+ All Categories
Home > Documents > Role of Rho-kinase and its inhibitors in pulmonary hypertension

Role of Rho-kinase and its inhibitors in pulmonary hypertension

Date post: 11-Dec-2016
Category:
Upload: anh-tuan
View: 216 times
Download: 3 times
Share this document with a friend
13
Associate Editor: M.G. Belvisi Role of Rho-kinase and its inhibitors in pulmonary hypertension Sy Duong-Quy a , Yihua Bei a, b , Zhongmin Liu b , Anh Tuan Dinh-Xuan a, b, a Paris Descartes University, Medical School, Assistance Publique Hôpitaux de Paris, Service de Physiologie, Explorations Fonctionnelles. Hôpital Cochin, 27 rue du faubourg Saint-Jacques, 75014 Paris, France b Clinical and Translational Research Center, Tongji University School of Medicine and Shanghai East Hospital, 150 Jimo Road, Shanghai, 200120, China abstract article info Keywords: RhoA Rho-kinase Pulmonary hypertension Pulmonary hypertension (PH) is an incurable disease with a dreadful survival rate. The disease is characterized by sustained vasoconstriction, progressive vascular remodeling, and irreversible right heart dysfunction. While hypoxic pulmonary vasoconstriction (HPV) is known to be the main pathophysiological factor causing the rise in pulmonary arterial pressure, biological mechanisms leading to HPV and vascular remodeling are multiple and complex and, as yet, incompletely understood. It is thought that molecular interactions and cross talks are involved in the pathogenesis of PH, perturbing the physiological balance between substances controlling vascu- lar tone, cell growth and apoptosis. This balance is achieved by subtle interactions between factors acting as both vasodilators and inhibitors of cell growth like nitric oxide, prostacyclin, vasoactive intestinal peptide and mole- cules with potent vasoconstrictor and cell growth activities like endothelin-1. Recent in vivo studies showed that the Rho GTPase/RhoA pathway and its downstream effectors, the Rho-kinases (ROCK-1 and ROCK-2), had an im- portant role in PH, due to its lasting effects on vasoconstriction and pulmonary cell proliferation leading to vas- cular remodeling. Benecial effects obtained in vivo with Rho-kinase inhibitors (e.g.Y-27632 and fasudil) in experimental PH will hopefully lead to future clinical trials with new compounds selectively targeting this path- way, which is now proven to be detrimental when over-activated in both experimental animals and human patients. © 2012 Elsevier Inc. All rights reserved. Contents 1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 353 2. Overview of pulmonary hypertension . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 353 3. Rho-kinase signaling pathway . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 355 4. Vascular downstream effect of Rho-kinase . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 356 5. Role of Rho-kinase signaling in pulmonary hypertension . . . . . . . . . . . . . . . . . . . . . . . . . 358 6. Rho-kinase inhibitors in the treatment of pulmonary hypertension . . . . . . . . . . . . . . . . . . . . . 359 7. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 361 Conict of interest statement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 361 References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 361 Pharmacology & Therapeutics 137 (2013) 352364 Abbreviations: 5-HT, serotonin; AA, arachidonic acid; AC, adenylate cyclase; Ang II, angiotensin II; ATP, adenosine triphosphate; BMP, bone morphogenetic proteins; CaM, cal- modulin; cAMP, cyclic adenosine monophosphate; cGMP, cyclic guanosine monophosphate; CH, chronic hypoxia; COPD, chronic obstructive pulmonary disease; EC, endothelial cell; ECE, endothelin converting enzyme; EGF, epidermal growth factor; eNOS or NOS-3, endothelial nitric oxide synthase; ERK, extracellular signal-regulated kinases; ET-1, endothelin-1; GDP, guanosine diphosphate; GTP, guanosine triphosphate; MAPK, mitogen-activated protein kinases; MCT, monocrotaline; MLC, myosin light chain; MLCK, myosin light chain kinase; MLCP, myosin light chain phosphatase; NE, norepinephrin; NO, nitric oxide; PAH, pulmonary arterial hypertension; PAP, pulmonary arterial pressure; PDGF, platelet-derived growth factor; PH, Pleckstrin-homology; PH, pulmonary hypertension; PIP 2 , phosphatidyl-inositol-diphosphate; PKC, protein kinase C; PLC, phospholipase C; PVR, pulmonary vascular resistance; RBD, Rho-binding domain; Rho-GAP, Rho GTPase activating protein; Rho-GEF, Rho guanine nucleotide exchange factor; ROCK, Rho-kinases; ROS, reactive oxygen species; RV, right ventricle; RVSP, right ventricle systolic pressure; SERT, serotonin transporter; sGC, soluble guanylate cyclase; SMC, smooth muscle cell; SPC, sphingosine phosphorylcholine; TXA 2 , thromboxane A 2 ; VEGF, vascular endothelial growth factor. Corresponding author at: Service de Physiologie, Explorations Fonctionnelles, Hôpital Cochin Hospital, Université Paris Descartes, 27, rue du faubourg Saint-Jacques, 75104 Paris, France. Tel.: +33 158412341; fax: +33 158412345. E-mail address: [email protected] (A.T. Dinh-Xuan). 0163-7258/$ see front matter © 2012 Elsevier Inc. All rights reserved. http://dx.doi.org/10.1016/j.pharmthera.2012.12.003 Contents lists available at SciVerse ScienceDirect Pharmacology & Therapeutics journal homepage: www.elsevier.com/locate/pharmthera
Transcript

Pharmacology & Therapeutics 137 (2013) 352–364

Contents lists available at SciVerse ScienceDirect

Pharmacology & Therapeutics

j ou rna l homepage: www.e lsev ie r .com/ locate /pharmthera

Associate Editor: M.G. Belvisi

Role of Rho-kinase and its inhibitors in pulmonary hypertension

Sy Duong-Quy a, Yihua Bei a,b, Zhongmin Liu b, Anh Tuan Dinh-Xuan a,b,⁎a Paris Descartes University, Medical School, Assistance Publique Hôpitaux de Paris, Service de Physiologie, Explorations Fonctionnelles. Hôpital Cochin, 27 rue du faubourg Saint-Jacques,75014 Paris, Franceb Clinical and Translational Research Center, Tongji University School of Medicine and Shanghai East Hospital, 150 Jimo Road, Shanghai, 200120, China

Abbreviations: 5-HT, serotonin; AA, arachidonic acidmodulin; cAMP, cyclic adenosine monophosphate; cGMcell; ECE, endothelin converting enzyme; EGF, epidermendothelin-1; GDP, guanosine diphosphate; GTP, guanolight chain kinase; MLCP, myosin light chain phosphataplatelet-derived growth factor; PH, Pleckstrin-homologPVR, pulmonary vascular resistance; RBD, Rho-bindingROS, reactive oxygen species; RV, right ventricle; RVSPSPC, sphingosine phosphorylcholine; TXA2, thromboxan⁎ Corresponding author at: Service de Physiologie, Explo

