H109. [DOI] [PubMed] [Google Scholar]
  • 146.Shimoda LA, Sham JS, Sylvester JT. Altered pulmonary vasoreactivity in the chronically hypoxic lung. Physiol Res 49: 549–560, 2000. [PubMed] [Google Scholar]
  • 147.Oka M, Homma N, Taraseviciene-Stewart L, Morris KG, Kraskauskas D, Burns N, Voelkel NF, McMurtry IF. Rho kinase-mediated vasoconstriction is important in severe occlusive pulmonary arterial hypertension in rats. Circ Res 100: 923–929, 2007. doi: 10.1161/01.RES.0000261658.12024.18. [DOI] [PubMed] [Google Scholar]
  • 148.Kourembanas S, Morita T, Liu Y, Christou H. Mechanisms by which oxygen regulates gene expression and cell-cell interaction in the vasculature. Kidney Int 51: 438–443, 1997. doi: 10.1038/ki.1997.58. [DOI] [PubMed] [Google Scholar]
  • 149.Fan C, Su Q, Li Y, Liang L, Angelini DJ, Guggino WB, Johns RA. Hypoxia-induced mitogenic factor/FIZZ1 induces intracellular calcium release through the PLC-IP(3) pathway. Am J Physiol Lung Cell Mol Physiol 297: L263–L270, 2009. doi: 10.1152/ajplung.90416.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Tian H, Liu L, Wu Y, Wang R, Jiang Y, Hu R, Zhu L, Li L, Fang Y, Yang C, Ji L, Liu G, Dai A. Resistin-like molecule β acts as a mitogenic factor in hypoxic pulmonary hypertension via the Ca2+ dependent PI3K/Akt/mTOR and PKC/MAPK signaling pathways. Respir Res 22: 8, 2021. doi: 10.1186/s12931-020-01598-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Jain PP, Lai N, Xiong M, Chen J, Babicheva A, Zhao T, Parmisano S, Zhao M, Paquin C, Matti M, Powers R, Balistrieri A, Kim NH, Valdez-Jasso D, Thistlethwaite PA, Shyy JY, Wang J, Garcia JGN, Makino A, Yuan JX. TRPC6, a therapeutic target for pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 321: L1161–L1182, 2021. doi: 10.1152/ajplung.00159.2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Liao J, Lu W, Chen Y, Duan X, Zhang C, Luo X, Lin Z, Chen J, Liu S, Yan H, Chen Y, Feng H, Zhou D, Chen X, Zhang Z, Yang Q, Liu X, Tang H, Li J, Makino A, Yuan JX-J, Zhong N, Yang K, Wang J. Upregulation of Piezo1 (piezo type mechanosensitive ion channel component 1) enhances the intracellular free calcium in pulmonary arterial smooth muscle cells from idiopathic pulmonary arterial hypertension patients. Hypertension 77: 1974–1989, 2021. doi: 10.1161/HYPERTENSIONAHA.120.16629. [DOI] [PubMed] [Google Scholar]
  • 153.Wang Z, Chen J, Babicheva A, Jain PP, Rodriguez M, Ayon RJ, Ravellette KS, Wu L, Balistrieri F, Tang H, Wu X, Zhao T, Black SM, Desai AA, Garcia JGN, Sun X, Shyy JY, Valdez-Jasso D, Thistlethwaite PA, Makino A, Wang J, Yuan JX. Endothelial upregulation of mechanosensitive channel Piezo1 in pulmonary hypertension. Am J Physiol Cell Physiol 321: C1010–C1027, 2021. doi: 10.1152/ajpcell.00147.2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Xiao R, Luo S, Zhang T, Lv Y, Wang T, Zhang J, Su Y, Ruiz M, Dupuis J, Zhu L, Hu Q. Peptide blocking self-polymerization of extracellular calcium-sensing receptor attenuates hypoxia-induced pulmonary hypertension. Hypertension 78: 1605–1616, 2021. doi: 10.1161/HYPERTENSIONAHA.120.16712. [DOI] [PubMed] [Google Scholar]
  • 155.Lyle MA, Davis JP, Brozovich FV. Regulation of pulmonary vascular smooth muscle contractility in pulmonary arterial hypertension: implications for therapy. Front Physiol 8: 614, 2017. doi: 10.3389/fphys.2017.00614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Yamamura A, Nayeem MJ, Sato M. The Rho kinase 2 (ROCK2)-specific inhibitor KD025 ameliorates the development of pulmonary arterial hypertension. Biochem Biophys Res Commun 534: 795–801, 2021. doi: 10.1016/j.bbrc.2020.10.106. [DOI] [PubMed] [Google Scholar]
  • 157.Homma N, Nagaoka T, Morio Y, Ota H, Gebb SA, Karoor V, McMurtry IF, Oka M. Endothelin-1 and serotonin are involved in activation of RhoA/Rho kinase signaling in the chronically hypoxic hypertensive rat pulmonary circulation. J Cardiovasc Pharmacol 50: 697–702, 2007. doi: 10.1097/FJC.0b013e3181593774. [DOI] [PubMed] [Google Scholar]