Tel.: +33 158412341; fax: +33 158412345.E-mail address: [email protected] (A

0163-7258/$ – see front matter © 2012 Elsevier Inc. Allhttp://dx.doi.org/10.1016/j.pharmthera.2012.12.003

a b s t r a c t

a r t i c l e i n f o

Keywords:

RhoARho-kinasePulmonary hypertension

Pulmonary hypertension (PH) is an incurable disease with a dreadful survival rate. The disease is characterizedby sustained vasoconstriction, progressive vascular remodeling, and irreversible right heart dysfunction. Whilehypoxic pulmonary vasoconstriction (HPV) is known to be the main pathophysiological factor causing the risein pulmonary arterial pressure, biological mechanisms leading to HPV and vascular remodeling are multipleand complex and, as yet, incompletely understood. It is thought that molecular interactions and cross talks areinvolved in the pathogenesis of PH, perturbing the physiological balance between substances controlling vascu-lar tone, cell growth and apoptosis. This balance is achieved by subtle interactions between factors acting as bothvasodilators and inhibitors of cell growth like nitric oxide, prostacyclin, vasoactive intestinal peptide and mole-culeswith potent vasoconstrictor and cell growth activities like endothelin-1. Recent in vivo studies showed thatthe Rho GTPase/RhoA pathway and its downstream effectors, the Rho-kinases (ROCK-1 and ROCK-2), had an im-portant role in PH, due to its lasting effects on vasoconstriction and pulmonary cell proliferation leading to vas-cular remodeling. Beneficial effects obtained in vivo with Rho-kinase inhibitors (e.g.Y-27632 and fasudil) inexperimental PHwill hopefully lead to future clinical trials with new compounds selectively targeting this path-way, which is now proven to be detrimental when over-activated in both experimental animals and humanpatients.

© 2012 Elsevier Inc. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3532. Overview of pulmonary hypertension . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3533. Rho-kinase signaling pathway . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3554. Vascular downstream effect of Rho-kinase . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3565. Role of Rho-kinase signaling in pulmonary hypertension . . . . . . . . . . . . . . . . . . . . . . . . . 3586. Rho-kinase inhibitors in the treatment of pulmonary hypertension . . . . . . . . . . . . . . . . . . . . . 3597. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 361Conflict of interest statement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 361

References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 361

; AC, adenylate cyclase; Ang II, angiotensin II; ATP, adenosine triphosphate; BMP, bone morphogenetic proteins; CaM, cal-P, cyclic guanosine monophosphate; CH, chronic hypoxia; COPD, chronic obstructive pulmonary disease; EC, endothelialal growth factor; eNOS or NOS-3, endothelial nitric oxide synthase; ERK, extracellular signal-regulated kinases; ET-1,

sine triphosphate; MAPK, mitogen-activated protein kinases; MCT, monocrotaline; MLC, myosin light chain; MLCK, myosinse; NE, norepinephrin; NO, nitric oxide; PAH, pulmonary arterial hypertension; PAP, pulmonary arterial pressure; PDGF,y; PH, pulmonary hypertension; PIP2, phosphatidyl-inositol-diphosphate; PKC, protein kinase C; PLC, phospholipase C;domain; Rho-GAP, Rho GTPase activating protein; Rho-GEF, Rho guanine nucleotide exchange factor; ROCK, Rho-kinases;, right ventricle systolic pressure; SERT, serotonin transporter; sGC, soluble guanylate cyclase; SMC, smooth muscle cell;e A2; VEGF, vascular endothelial growth factor.rations Fonctionnelles, Hôpital CochinHospital, Université ParisDescartes, 27, rue du faubourg Saint-Jacques, 75104Paris, France.

.T. Dinh-Xuan).

rights reserved.

353S. Duong-Quy et al. / Pharmacology & Therapeutics 137 (2013) 352–364

1. Introduction

Pulmonary hypertension (PH) is a rare disorder with a dreadful prog-nosis. Despite substantial advances in the knowledge of pathogenesis andtherapy during the last decade, PH still remains an incurable disease. Inthe 90s, the median survival rate of untreated patients was around2.8 years from diagnosis (D'Alonzo et al., 1991). PH is defined by an in-crease in mean pulmonary arterial pressure (PAP) ≥25 mm Hg at rest,a pulmonary wedge pressure (PWP) ≤15 mm Hg and a normal or re-duced cardiac output, assessed by right heart catheterization (Galiè etal., 2009). PH is currently classified into five groups and one subgroupaccording to pathological, clinical, and therapeutic features (Galiè et al.,2009).

Current treatment of PH consists of the use of conventional therapy incombination with specific treatments with continuous prostacyclin infu-sion or inhalation, oral phosphodiesterase-5 inhibitors (Olschewski etal., 2002; Sitbon et al., 2002; Sastry et al., 2004) and oral endothelin-1 re-ceptor antagonists (Channick et al., 2001; Rubin et al., 2002; Naeije &Huez, 2007). None of these drugs, however, can be considered as an opti-mal treatment for PH as theymainly act as vasodilators and lack inhibito-ry effects on remodeling of the pulmonary vasculature. There is thereforea need for new treatments to be found and tested by randomized clinicaltrials (Hoeper & Dinh-Xuan, 2004).

Recently, accumulating evidence showed that RhoA and its down-stream effectors, the Rho-kinases, have a preponderant role in the phys-iopathology of PH due to their potent effects on pulmonary arterialsmooth muscle cell (SMCs) contraction and proliferation (Nagaoka etal., 2006; Homma et al., 2008; Guilluy et al., 2009). This paperwill reviewthe biological role of Rho-kinase signaling pathway and the efficacy ofRho-kinase inhibitors in the treatment of PH, targeting both its vasocon-strictor and vascular remodeling components.

2. Overview of pulmonary hypertension

2.1. Classification of pulmonary hypertension

The first international classification of PH, established in 1973 at theWorld Health Organization (WHO) Symposium held in Evian, catego-rized the disorder as “primary”whenno underlying etiology or risk factorcould be identified, and “secondary”when clearly associated to a clearlyidentified primary disorder (Hatano & Strasser, 1975). Since then, theclinical classification of PH has been revised thrice in 1998 (Evian,France), 2003 (Venice, Italy) (Simonneau et al., 2004), and 2008 (DanaPoint, USA) (Galiè et al., 2009). In these classifications, clinical conditionswith PH were divided into five groups according to pathological, patho-physiological, and therapeutic characteristics.

The latest clinical classification, made in Dana Point in 2008(Galiè et al., 2009) maintains the overall structure of the Evian-Venice classifications while amending some specific points toavoid possible confusion among the terms PH and PAH (pulmonaryarterial hypertension). This new classification of PH is summarizedas follows:

Group 1 (PAH): including idiopathic PAH, heritable PAH (germlinemutations of BMPR2, ALK1 or endoglin genes), drugs and toxins in-duced PAH, PAH associated with other conditions (connective tissuediseases, HIV infection, portal hypertension, congenital heart disease,schistosomiasis, chronic hemolytic anemia), and persistent pulmo-nary hypertension of the newborn.Group1′: PH due to pulmonary veno-occlusive disease and/orpulmonary capillary hemangiomatosis. These conditions havebeen classified as a distinct category but not completely separat-ed from PAH, and designated as Group 1′.Group 2: PH related to left heart disease, including systolic dys-function, diastolic dysfunction, and valvular disorders.

Group 3: PH related to lung disease and/or hypoxia, including chron-ic obstructive pulmonary disease, interstitial lung disease, other pul-monary disease with mixed restrictive and obstructive patterns,sleep-disordered breathing, alveolar hypoventilation disorders,chronic exposure to high altitude, and developmental abnormalities.Group 4: PH due to chronic thromboembolic disease (CTEPH:chronic thromboembolic pulmonary hypertension) without pre-cise criterion to distinguish between proximal and distal forms.Group 5: PH with unclear and/or multifactorial mechanisms, includ-ing heterogeneous conditions with different pathological featuressuch as hematological disorders (myeloproliferative disorders, sple-nectomy), systemic disorders (sarcoidosis, pulmonary Langerhanscell histiocytosis, lymphangioleiomyomatosis, neurofibromatosis,vasculitis), metabolic disorders (glycogen storage disease, Gaucherdisease, thyroid disorders), and others (tumoral obstruction, fibrosingmediastinitis, chronic renal failure on dialysis).

2.2. Physiopathology and pathogenesis of pulmonary hypertension

Pulmonary vasoconstriction is an important feature of all forms ofPH. Decrease of alveolar partial pressure of oxygen results in acutepulmonary vasoconstriction which, when prolonged, will eventuallylead to the occurrence of PH (Sommer et al., 2008). Chronic hypoxicpulmonary vasoconstriction is sustained and worsened by an imbal-ance between vasodilators and vasoconstrictors acting on the pulmo-nary vascular smooth muscle cells. Recent evidence also suggestsdeleterious roles of growth factors (Giaid & Saleh, 1995; Tuder et al.,1999; Galié et al., 2004). Other mechanisms involved in the physiopa-thology of PH include 1/reduced cross-sectional area of pulmonary vas-cular bed due to disorders affecting the lung parenchyma, 2/thrombiobstructing pulmonary vascular lumen (Mahapatra et al., 2006a,2006b), 3/overloaded volume and pressure caused by left-right intra-cardiac shunts, 4/obstruction of pulmonary venousflow, and 5/a combi-nation of the above mechanisms in advanced stage of PH (Ito et al.,2003; Montani et al., 2009).

Whilst the classification of PH is well defined and largely dependson clinical features, the pathogenesis of PH is complex and incom-pletely understood. It is thought that the underlying mechanismsare multiple and lead to both functional and structural changes ofthe pulmonary vasculature (Humbert et al., 2004; Yuan & Rubin,2005).

2.3. Pathology and pathobiology of pulmonary hypertension

Although differentmechanisms are involved in the physiopathologyand pathogenesis of the disease, all forms of PH have common patho-logic features in pulmonary arteries. It is mostly characterized by vascu-lar remodeling (medial hypertrophy, intimal fibrosis, and adventitialthickening), and organized thrombotic (Pietra et al., 2004; Yuan &Rubin, 2005; Tuder et al., 2009). Pulmonary vascular remodeling ischaracterized by the increasing of external diameter of pulmonary ar-teries due to predominantly SMC hypertrophy and extracellular matrixand collagen deposit (Palevsky et al., 1989; Yi et al., 2000). However, thecorrelation betweenmorphological changes, degree of disease severity,and potential response to vasodilators is not clearly established in PH(Tuder & Zaiman, 2004).

The pathobiology of PH is very complex andmultifactorial (Humbertet al., 2004; Hassoun et al., 2009; Morrell et al., 2009) (Fig. 1). Manymediator and signaling pathway with its downstream effectors areimplicated in the pathobiology of PH. It is involved in the regulation ofpulmonary vasoactivity, endothelial function, SMC proliferation, andvascular inflammation or thrombosis. Impairment of vasoconstriction–vasodilatation balance is the earliest disorder occurring in the pathogen-esis of PAH (Christman et al., 1992; Giaid & Saleh, 1995). Endothelial

Fig. 1. Potential mechanisms involved in the development of PH. Top half: Relaxation of smooth muscle cells induced by NO, prostacyclin, and VIP is mediated by cGMP and cAMPwhereas constriction induced by TXA2, ET-1 and 5-HT is mediated by membrane receptors coupled to G proteins and intracellular calcium modifiers such as IP3 (inositol triphos-phate) or mediators of RhoA/Rho-kinase pathways. Bottom half: Proliferation of smooth muscle cells is induced by cytokines, growth factors and TGF-β. Control of transcription isregulated by the BMP proteins and its membrane receptors via Smad signaling effectors, and MAP kinase pathways. Abbreviations: 5-HT1B/2A, serotonin receptors 1B or 2A; BMPR2,bone morphogenetic protein receptor 2; ETA/B, endothelin receptors A or B; GF, growth factors; GPCR, G protein-coupled receptors; HHV-8, human herpesvirus 8; JNK, c-JunN-terminal kinase; Kv, potassium voltage-gated channels; P, phosphorylation; PGH2, prostaglandin H2; PGI2, prostaglandine I2 (prostacyclin); PGI2-R, prostacyclin receptors;R-TXA2, thromboxane receptors; VPAC-1/2, vasoactive intestinal peptide receptors 1 or 2; TGF-β, transforming growth factor-β; TPH1, tryptophan hydroxylase 1; VIP, vasoactiveintestinal peptide.

354 S. Duong-Quy et al. / Pharmacology & Therapeutics 137 (2013) 352–364

dysfunction leads to chronically impaired production of vasodilator andanti-proliferative agents such as NO and prostacyclin, along withoverexpression of vasoconstrictor and proliferative substances such asthromboxane A2 and endothelin-1 (ET-1). Inflammatory cells, platelets(through the serotonin pathway), andprothrombotic abnormalities alsoplay a significant role in pathogenesis of PH.

However, the role of other molecular and signaling pathway, whichismediated bymany factors such as ET-1, serotonin (5-HT), angiotensinII (Ang II), growth factor etc.…, in pathobiology and pathogenesis of PHis not clearly clarified.

2.4. Genetic disorders in pulmonary hypertension

The best characterized genetic defects in heritable pulmonaryarterial hypertension are mutations of the gene encoding bonemorphogenetic protein receptor type 2 (BMPR2), a member of thetransforming growth factor-β signaling family. BMPR2 modulatesthe growth of vascular cells by activating the intracellular pathwayof Smad and LIM kinase (Foletta et al., 2003). The germline muta-tions in BMPR2 gene are detected in at least 70% of cases of

heritable pulmonary arterial hypertension (Machado et al., 2006,2009). BMPR2 gene mutations are also detected in 11%–40% of ap-parently sporadic cases, thus representing the major geneticpredisposing factor for PAH (Sztrymf et al., 2008). More than 45 dif-ferent mutations of BMPR2 gene have been identified in patientswith heritable PAH (Lane et al., 2000; Newman et al., 2001). Func-tional studies have shown that point mutations and truncations in thekinase domain exert dominant negative effects on receptor function(Newman et al., 2004), resulting in incomplete penetrance and geneticanticipation.