  • 158.Liu P, Huang W, Ding Y, Wu J, Liang Z, Huang Z, Xie W, Kong H. Fasudil dichloroacetate alleviates SU5416/hypoxia-induced pulmonary arterial hypertension by ameliorating dysfunction of pulmonary arterial smooth muscle cells. Drug Des Devel Ther 15: 1653–1666, 2021. doi: 10.2147/DDDT.S297500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Liu Z, Khalil RA. Evolving mechanisms of vascular smooth muscle contraction highlight key targets in vascular disease. Biochem Pharmacol 153: 91–122, 2018. doi: 10.1016/j.bcp.2018.02.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Shi Y, Jiang R, Qin X, Gao A, Hou X, Chen L, Xu X, Guo Y, Chai L, Zhao L, Du X, Wu F. Up-regulation of nPKC contributes to proliferation of mice pulmonary artery smooth muscle cells in hypoxia-induced pulmonary hypertension. Eur J Pharmacol 900: 174046, 2021. doi: 10.1016/j.ejphar.2021.174046. [DOI] [PubMed] [Google Scholar]
  • 161.Mohammed S, Vijayvergiya R, Malhotra S, Rohit MK. A randomized, double-blind, placebo-controlled study to evaluate sildenafil, ambrisentan combination therapy in pulmonary hypertension, particularly of Eisenmenger syndrome. Indian Heart J 73: 633–636, 2021. doi: 10.1016/j.ihj.2021.07.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Chin KM, Sitbon O, Doelberg M, Feldman J, Gibbs JSR, Grünig E, Hoeper MM, Martin N, Mathai SC, McLaughlin VV, Perchenet L, Poch D, Saggar R, Simonneau G, Galiè N. Three- versus two-drug therapy for patients with newly diagnosed pulmonary arterial hypertension. J Am Coll Cardiol 78: 1393–1403, 2021. doi: 10.1016/j.jacc.2021.07.057. [DOI] [PubMed] [Google Scholar]
  • 163.Barst RJ, McGoon M, Torbicki A, Sitbon O, Krowka MJ, Olschewski H, Gaine S. Diagnosis and differential assessment of pulmonary arterial hypertension. J Am Coll Cardiol 43: 40S–47S, 2004. doi: 10.1016/j.jacc.2004.02.032. [DOI] [PubMed] [Google Scholar]
  • 164.McGoon M, Gutterman D, Steen V, Barst R, McCrory DC, Fortin TA, Loyd JE; American College of Chest Physicians. Screening, early detection, and diagnosis of pulmonary arterial hypertension: ACCP evidence-based clinical practice guidelines. Chest 126: 14S–34S, 2004. doi: 10.1378/chest.126.1_suppl.14S. [DOI] [PubMed] [Google Scholar]
  • 165.Miano JM, Fisher EA, Majesky MW. Fate and state of vascular smooth muscle cells in atherosclerosis. Circulation 143: 2110–2116, 2021. doi: 10.1161/CIRCULATIONAHA.120.049922. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Rubin LJ. Therapy of pulmonary hypertension: the evolution from vasodilators to antiproliferative agents. Am J Respir Crit Care Med 166: 1308–1309, 2002. doi: 10.1164/rccm.2208008. [DOI] [PubMed] [Google Scholar]
  • 167.Mitani Y, Ueda M, Komatsu R, Maruyama K, Nagai R, Matsumura M, Sakurai M. Vascular smooth muscle cell phenotypes in primary pulmonary hypertension. Eur Respir J 17: 316–320, 2001. doi: 10.1183/09031936.01.17203160. [DOI] [PubMed] [Google Scholar]
  • 168.Pietra GG, Edwards WD, Kay JM, Rich S, Kernis J, Schloo B, Ayres SM, Bergofsky EH, Brundage BH, Detre KM. Histopathology of primary pulmonary hypertension. A qualitative and quantitative study of pulmonary blood vessels from 58 patients in the National Heart, Lung, and Blood Institute, Primary Pulmonary Hypertension Registry. Circulation 80: 1198–1206, 1989. doi: 10.1161/01.cir.80.5.1198. [DOI] [PubMed] [Google Scholar]
  • 169.Cooper AL, Beasley D. Hypoxia stimulates proliferation and interleukin-1α production in human vascular smooth muscle cells. Am J Physiol Heart Circ Physiol 277: H1326–H1337, 1999. doi: 10.1152/ajpheart.1999.277.4.H1326. [DOI] [PubMed] [Google Scholar]
  • 170.Chen SC, Wang BW, Wang DL, Shyu KG. Hypoxia induces discoidin domain receptor-2 expression via the p38 pathway in vascular smooth muscle cells to increase their migration. Biochem Biophys Res Commun 374: 662–667, 2008. doi: 10.1016/j.bbrc.2008.07.092. [DOI] [PubMed] [Google Scholar]