Mutations of other receptors such as activin receptor-like kinase 1(ALK1) and endoglin have also been identified in PAH patients usuallyfrom families with coexistent hereditary hemorrhagic telangiectasia(Trembath et al., 2001). Mutations in ALK1 are believed to result incellular growth-promoting via Smad-depending signaling. Geneticmutations of the serotonin transporter (5-HTT) are more frequentin idiopathic PAH than control subjects (Eddahibi et al., 2001). TheL-allelic variant of the 5HTT gene is associated with an increased ex-pression of the transporter and increased proliferation of vascularSMC. Serotonin gene polymorphism has also been found in PH

355S. Duong-Quy et al. / Pharmacology & Therapeutics 137 (2013) 352–364

patients with hypoxemic chronic obstructive pulmonary disease(COPD) (Eddahibi et al., 2003).

2.5. Role of inflammation

The roles of inflammation and autoimmunity in vascular remod-eling seen in idiopathic PH and PH associated with systemic diseaseshave been recently highlighted (Tuder & Voelkel, 1998; Voelkel et al.,1998). First, circulating autoantibodies directed against endothelialcells and cell nuclei are frequently found in PH (Isern et al., 1992;Dorfmüller et al., 2003; Tamby et al., 2005). Secondly, perivascularinfiltration of inflammatory cells including lymphocytes (T and B),macrophages, and dendritic cells, is a constant feature of plexiformvascular lesions (Tuder et al., 1994; Pietra et al., 2004; Perros et al.,2007b).

Cytokines and inflammatory chemokines are also involved in thepathogenesis of PH. Increased concentrations and expressions ofpro-inflammatory cytokines (IL-1β, IL-6) were found in plasma andlung tissue of patients with severe idiopathic PAH (Humbert et al.,1995). In mice, high expression of IL-6 was associated with increasedpulmonary vascular resistance, and extensive pulmonary vascularlesions (Steiner et al., 2009). Efficacy of tocilizumab, a monoclonal an-tibody directed against IL-6 receptors, has been recently reported in apatient with PH (Furuya et al., 2010). Plasma concentrations and ex-pressions of fractalkine and its receptors (CX3CR1 and CX3CL1) areincreased in circulating lymphocytes (CD4+ and CD8+) and lung tis-sue (Balabanian et al., 2002), and the role of the chemokines CCL5(RANTES) and CCL2 (chemokine ligand 2) studied in PH patients(Perros et al., 2007a; Sanchez et al., 2007).

2.6. Pulmonary vascular remodeling

Pulmonary vascular remodeling is a major pathological feature ofPH. It is due to encroach of vascular walls into the lumen, thus reducingthe vessels inner diameter. Many cell types of pulmonary vessel and cir-culation contribute to the development of pulmonary vascular remod-eling. This remodeling is characterized by the thickening of all thethree layers of pulmonary vessels, including the media, the intima,and the adventitia. It is marked by the hypertrophy and hyperplasia ofdifferent cell types such as endothelial cells (EC), smooth muscle cells(SMC), and fibroblasts. These structural changes are enhanced by theaccumulation of extracellular matrix components, including collagen,elastin, fibronectin, and tenascin, in the vascular walls, and predomi-nantly in the adventitia (Jeffery & Wanstall, 2001).

In the remodeled of pulmonary vessels, the thickened andhypertrophied media layer critically reduces vascular lumen whilstperturbing the fragile balance between vascular contraction andrelaxation mediators. Medial hypertrophy results from imbalancebetween proliferation and apoptosis of SMC, occurring at all levelsof pulmonary vascular tree, from large arteries to small arterioles(Dingemans & Wagenvoort, 1978). Furthermore, medial hypertro-phy further obstructs vascular lumen through extension of newmuscle cells into muscular and non muscular pulmonary arterioles,termed as the muscularization (Wagenvoort & Wagenvoort, 1970).Muscularization of pulmonary vessels is initiated by the differentia-tion of many precursor cells and fibroblasts into SMC (Jones et al.,1999; Sata, 2006).

Pulmonary vascular remodeling is also characterized by hypertrophyof the intima. Lung injury caused by hypoxia, inflammation, and shearstress, results in endothelial cells damage and dysfunction, uncontrolledcell proliferation, excessive vasoconstriction, and in situ thrombosis.These processes are linked to complex patterns of inflammation, angio-genesis, and trans-differentiation of endothelial cells to pulmonary vas-cular SMC (Zhu et al., 2006; Sakao et al., 2007), leading to intimalproliferation and lamina-intima fibrosis and yielding plexiform lesionsof pulmonary arteries.

3. Rho-kinase signaling pathway

3.1. Molecular structure and expression of Rho-kinase

Rho-kinase, also namedRho-associated kinase and identified inmild1990s, is one of the effectors of Rho families (Ishizaki et al., 1996).Rho-kinase or ROCKs having two different isoforms, ROCK-1or ROCK-β and ROCK-2 or ROCK-α, is the main downstream effectors ofGTPase-RhoA (Ishizaki et al., 1996). ROCKs are serine/threoninekinases with a molecular mass of ~160 kD. These kinases are formedby parallel homodimers including a catalytic (kinase) domain in itsamino-terminus (NH2- or N-terminal domain), a coiled-coil in itsmiddle dimerization portion, and a putative Pleckstrin-homology(PH) domain in its cystein-riche domain (COOH- or C-terminaldomain, CRD) (Fig. 2) (Ishizaki et al., 1996; Matsui et al., 1996;Fukata et al., 2001). These carboxyl terminal domains constitute anautoinhibitory region that reduces the kinase activity of ROCKs(Amano et al., 1999). The Rho-binding domain (RBD) of ROCKs is lo-calized in the C-terminal portion of the coiled-coil region, and itshows sequence homology to the Rho-interaction domain of kinectinwhich is a regulating protein of microtubule-based organelle motility(Alberts et al., 1998) (Fig. 2). The coiled-coil region of ROCKs is showedto interact with other α-helical proteins, whereas the PH domain is in-volved in protein localization (Riento & Ridley, 2003).

ROCKs mRNAs are expressed in invertebrates and in vertebrates.In human, ROCK-1 and ROCK-2 are encoded by two different geneslocalizing on chromosome 18 (18q11.1) and chromosome 2 (2p24),respectively (Nakagawa et al., 1996; Takahashi et al., 1999). Twoisoformes of Rho-kinase in human have the homologue structurewith about 65% of amino acid and 58% of RBD. The highest similarity(92%) is presented at kinase domain (Leung et al., 1995; Nakagawa etal., 1996). The mRNAs of ROCK-1 and ROCK-2 are ubiquitouslyexpressed, with a preferential expression of ROCK-2 mRNA in brainand skeletal muscle. Both ROCK-1 and ROCK-2 are expressed in vas-cular smooth muscle and in heart (Leung et al., 1996; Wibberley etal., 2003). Cell-fractionation studies show that ROCKs are mainly dis-tributed in the cytoplasm fraction but a small amount of ROCKs is alsofound in the membrane fraction (Leung et al., 1996; Matsui et al.,1996). In addition, ROCK-1 might colocalize with centrosomes(Chevrier et al., 2002). However, ROCKs are also found in subcellularlocalization at the vimentin intermediate-filament network and atactin stress fibers (Sin et al., 1998; Kawabata et al., 2004).

3.2. Regulation of Rho-kinase activity

In the structure of ROCKs, the C-terminus (Rho-binding domain andPH domain) plays a role as a dominant-negative autoinhibitor (Fig. 3)due to its independent interaction with the catalytic domain (N-termi-nus) (Amano et al., 1999). Lacking of the C-terminus of ROCKs (truncat-ed forms) are constitutively formed the active form kinase (Amano etal., 2000). Beside of the self-associative autoinhibition, the activity ofROCKs is also influenced by its affinity for ATP which is regulated bythe dimerization of kinases (Doran et al., 2004).

Binding of GTPase-RhoA (amino acids 23–40 and 75–119 ofRhoA) to ROCKs at Rho-binding domain induces conformationalchanges of ROCKs, resulting in relieve of autoinhibitory blockageof kinase activity (Fujisawa et al., 1998). This binding is believedto stimulate the phosphotransferase activity of ROCKs (positive reg-ulation) by disrupting the interaction between catalytic and theC-terminal region of proteins (Loirand et al., 2006), which therebyfrees the kinase activity (Fig. 4a).

Independently of RhoA, ROCK activity might be activated by otherstimulators such as arachidonic acid (AA), sphingosine phosphoryl-choline (SPC), caspase-3 or granzyme B (Feng et al., 1999b; Shirao etal., 2002; Sebbagh et al., 2001, 2005). AA and SPC interact with thenegative regulatory region at PH domain, thus disrupting its inhibitory

Fig. 2. The molecular structure of ROCKs. The kinase domain is located at the amino terminus (N-terminus) of the protein, followed by coiled-coil region containing the Rho-bindingdomain (RBD). The Pleckstrin-homology (PH) with an internal cystein-rich domain (CRD) is situated in the carboxyl terminus (C-terminus).

356 S. Duong-Quy et al. / Pharmacology & Therapeutics 137 (2013) 352–364

property on the catalytic activity of ROCKs (Feng et al., 1999b; Shirao etal., 2002). Caspase-3 cleaves ROCK-1 at the cleavage site DETD1113whereas granzyme B cleaves the ROCK-2 at the C-terminus at IGLD1131,thus removing an inhibitory region (Fig. 4a) (Sebbagh et al., 2001,2005).

The activity of ROCKs is also negatively regulated by other smallG-binding proteins such as RhoE, Gem, and Rad (Ward et al., 2002;Riento et al., 2003). RhoE binds to the N-terminal region of kinasedomain of ROCK-1 (amino acids 1–420) and therefore interfereswith the kinase activity and prevents GTPase-RhoA binding toRho-binding domain (Fig. 4b) (Riento et al., 2003). Overexpressionof Gem and Rad might inhibit respectively the downstream re-sponses of ROCK-1 and ROCK-2, but the mechanism is not clearlydemonstrated (Ward et al., 2002). In addition, the negative regula-tion of ROCK-mediated target effects by these small G-binding pro-teins are localized at the different cellular structure (RhoE in theGolgi and Gem and Rad in the cytoskeleton) (Bilan et al., 1998;Piddini et al., 2001).

4. Vascular downstream effect of Rho-kinase

4.1. Rho-kinase and vascular smooth muscle cells (VSMC)

4.1.1. Rho-kinase and vascular smooth muscle cells contractilityVascular smooth muscle cell (VSMC) contractility is dependent on

the level of phosphorylation of the 20 kDamyosin light chain (MLC20)that is determined by both Ca2+-dependent and Ca2+-independentmechanisms (Fig. 5).

VSMC Ca2+-dependent contractility is related to the concentrationof Ca2+ in cytosolic ([Ca2+]i) which is regulated by the release ofCa2+ from the sarcoplasmic reticulum and the entry of Ca2+ from theextracellular space via voltage-dependent Ca2+ channels (VDCCs) orthrough non-selective cation channels (NSCCs) (VanBavel et al., 2002;Moosmang et al., 2003). Increase of [Ca2+]i in cytosolic promotes there-fore the binding of Ca2+ to a specific pool of calmodulin (CaM) tetheredtomyosin light chain kinase (MLCK) (Wilson et al., 2002). It results thento the activation of MLCK and MLCK-mediated phosphorylation ofMLC20, thereby activating cross-bridge cycling and contraction.

Fig. 3. The autoinhibition form of ROCKs. The Pleckstrin-homology (PH) and Rho-binding

Vascular Ca2+-independent contractivity is related to Ca2+ sensitiza-tion of VSMC that is dependent on an increase of MLC20 phosphorylationand a force of contractile myofilaments without eliciting a change in[Ca2+]i. Many cellular signaling pathways contribute to culminate the in-hibition of myosin light chain phosphatase (MLCP) activity, increasingtherefore the level of phosphorylated MLC20 (Somlyo & Somlyo, 2003).ROCKs, downstream effectors of GTPase-RhoA, phosphorylate theMLCP targeting subunit (MYPT1) at threonine (Thr)-696, Thr-853,and Thr-855 (Somlyo & Somlyo, 2003; Wilson et al., 2005; Knock etal., 2009), resulting in inhibition of MLCP activity and increasing ofMLC20 phosphorylation (Feng et al., 1999a; Velasco et al., 2002).This in turn, enhances the actin binding and actin-induced ATPaseactivity of myosin, facilitating interaction of myosin with F-actin,then VSMC contractility (Fig. 5).

However, the inhibition of MLCP activity might bemediated by a di-rect interaction of the small 17 kDa protein kinase C-potentiated myo-sin phosphatase inhibitor protein (CPI-17) with protein phosphatase 1catalytic subunit (PP1c) of MLCP. Phosphorylation of CPI-17 by ROCKsat Thr-38 (Koyama et al., 2000), and protein kinase C (PKC) (Eto et al.,1997) also enhances the potency of CPI-17 for inhibiting MCLP activity(Takizawa et al., 2002), and VSMC contractility.