  • 171.Schultz K, Fanburg BL, Beasley D. Hypoxia and hypoxia-inducible factor-1α promote growth factor-induced proliferation of human vascular smooth muscle cells. Am J Physiol Heart Circ Physiol 290: H2528–H2534, 2006. doi: 10.1152/ajpheart.01077.2005. [DOI] [PubMed] [Google Scholar]
  • 172.Han XJ, Zhang WF, Wang Q, Li M, Zhang CB, Yang ZJ, Tan RJ, Gan LJ, Zhang LL, Lan XM, Zhang FL, Hong T, Jiang LP. HIF-1α promotes the proliferation and migration of pulmonary arterial smooth muscle cells via activation of Cx43. J Cell Mol Med 25: 10663–10673, 2021. doi: 10.1111/jcmm.17003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Xiao G, Lian G, Wang T, Chen W, Zhuang W, Luo L, Wang H, Xie L. Zinc-mediated activation of CREB pathway in proliferation of pulmonary artery smooth muscle cells in pulmonary hypertension. Cell Commun Signal 19: 103, 2021. doi: 10.1186/s12964-021-00779-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Chen DD, Hu WP, Xie L, Xiang GL, Wu QH, Qu JM, Li SQ, Guan LH, Liu D. Serum cartilage oligomeric matrix protein is decreased in patients with pulmonary hypertension: a potential protective factor. Pulm Circ 11: 0271678X20978861, 2021. doi: 10.1177/20458940211031111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Baber SR, Deng W, Master RG, Bunnell BA, Taylor BK, Murthy SN, Hyman AL, Kadowitz PJ. Intratracheal mesenchymal stem cell administration attenuates monocrotaline-induced pulmonary hypertension and endothelial dysfunction. Am J Physiol Heart Circ Physiol 292: H1120–H1128, 2007. doi: 10.1152/ajpheart.00173.2006. [DOI] [PubMed] [Google Scholar]
  • 176.Wang X-X, Zhang F-R, Shang Y-P, Zhu J-H, Xie X-D, Tao Q-M, Zhu J-H, Chen J-Z. Transplantation of autologous endothelial progenitor cells may be beneficial in patients with idiopathic pulmonary arterial hypertension: a pilot randomized controlled trial. J Am Coll Cardiol 49: 1566–1571, 2007. doi: 10.1016/j.jacc.2006.12.037. [DOI] [PubMed] [Google Scholar]
  • 177.Fried ND, Morris TM, Whitehead A, Lazartigues E, Yue X, Gardner JD. Angiotensin II type 1 receptor mediates pulmonary hypertension and right ventricular remodeling induced by inhaled nicotine. Am J Physiol Heart Circ Physiol 320: H1526–H1534, 2021. doi: 10.1152/ajpheart.00883.2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Humbert M, McLaughlin V, Gibbs JSR, Gomberg-Maitland M, Hoeper MM, Preston IR, Souza R, Waxman A, Escribano Subias P, Feldman J, Meyer G, Montani D, Olsson KM, Manimaran S, Barnes J, Linde PG, de Oliveira Pena J, Badesch DB. Sotatercept for the treatment of pulmonary arterial hypertension. N Engl J Med 384: 1204–1215, 2021. doi: 10.1056/NEJMoa2024277. [DOI] [PubMed] [Google Scholar]
  • 179.Dieffenbach PB, Haeger CM, Coronata AMF, Choi KM, Varelas X, Tschumperlin DJ, Fredenburgh LE. Arterial stiffness induces remodeling phenotypes in pulmonary artery smooth muscle cells via YAP/TAZ-mediated repression of cyclooxygenase-2. Am J Physiol Lung Cell Mol Physiol 313: L628–L647, 2017. doi: 10.1152/ajplung.00173.2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Wang L, Chennupati R, Jin YJ, Li R, Wang S, Gunther S, Offermanns S. YAP/TAZ are required to suppress osteogenic differentiation of vascular smooth muscle cells. iScience 23: 101860, 2020. doi: 10.1016/j.isci.2020.101860. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Burg ED, Remillard CV, Yuan JX. Potassium channels in the regulation of pulmonary artery smooth muscle cell proliferation and apoptosis: pharmacotherapeutic implications. Br J Pharmacol 153, Suppl 1: S99–S111, 2008. doi: 10.1038/sj.bjp.0707635. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Antigny F, Hautefort A, Meloche J, Belacel-Ouari M, Manoury B, Rucker-Martin C, Pechoux C, Potus F, Nadeau V, Tremblay E, Ruffenach G, Bourgeois A, Dorfmüller P, Breuils-Bonnet S, Fadel E, Ranchoux B, Jourdon P, Girerd B, Montani D, Provencher S, Bonnet S, Simonneau G, Humbert M, Perros F. Potassium channel subfamily K member 3 (KCNK3) contributes to the development of pulmonary arterial hypertension. Circulation 133: 1371–1385, 2016. doi: 10.1161/CIRCULATIONAHA.115.020951. [DOI] [PubMed] [Google Scholar]
  • 183.Brevnova EE, Platoshyn O, Zhang S, Yuan JX. Overexpression of human KCNA5 increases IK V and enhances apoptosis. Am J Physiol Cell Physiol 287: C715–C722, 2004. doi: 10.1152/ajpcell.00050.2004. [DOI] [PubMed] [Google Scholar]
  • 184.Shults NV, Rybka V, Suzuki YJ, Brelidze TI. Increased smooth muscle Kv11.1 channel expression in pulmonary hypertension and protective role of Kv11.1 channel blocker dofetilide. Am J Pathol 190: 48–56, 2020. doi: 10.1016/j.ajpath.2019.09.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Christou H, Reslan OM, Mam V, Tanbe AF, Vitali SH, Touma M, Arons E, Mitsialis SA, Kourembanas S, Khalil RA. Improved pulmonary vascular reactivity and decreased hypertrophic remodeling during nonhypercapnic acidosis in experimental pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 302: L875–L890, 2012. doi: 10.1152/ajplung.00293.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Christou H, Bailey N, Kluger MS, Mitsialis SA, Kourembanas S. Extracellular acidosis induces heme oxygenase-1 expression in vascular smooth muscle cells. Am J Physiol Heart Circ Physiol 288: H2647–H2652, 2005. doi: 10.1152/ajpheart.00937.2004. [DOI] [PubMed] [Google Scholar]
  • 187.Guan J, Wu X, Arons E, Christou H. The p38 mitogen-activated protein kinase pathway is involved in the regulation of heme oxygenase-1 by acidic extracellular pH in aortic smooth muscle cells. J Cell Biochem 105: 1298–1306, 2008. doi: 10.1002/jcb.21930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Brenninkmeijer L, Kuehl C, Geldart AM, Arons E, Christou H. Heme oxygenase-1 does not mediate the effects of extracellular acidosis on vascular smooth muscle cell proliferation, migration, and susceptibility to apoptosis. J Vasc Res 48: 285–296, 2011. doi: 10.1159/000321555. [DOI] [PubMed] [Google Scholar]
  • 189.Henry EK, Sy CB, Inclan-Rico JM, Espinosa V, Ghanny SS, Dwyer DF, Soteropoulos P, Rivera A, Siracusa MC. Carbonic anhydrase enzymes regulate mast cell-mediated inflammation. J Exp Med 213: 1663–1673, 2016. doi: 10.1084/jem.20151739. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Supuran CT. Carbonic anhydrases: novel therapeutic applications for inhibitors and activators. Nat Rev Drug Discov 7: 168–181, 2008. doi: 10.1038/nrd2467. [DOI] [PubMed] [Google Scholar]
  • 191.Lichtblau M, Berlier C, Saxer S, Carta AF, Mayer L, Groth A, Bader PR, Schneider SR, Furian M, Schwarz EI, Swenson ER, Bloch KE, Ulrich S. Acute hemodynamic effect of acetazolamide in patients with pulmonary hypertension whilst breathing normoxic and hypoxic gas: a randomized cross-over trial. Front Med (Lausanne) 8: 681473, 2021. doi: 10.3389/fmed.2021.681473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Mokhtari RB, Kumar S, Islam SS, Yazdanpanah M, Adeli K, Cutz E, Yeger H. Combination of carbonic anhydrase inhibitor, acetazolamide, and sulforaphane, reduces the viability and growth of bronchial carcinoid cell lines. BMC Cancer 13: 378, 2013. doi: 10.1186/1471-2407-13-378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Pichon A, Connes P, Quidu P, Marchant D, Brunet J, Levy BI, Vilar J, Safeukui I, Cymbalista F, Maignan M, Richalet JP, Favret F. Acetazolamide and chronic hypoxia: effects on haemorheology and pulmonary haemodynamics. Eur Respir J 40: 1401–1409, 2012. doi: 10.1183/09031936.00216011. [DOI] [PubMed] [Google Scholar]