4.1.2. Rho-kinase and vascular smooth muscle cells proliferationThe proliferation of VSMCs is related to an activation of multiple

pro-mitogenic signals, which interferes in the regulation of cell cycle(Assoian & Marcantonio, 1996). During the cell cycle, the progressionthrough the G1 phase is regulated by phosphorylation and inactivationof retinoblastoma (Rb) proteins, which is promoted by the Cyclins(Cyclin A, D, and E), inducing an activation of cyclin-dependent kinases(cdk2, cdk4, and cdk6). These cdk-cyclin complexes are however, neg-atively regulated by the Cip/Kip family (p21Cip1, p27Kip1, and p57Kip2)of Cyclin-dependent kinase inhibitors (CDKIs) (Morgan, 1995; Sherr &Roberts, 1995). Hence, the cyclin-dependent kinase inhibitor p27Kip1

plays a crucial role in VSMC proliferation.There is increasing evidence that ROCK effectors downregulate

p27Kip1 expression, leading to the acceleration of cell cycle progressionand VSMC proliferation (Laufs et al., 1999b; Sawada et al., 2000). Theeffect of ROCK inhibitors as antiproliferators has been also ascribed to

domain (RBD) bind to the N-terminus of the enzyme, forming an autoinhibitory loop.

Fig. 4. a. The mechanism of positive regulation of ROCKs activity. Binding of GTPase-RhoA to Rho-binding domain (RBD) relieves the autoinhibitory blockage of kinase activity. Ar-achidonic acid (AA) and sphingosine phosphorylcholine (SPC) interact with the negative regulatory region at PH domain, disrupting the inhibitory property of ROCKs. Caspase-3and granzyme B cleave ROCKs at cleavage sites, removing the inhibitory regions. b. The mechanism of negative regulation of ROCKs activity. RhoE binds to the N-terminal region ofkinase domain of ROCKs (ROCK-1) prevents GTPase-RhoA binding to Rho-binding domain (RBD).

357S. Duong-Quy et al. / Pharmacology & Therapeutics 137 (2013) 352–364

the upregulation of p27Kip1 expression (Sawada et al., 2000; Kanda etal., 2005). In addition, the role of ROCKs in the proliferation of VSMCme-diated by platelet-derived growth factor (PDGF) has been demonstratedby a previous study (Kamiyama et al., 2003). Inhibition of ROCKs abol-ishes the PDGF-induced activation of extracellular-regulated kinase 1/2(ERK1/2) and proliferation of VSMCs. Moreover, inhibition of ROCKsalso suppresses VSMC proliferation mediated by G-protein-coupledreceptor-stimulated cell proliferation which is promoted by many medi-ators such as thrombin, urotensin-II, and angiotensin-II (Seasholtz et al.,1999; Yamakawa et al., 2000; Sauzeau et al., 2001).

4.2. Rho-kinase and endothelial cells (EC)

4.2.1. Rho-kinase and endothelial nitric oxide synthase/nitric oxide signalingIn endothelium, nitric oxide (NO) is synthesized from the conver-

sion of L-arginine to L-citrulline in the presence of endothelial nitricoxide synthase (eNOS) by using oxygen, NADPH, and substrates(Moncada et al., 1989). After diffusing into the VSMCs, NO activatessoluble guanylate cyclase (sGC), which catalyzes the formation of

guanosine monophosphate (cGMP) and the subsequent activation ofcGMP-dependent protein kinase (cGK). The effect of NO/cGK cascadesignalization in VSMC relaxation results from the decrease of [Ca2+]iintracellular due to the activation of the sarcoplasmatic Ca2+-ATPase,reducing Ca2+ release from intracellular store and the phosphorylationand activation of MLP20. In addition, in VSMCs, the role of eNOS/NO sig-nalization as antiproliferation has been demonstrated (Emerson et al.,1999; Keil et al., 2002).

Recent studies showed that Rho-kinase/ROCKs might negativelyregulate eNOS expression, eNOS activity, and NO bioavailability(Laufs & Liao, 1998; Laufs et al., 1999a; Takemoto et al., 2002;Rikitake & Liao, 2005). Inhibition of ROCKs increases eNOS mRNAstability and eNOS expression (Rikitake et al., 2005). ROCK inhibi-tors also enhance phosphorylation and activation of the Akt/phos-phatidylinositol-3 kinase (PI-3K) pathway, leading to increase NOproduction (Wolfrum et al., 2004). In addition, the pleiotropic effectof statins, a nonselective ROCKs inhibitors, on upregulation of eNOShas been demonstrated (Ni et al., 2001; Girgis et al., 2003;Nishimura et al., 2003). Statins upregulate eNOS/NO signalization

Fig. 5. Ca2+ sensitization of VSMC mediated by RhoA/Rho-kinase signaling pathway. In basal state of VSMCs, there is a balance between vasoconstriction and vasodilatation that iscontrolled by the activity of MLCK and MLCP. When Rho-kinase agonists are coupled on GPCR, it converts GDP-RhoA (inactive form) to GTP-RhoA (active form). GTP-RhoA trans-locates into the membrane and activates its downstream effectors: Rho-kinase or ROCKs. ROCKs increase the vasoconstriction by inhibiting the activity of MLCP via the phosphor-ylation of this enzyme.

358 S. Duong-Quy et al. / Pharmacology & Therapeutics 137 (2013) 352–364

via the inhibition of geranylgeranylation of the small G-protein Rho,and translocate inactive Rho form the cytosol to themembrane (Liao& Laufs, 2005).

4.2.2. Endothelial nitric oxide synthase/nitric oxide signaling and GTPase-RhoA/Rho-kinase

On the other hand, the effect of eNOS/NO signalization on RhoA/Rho-kinase pathway has been recently studied. Previous studiesfounded that NO through cGMP/cGK might serine-phosphorylateRhoA leading to decrease the association of RhoA protein on cellmembrane (Sauzeau et al., 2000; Begum et al., 2002; Chitaley &Webb, 2002; Krepinsky et al., 2003). As a result, it reducedRhoA-GTP active form, and therefore, decreased the activity ofdownstream target effects of ROCKs. Sauzeau et al. (Sauzeau et al.,2000) showed that exogenous NO attenuated Ca2+-dependentROCKs sensitization of blood vessel contraction by inhibiting RhoAtranslocation from the cytosol to membrane in VSMCs via cGK path-way. Moreover, NOmight also inhibit RhoA/ROCKs independently ofcGMP by S-nitrosating RhoA via interaction with cysteine to formnitrothiols (Hess et al., 2001; Stamler et al., 2001; Zuckerbraun etal., 2007).

5. Role of Rho-kinase signaling in pulmonary hypertension

5.1. In animal models

The role of RhoA/Rho-kinase pathway in PAH has been principallystudied in chronic hypoxia (CH)-induced PH and monocrotaline(MCT)-induced PH (Table 1). Chronic hypoxia due to different disor-ders or pathologies, is one of the major causes of PH. Exposure tochronic hypoxia induces structural and functional changes in pulmo-nary arterial bed (Rabinovitch et al., 1979; Pierson, 2000) leading toendothelial dysfunction (Higenbottam & Laude, 1998), and modifica-tion of pulmonary vascular contractile properties (Yuan et al., 1998)as described above. Furthermore, in hypoxia-induced PH, the balanceof vasoconstriction–vasodilatation tonus has been impaired by other

vasoconstrictors such as ET-1, 5-HT, Ang II. These vasoconstrictorsalso mediate RhoA/Rho-kinase pathway (Barman, 2007; Homma etal., 2007), and it in turn, is involved in downregulation of eNOS sig-nalization, VSMC hyperconstriction (Batchelor et al., 2001; Sakuradaet al., 2001; Nagaoka et al., 2004, 2006; McNamara et al., 2008;Desbuards et al., 2009), and pulmonary vascular remodeling (Laufset al., 1999a; Sauzeau et al., 2001; Keil et al., 2002). It is known thatdistal muscularization of pulmonary circulation has been increasedduring chronic hypoxia and RhoA/Rho-kinase pathway is involvedin growth and hypertrophy of SMC via the mitogenic effectors(ET-1, 5-HT) in pulmonary vascular remodeling (Emerson et al.,1999; Fagan et al., 1999; Aznar and Lacal, 2001; Keil et al., 2002).

In animal model, PH can be achieved by injection with a singlesubcutaneous or intraperitoneal of monocrotaline (MCT). Althoughthe exact mechanism though which MCT-induced PH is not wellunderstood. It is suggested that injection of MCT-induced severe orlethal PH is mediated by its direct toxic effect on endothelial cellsand dramatic accumulation of mononuclear inflammatory cells, par-ticularly macrophage, in the small intraacinar vessels (Wilson et al.,1989; Jasmin et al., 2001). The role of Rho-kinase in MCT-inducedPH in rats has been firstly demonstrated by Khan et al. (2005). Inthis study, the authors showed that while pulmonary arterialcontraction induced by noradrenaline was attenuated and due topoor-coupling to G-protein-coupled receptor, the Rho-kinase activ-ity was increased with principally ROCK-2. Increase of Rho-kinaseactivity, mediated by HMG-CoA reductase and cleaved caspase-3,is also involved in MCT-induced PH in pneumonectomized rats(Homma et al., 2008). Recent study showed that increases asym-metric dimethylarginine (ADMA) which is involved in vascular re-modeling in MCT-induced PH, is mediated by Rho-kinase activity(Li et al., 2010).

Rho-kinase signaling pathway also plays a important role in thepathogenesis of other models of PH. Li F et al. demonstrated that Wis-ter rats with carotid-external jugular vein communication, a model ofhigh-pressure systemic–to-pulmonary shunts, developed PH after 4wk with excessive vascular remodeling at week 8 (Li et al., 2007). In

Table 1The role of Rho-kinase signaling pathway in pulmonary hypertension.

Authors,Year

PH model Involved role of Rho-kinasesignaling in PH

(Li et al.,2010)

MCT-induced PH in rats PASMCs proliferation mediatedby ADMA

(Do et al.,2009)

iPAH patients Impaired endothelium-dependentrelaxation; serotonin-inducedhypercontraction

(Guilluy etal., 2009)

iPAH patients 5-HT -mediated PASMCsproliferation and plateletactivation

(Desbuardset al.,2009)

Hypoxic associated withdiscontinuateddexfenfluramine-induced PH inrats

PH due to discontinuation ofanorexia

(Gien et al.,2008)

Partial ligation of the ductusarteriosus in utero (PPHN)sheep

Impaired angiogenesis;downregulated eNOS and NO

(Homma etal., 2008)

MCT-induced PH inpneumonectomized rats

PH mediated by HMG-CoAreductase and cleaved caspase-3

(McNamaraet al.,2008)

CH- and bleomycin-induced PHin rats

Vasoconstriction

(Hemnes etal., 2008)

Bleomycin-induced PH in rats ROS-mediated lung fibrosis andNOS uncoupling

(Homma etal., 2007)

CH-induced PH in rats Vasoconstriction in response toKCl mediated by ET-1 and 5-HT

(Barman,2007)

CH-induced PH in rats ET-1-induced vasoconstriction

(Oka et al.,2007)

Severe occlusive PAH in rats Angioproliferation in pulmonaryarterioles; vasoconstriction

(Li et al.,2007)

High flow-induced PH in rats Vascular remodeling

(Nagaokaet al.,2006)

PH of Denver FHR Sustained vasoconstriction

(Khan et al.,2005)

MCT-induced PH in rats NA-induced PA contraction

(Nagaokaet al.,2004)

CH-induced PH in rats Vasoconstriction

MCT, monocrotaline; PH, pulmonary hypertension; ADMA, asymmetric dimethylarginine;PASMCs, pulmonary arterial smoothmuscle cells; iPAH, idiopathic pulmonary arterial hyper-tension; ROS, reactive oxygen species; 5-HT, serotonin; PPHN, persistent pulmonary hyper-tension of the newborn; CH, chronic hypoxia; ET-1, endothelin-1; FHR, fawn hooded rat;NA, noradrenalin.

359S. Duong-Quy et al. / Pharmacology & Therapeutics 137 (2013) 352–364

this study, Rho-kinase signaling pathway was involved in increasedproliferation and decreased apoptosis of SMCs. In severe PHmodel in-duced by hypoxia associated with injected Sugen 5416, a VEGF recep-tor inhibitor, Oka et al. (2007) founded that Rho-kinase activity had acrucial role in the development of occlusive pulmonary vascular le-sions and sustained vasoconstriction. The role of Rho-kinase signalingpathway in bleomycin-induced PH has been demonstrated (Hemneset al., 2008). It is speculated that in this model, Rho-kinase activityis mediated by enhanced reactive oxygen species (ROS), resultinglung fibrosis and NOS uncoupling. Another role of Rho-kinase in ani-mal model with PH has been also reported (Gien et al., 2008).

5.2. In humans

Although the role of Rho-kinase signaling pathway in pathogene-sis of PH has been confirmed in diverse animal models, its role inhuman pulmonary hypertension is not well studied. The first pub-lished study in this field realized by Guilluy et al. (2009), demonstrat-ed that RhoA/Rho-kinase activity was increased in patients withidiopathic pulmonary arterial hypertension (iPAH). The upregulationof RhoA/Rho-kinase in these patients has been mediated by 5-HT viathe serotonylation of Rho protein, contributing to the proliferation ofpulmonary arterial SMCs. In the study from patients with pulmonaryhypertension who underwent lung transplantation, Do et al. (2009)

showed that Rho-kinase activity was correlated to severity (meanpulmonary arterial pressure, mPAP) and evolution of the disease. Inthis study, Rho-kinase activity was involved in pulmonary arterialhypercontraction mediated by PGK and endothelial dysfunction.

6. Rho-kinase inhibitors in the treatment of pulmonary hypertension

6.1. In animal models

6.1.1. Y-27632 in the treatment of pulmonary hypertensionY-27632 [−R-trans-4-(1-aminoethyl)-N-(4-pyridyl) cyclohexane-

carboxamide dihydrochloride]] is one of the compounds derived frompyridine with potent ROCK inhibitory effect (Uehata et al., 1997).Y-27632 is a non-specific inhibitor of ROCKs, competing with ATP forbinding to ROCK catalytic sites (Ishizaki et al., 2000). It has also a potentinhibitory effect of Rho-dependent protein kinase C (Uehata et al.,1997).