  • 194.Hudalla H, Michael Z, Christodoulou N, Willis GR, Fernandez-Gonzalez A, Filatava EJ, Dieffenbach P, Fredenburgh LE, Stearman RS, Geraci MW, Kourembanas S, Christou H. Carbonic anhydrase inhibition ameliorates inflammation and experimental pulmonary hypertension. Am J Respir Cell Mol Biol 61: 512–524, 2019. doi: 10.1165/rcmb.2018-0232OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Sharmin N, Nganwuchu CC, Nasim MT. Targeting the TGF-β signaling pathway for resolution of pulmonary arterial hypertension. Trends Pharmacol Sci 42: 510–513, 2021. doi: 10.1016/j.tips.2021.04.002. [DOI] [PubMed] [Google Scholar]
  • 196.Tran HB, Maiolo S, Harper R, Zalewski PD, Reynolds PN, Hodge S. Dysregulated zinc and sphingosine-1-phosphate signaling in pulmonary hypertension: potential effects by targeting of bone morphogenetic protein receptor type 2 in pulmonary microvessels. Cell Biol Int 45: 2368–2379, 2021. doi: 10.1002/cbin.11682. [DOI] [PubMed] [Google Scholar]
  • 197.Woo KV, Shen IY, Weinheimer CJ, Kovacs A, Nigro J, Lin CY, Chakinala M, Byers DE, Ornitz DM. Endothelial FGF signaling is protective in hypoxia-induced pulmonary hypertension. J Clin Invest 131: e141467, 2021. doi: 10.1172/JCI141467. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Ambade AS, Hassoun PM, Damico RL. Basement membrane extracellular matrix proteins in pulmonary vascular and right ventricular remodeling in pulmonary hypertension. Am J Respir Cell Mol Biol 65: 245–258, 2021. doi: 10.1165/rcmb.2021-0091TR. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Sanz J, Kariisa M, Dellegrottaglie S, Prat-González S, Garcia MJ, Fuster V, Rajagopalan S. Evaluation of pulmonary artery stiffness in pulmonary hypertension with cardiac magnetic resonance. JACC Cardiovasc Imaging 2: 286–295, 2009. doi: 10.1016/j.jcmg.2008.08.007. [DOI] [PubMed] [Google Scholar]
  • 200.Dieffenbach PB, Mallarino Haeger C, Rehman R, Corcoran AM, Coronata AMF, Vellarikkal SK, Chrobak I, Waxman AB, Vitali SH, Sholl LM, Padera RF, Lagares D, Polverino F, Owen CA, Fredenburgh LE. A novel protective role for matrix metalloproteinase-8 in the pulmonary vasculature. Am J Respir Crit Care Med 204: 1433–1451, 2021. doi: 10.1164/rccm.202108-1863OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Pang W, Zhang Z, Zhang Y, Zhang M, Miao R, Yang Y, Xie W, Wan J, Zhai Z, Wang C. Extracellular matrix collagen biomarkers levels in patients with chronic thromboembolic pulmonary hypertension. J Thromb Thrombolysis 52: 48–58, 2021. doi: 10.1007/s11239-020-02329-8. [DOI] [PubMed] [Google Scholar]
  • 202.Lammers SR, Kao PH, Qi HJ, Hunter K, Lanning C, Albietz J, Hofmeister S, Mecham R, Stenmark KR, Shandas R. Changes in the structure-function relationship of elastin and its impact on the proximal pulmonary arterial mechanics of hypertensive calves. Am J Physiol Heart Circ Physiol 295: H1451–H1459, 2008. doi: 10.1152/ajpheart.00127.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Wang R, Xu J, Wu J, Gao S, Wang Z. Angiotensin-converting enzyme 2 alleviates pulmonary artery hypertension through inhibition of focal adhesion kinase expression. Exp Ther Med 22: 1165, 2021. doi: 10.3892/etm.2021.10599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Barman SA, Bordan Z, Batori R, Haigh S, Fulton DJR. Galectin-3 promotes ROS, inflammation, and vascular fibrosis in pulmonary arterial hypertension. Adv Exp Med Biol 1303: 13–32, 2021. doi: 10.1007/978-3-030-63046-1_2. [DOI] [PubMed] [Google Scholar]
  • 205.Yuan K, Agarwal S, Chakraborty A, Condon DF, Patel H, Zhang S, Huang F, Mello SA, Kirk OI, Vasquez R, de Jesus Perez VA. Lung pericytes in pulmonary vascular physiology and pathophysiology. Compr Physiol 11: 2227–2247, 2021. doi: 10.1002/cphy.c200027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Sheikh AQ, Misra A, Rosas IO, Adams RH, Greif DM. Smooth muscle cell progenitors are primed to muscularize in pulmonary hypertension. Sci Transl Med 7: 308ra159, 2015. doi: 10.1126/scitranslmed.aaa9712. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Steffes LC, Froistad AA, Andruska A, Boehm M, McGlynn M, Zhang F, Zhang W, Hou D, Tian X, Miquerol L, Nadeau K, Metzger RJ, Spiekerkoetter E, Kumar ME. A Notch3-marked subpopulation of vascular smooth muscle cells is the cell of origin for occlusive pulmonary vascular lesions. Circulation 142: 1545–1561, 2020. doi: 10.1161/CIRCULATIONAHA.120.045750. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Dierick F, Solinc J, Bignard J, Soubrier F, Nadaud S. Progenitor/stem cells in vascular remodeling during pulmonary arterial hypertension. Cells 10: 1338, 2021. doi: 10.3390/cells10061338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Xu J, Li X, Zhou S, Wang R, Wu M, Tan C, Chen J, Wang Z. Inhibition of CXCR4 ameliorates hypoxia-induced pulmonary arterial hypertension in rats. Am J Transl Res 13: 1458–1470, 2021. [PMC free article] [PubMed] [Google Scholar]
  • 210.Pugliese SC, Poth JM, Fini MA, Olschewski A, El Kasmi KC, Stenmark KR. The role of inflammation in hypoxic pulmonary hypertension: from cellular mechanisms to clinical phenotypes. Am J Physiol Lung Cell Mol Physiol 308: L229–L252, 2015. doi: 10.1152/ajplung.00238.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.Rabinovitch M, Guignabert C, Humbert M, Nicolls MR. Inflammation and immunity in the pathogenesis of pulmonary arterial hypertension. Circ Res 115: 165–175, 2014. doi: 10.1161/CIRCRESAHA.113.301141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Savai R, Pullamsetti SS, Kolbe J, Bieniek E, Voswinckel R, Fink L, Scheed A, Ritter C, Dahal BK, Vater A, Klussmann S, Ghofrani HA, Weissmann N, Klepetko W, Banat GA, Seeger W, Grimminger F, Schermuly RT. Immune and inflammatory cell involvement in the pathology of idiopathic pulmonary arterial hypertension. Am J Respir Crit Care Med 186: 897–908, 2012. doi: 10.1164/rccm.201202-0335OC. [DOI] [PubMed] [Google Scholar]
  • 213.Soon E, Holmes AM, Treacy CM, Doughty NJ, Southgate L, Machado RD, Trembath RC, Jennings S, Barker L, Nicklin P, Walker C, Budd DC, Pepke-Zaba J, Morrell NW. Elevated levels of inflammatory cytokines predict survival in idiopathic and familial pulmonary arterial hypertension. Circulation 122: 920–927, 2010. doi: 10.1161/CIRCULATIONAHA.109.933762. [DOI] [PubMed] [Google Scholar]
  • 214.Vergadi E, Chang MS, Lee C, Liang OD, Liu X, Fernandez-Gonzalez A, Mitsialis SA, Kourembanas S. Early macrophage recruitment and alternative activation are critical for the later development of hypoxia-induced pulmonary hypertension. Circulation 123: 1986–1995, 2011. doi: 10.1161/CIRCULATIONAHA.110.978627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Minamino T, Christou H, Hsieh CM, Liu Y, Dhawan V, Abraham NG, Perrella MA, Mitsialis SA, Kourembanas S. Targeted expression of heme oxygenase-1 prevents the pulmonary inflammatory and vascular responses to hypoxia. Proc Natl Acad Sci USA 98: 8798–8803, 2001. doi: 10.1073/pnas.161272598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216.Groth A, Vrugt B, Brock M, Speich R, Ulrich S, Huber LC. Inflammatory cytokines in pulmonary hypertension. Respir Res 15: 47, 2014. doi: 10.1186/1465-9921-15-47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.Stacher E, Graham BB, Hunt JM, Gandjeva A, Groshong SD, McLaughlin VV, Jessup M, Grizzle WE, Aldred MA, Cool CD, Tuder RM. Modern age pathology of pulmonary arterial hypertension. Am J Respir Crit Care Med 186: 261–272, 2012. doi: 10.1164/rccm.201201-0164OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.Frid MG, Brunetti JA, Burke DL, Carpenter TC, Davie NJ, Reeves JT, Roedersheimer MT, van Rooijen N, Stenmark KR. Hypoxia-induced pulmonary vascular remodeling requires recruitment of circulating mesenchymal precursors of a monocyte/macrophage lineage. Am J Pathol 168: 659–669, 2006. doi: 10.2353/ajpath.2006.050599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Fan Y, Hao Y, Gao D, Li G, Zhang Z. Phenotype and function of macrophage polarization in monocrotaline-induced pulmonary arterial hypertension rat model. Physiol Res 70: 213–226, 2021. doi: 10.33549/physiolres.934456. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Bellan M, Piccinino C, Tonello S, Minisini R, Giubertoni A, Sola D, Pedrazzoli R, Gagliardi I, Zecca E, Calzaducca E, Mazzoleni F, Piffero R, Patti G, Pirisi M, Sainaghi PP. Role of osteopontin as a potential biomarker of pulmonary arterial hypertension in patients with systemic sclerosis and other connective tissue diseases (CTDs). Pharmaceuticals (Basel) 14: 394, 2021. doi: 10.3390/ph14050394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221.Hurst LA, Dunmore BJ, Long L, Crosby A, Al-Lamki R, Deighton J, Southwood M, Yang X, Nikolic MZ, Herrera B, Inman GJ, Bradley JR, Rana AA, Upton PD, Morrell NW. TNFα drives pulmonary arterial hypertension by suppressing the BMP type-II receptor and altering NOTCH signalling. Nat Commun 8: 14079, 2017. doi: 10.1038/ncomms14079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.Hashimoto-Kataoka T, Hosen N, Sonobe T, Arita Y, Yasui T, Masaki T, Minami M, Inagaki T, Miyagawa S, Sawa Y, Murakami M, Kumanogoh A, Yamauchi-Takihara K, Okumura M, Kishimoto T, Komuro I, Shirai M, Sakata Y, Nakaoka Y. Interleukin-6/interleukin-21 signaling axis is critical in the pathogenesis of pulmonary arterial hypertension. Proc Natl Acad Sci USA 112: E2677–E2686, 2015. doi: 10.1073/pnas.1424774112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223.Otsuki S, Sawada H, Yodoya N, Shinohara T, Kato T, Ohashi H, Zhang E, Imanaka-Yoshida K, Shimpo H, Maruyama K, Komada Y, Mitani Y. Potential contribution of phenotypically modulated smooth muscle cells and related inflammation in the development of experimental obstructive pulmonary vasculopathy in rats. PLoS One 10: e0118655, 2015. doi: 10.1371/journal.pone.0118655. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Ali MS, Starke RM, Jabbour PM, Tjoumakaris SI, Gonzalez LF, Rosenwasser RH, Owens GK, Koch WJ, Greig NH, Dumont AS. TNF-α induces phenotypic modulation in cerebral vascular smooth muscle cells: implications for cerebral aneurysm pathology. J Cereb Blood Flow Metab 33: 1564–1573, 2013. doi: 10.1038/jcbfm.2013.109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225.Li P, Li YL, Li ZY, Wu YN, Zhang CC, A X, Wang CX, Shi HT, Hui MZ, Xie B, Ahmed M, Du J. Cross talk between vascular smooth muscle cells and monocytes through interleukin-1β/interleukin-18 signaling promotes vein graft thickening. Arterioscler Thromb Vasc Biol 34: 2001–2011, 2014. doi: 10.1161/ATVBAHA.113.303145. [DOI] [PubMed] [Google Scholar]
  • 226.Tian W, Jiang X, Tamosiuniene R, Sung YK, Qian J, Dhillon G, Gera L, Farkas L, Rabinovitch M, Zamanian RT, Inayathullah M, Fridlib M, Rajadas J, Peters-Golden M, Voelkel NF, Nicolls MR. Blocking macrophage leukotriene b4 prevents endothelial injury and reverses pulmonary hypertension. Sci Transl Med 5: 200ra117, 2013. doi: 10.1126/scitranslmed.3006674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227.Daley E, Emson C, Guignabert C, de Waal Malefyt R, Louten J, Kurup VP, Hogaboam C, Taraseviciene-Stewart L, Voelkel NF, Rabinovitch M, Grunig E, Grunig G. Pulmonary arterial remodeling induced by a Th2 immune response. J Exp Med 205: 361–372, 2008. doi: 10.1084/jem.20071008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228.Tian W, Jiang SY, Jiang X, Tamosiuniene R, Kim D, Guan T, Arsalane S, Pasupneti S, Voelkel NF, Tang Q, Nicolls MR. The role of regulatory t cells in pulmonary arterial hypertension. Front Immunol 12: 684657, 2021. doi: 10.3389/fimmu.2021.684657. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.Tamosiuniene R, Manouvakhova O, Mesange P, Saito T, Qian J, Sanyal M, Lin YC, Nguyen LP, Luria A, Tu AB, Sante JM, Rabinovitch M, Fitzgerald DJ, Graham BB, Habtezion A, Voelkel NF, Aurelian L, Nicolls MR. Dominant role for regulatory T cells in protecting females against pulmonary hypertension. Circ Res 122: 1689–1702, 2018. doi: 10.1161/CIRCRESAHA.117.312058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.Nicolls MR, Voelkel NF. The roles of immunity in the prevention and evolution of pulmonary arterial hypertension. Am J Respir Crit Care Med 195: 1292–1299, 2017. doi: 10.1164/rccm.201608-1630PP. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231.Rampa DR, Murugesan P, Chao H, Feng H, Dai W, Lee D, Pekcec A, Doods H, Wu D. Reversal of pulmonary arterial hypertension and neointimal formation by kinin B1 receptor blockade. Respir Res 22: 281, 2021. doi: 10.1186/s12931-021-01875-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Liu G, Wan N, Liu Q, Chen Y, Cui H, Wang Y, Ren J, Shen X, Lu W, Yu Y, Shen Y, Wang J. Resolvin E1 attenuates pulmonary hypertension by suppressing Wnt7a/β-catenin signaling. Hypertension 78: 1914–1926, 2021. doi: 10.1161/HYPERTENSIONAHA.121.17809. [DOI] [PubMed] [Google Scholar]
  • 233.Youm YH, Nguyen KY, Grant RW, Goldberg EL, Bodogai M, Kim D, D'Agostino D, Planavsky N, Lupfer C, Kanneganti TD, Kang S, Horvath TL, Fahmy TM, Crawford PA, Biragyn A, Alnemri E, Dixit VD. The ketone metabolite β-hydroxybutyrate blocks NLRP3 inflammasome-mediated inflammatory disease. Nat Med 21: 263–269, 2015. doi: 10.1038/nm.3804. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234.Colegio OR, Chu NQ, Szabo AL, Chu T, Rhebergen AM, Jairam V, Cyrus N, Brokowski CE, Eisenbarth SC, Phillips GM, Cline GW, Phillips AJ, Medzhitov R. Functional polarization of tumour-associated macrophages by tumour-derived lactic acid. Nature 513: 559–563, 2014. doi: 10.1038/nature13490. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 235.Mogi C, Tobo M, Tomura H, Murata N, He XD, Sato K, Kimura T, Ishizuka T, Sasaki T, Sato T, Kihara Y, Ishii S, Harada A, Okajima F. Involvement of proton-sensing TDAG8 in extracellular acidification-induced inhibition of proinflammatory cytokine production in peritoneal macrophages. J Immunol 182: 3243–3251, 2009. doi: 10.4049/jimmunol.0803466. [DOI] [PubMed] [Google Scholar]
  • 236.Fließer E, Birnhuber A, Marsh LM, Gschwandtner E, Klepetko W, Olschewski H, Kwapiszewska G. Dysbalance of ACE2 levels—a possible cause for severe COVID-19 outcome in COPD. J Pathol Clin Res 7: 446–458, 2021. doi: 10.1002/cjp2.224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237.Cueto-Robledo G, Porres-Aguilar M, Puebla-Aldama D, Barragán-Martínez MDP, Jurado-Hernandez MY, García-César M, Rojas MBT, García-Treminio C, Roldan-Valadez E. Severe pulmonary hypertension: an important sequel after severe post-acute COVID-19 pneumonia. Curr Probl Cardiol 47: 101004, 2022. doi: 10.1016/j.cpcardiol.2021.101004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238.Suzuki YJ, Nikolaienko SI, Shults NV, Gychka SG. COVID-19 patients may become predisposed to pulmonary arterial hypertension. Med Hypotheses 147: 110483, 2021. doi: 10.1016/j.mehy.2021.110483. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 239.Vlachou M, Drebes A, Candilio L, Weeraman D, Mir N, Murch N, Davies N, Coghlan JG. Pulmonary thrombosis in Covid-19: before, during and after hospital admission. J Thromb Thrombolysis 51: 978–984, 2021. doi: 10.1007/s11239-020-02370-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240.Deniz S, Uysal TK, Capasso C, Supuran CT, Ozensoy Guler O. Is carbonic anhydrase inhibition useful as a complementary therapy of Covid-19 infection? J Enzyme Inhib Med Chem 36: 1230–1235, 2021. doi: 10.1080/14756366.2021.1924165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241.Smith AM, Jones RD, Channer KS. The influence of sex hormones on pulmonary vascular reactivity: possible vasodilator therapies for the treatment of pulmonary hypertension. Curr Vasc Pharmacol 4: 9–15, 2006. doi: 10.2174/157016106775203090. [DOI] [PubMed] [Google Scholar]

Articles from American Journal of Physiology - Heart and Circulatory Physiology are provided here courtesy of American Physiological Society

RESOURCES