In the initial study on the effect of inhibition of Rho-kinase in micewith CH-induced PAH, Fagan et al. (2004) reported that Y-27632 de-creased pulmonary vasoconstriction via mediating Ca2+ sensitizationand attenuated the risk of developing PH and vascular remodeling.The study showed that Y-27632 at 30 mg−1.day−1 reduced right ven-tricle systolic pressure (RVSP), right ventricle hypertrophy (RV hy-pertrophy), and neomuscularization of the distal pulmonaryvasculature. Particularly, treatment with Y-27632 increased the ex-pression of eNOS protein in lung tissues of CH-induced PH.

Many other studies also showed that in CH-induced PH in rats,Y-27632 attenuated significantly hemodynamic changes and vascularremodeling in treated rats in comparison with non treated group(McNamara et al., 2008; Xu et al., 2010; Ziino et al., 2010). Recently,Vanderpool et al. (2011) showed that, by using isolated lung ofCH-induced PH, Y-27632 decreased the pulmonary arterial imped-ance, the diameter of main pulmonary arterial, and the right pulmo-nary compliance.

However, Y-27632 is not a high selective pulmonary vasodilator be-cause of its effects on systemic circulation when used intravenously ororally. To avoid the systemic vasodilator effect of Y-27632, Nagaoka etal. (2005) demonstrated that inhalation of aerosolized Y-27632 had aselective effect by reducing mean pulmonary arterial pressure (mPAP)without systemic effect in chronic hypoxia Sprague–Dawley Rat (SDR).The selective pulmonary hypotension effect of aerosolized Y-27632lasted more than 5 h after 5 min of inhalation. Especially, its effect wasstronger than that with inhalation of NO.

6.1.2. Fasudil in the treatment of pulmonary hypertensionFasudil (HA-1077) is the first approved ROCK for clinical use in the

treatment of ischemia-induced brain damage (Toshima et al., 2000;Satoh et al., 2001; Rikitake et al., 2005). Fasudil is a selective ROCK in-hibitor, competing with ATP for the binding to the kinase (Davies etal., 2000). After oral administration, fasudil is metabolized in hydroxylfasudil (HA-1100), having more selective inhibitory effect on ROCKs(Shimokawa et al., 1999).

Fasudil is more effective than Y-27632 in prevention of CH-inducedPH (Shimokawa, 2002; Shimokawa et al., 2002; Abe et al., 2006). Thefirst use of fasudil in the treatment of PH in animal model has been dem-onstrated byAbe et al. (2004). In this study, the long-term treatmentwithfasudil improved hemodynamic parameters (PAP, RVSP, RV hypertro-phy), pulmonary vascular remodelingwith suppression of VSMCprolifer-ation and enhancing of VSMC apoptosis in MCT-induced PH in rats.Fasudil ameliorated endothelial dysfunction and VSMC hypercontraction.The beneficial effect of fasudil in hemodynamic parameters in MCT-induced PH has been demonstrated by other studies (Jiang et al., 2007;Mouchaers et al., 2010).

The role of fasudil in the treatment of PH also has been demonstrat-ed in othermodel of PH (Table 2). InWistar ratswith highflow-inducedPH (Li et al., 2007), treatment with fasudil suppressed Rho-kinase

360 S. Duong-Quy et al. / Pharmacology & Therapeutics 137 (2013) 352–364

hyperactivity, leading to improve PAP, RV hypertrophy, and pulmonaryvascular remodeling. These results were similar to CH-induced PH orsevere occlusive PH, and left-ventricular dysfunction-induced PH(Yamakawa et al., 2000; Guilluy et al., 2005; Abe et al., 2006; Nagaokaet al., 2006; Oka et al., 2007; Ziino et al., 2010; Dai et al., 2011). Particu-larly, fasudil improved the expression of eNOS (Abe et al., 2006) andnormalized pulmonary vascular resistance (PVR) in PH refractory to

Table 2Effects of Rho-kinase inhibitors in PH animal model.

Authors,Year

PH model Rho-kinaseinhibitors(daily dose)

Effect of Rho-kinaseinhibitors

(Vanderpoolet al.,2011)

CH-induced PH inmice

Y-27632 (10−5M, isolatedmouse lungs)

Decreased: PA impedance,diameter of main PA, andright PA compliance

(Dai et al.,2011)

Left-ventriculardysfunction-inducedPH in rats

Fasudil(30 mg/kg)

Improved: mPAP, RVhypertrophy, PA medialthickness

(Xu et al.,2010)

CH-induced PH injuvenile rats

Y-27632(15 mg/kg)

Attenuated: RVhypertrophy, PA wallremodeling; normalized:RVSF

(Dahal et al.,2010)

MCT-induced PH inrats; CH-induced PHin mice

Azaindole-1(10–30 mg/kg)

Improved: RVSP, TPR, RVhypertrophy; anti-PASMCproliferation

(Mouchaerset al.,2010)

MCT-induced PH inrats

Fasudil(40 mg/kg)

Reduced: mPAP, PVR,RVSP, RV hypertrophy

(Ziino et al.,2010)

CH-induced PH inneonatal rats

Y-27632(10 mg/kg)Fasudil(20 mg/kg)

Attenuated: hemodynamicand structural changes

(McNamaraet al.,2008)

CH- andbleomycin-inducedPH in rats

Y-27632(15 mg/kg)Fasudil(30 mg/kg)

Normalized: PVR(PH refractory to NO)

(Tawara etal., 2007)

MCT-induced PH inrats

Fasudil (30 mg/kg) associatedwith Beraprostsodium

Decreased: mPAP, RVhypertrophy, medialthickening (synergisticeffect)

(Oka et al.,2007)

Severe occlusivePAH in rats

Fasudil (10 mg/kg, intravenousadministration)

Decreased: RVSP,vasoconstrictionIncreased: CO

(Jiang et al.,2007)

MCT-induced PH inrats

Fasudil(10–30 mg/kg)

Reduced: mPAP

(Girgis et al.,2007)

CH-induced PH inrats

Simvastatine(non selectiveinhibitor)

Reduced: mPAP, RVhypertrophie, medialthickening

(Li et al.,2007)

High flow-inducedPH in rats

Fasudil(30 mg/kg)

Suppressed: PASMCproliferationAttenuated: RVhypertrophy

(Abe et al.,2006)

CH-induced PH inmice

Fasudil(100 mg/kg)

Improved: mPAP, RVhypertrophie, eNOSexpression

(Nagaoka etal., 2006)

PH of Denver FHR Fasudil(30 mg/kg)

Reduced: elevated PAPImproved: alveolarizationvascularization

(Guilluy etal., 2005)

CH-induced PH inrats

Fasudil(30 mg/kg)

Reduced: PAP, RVhypertrophy

(Nagaoka etal., 2005)

CH-induced PH inratsMCT-induced PH inrats

Y-27632(30 mg/kg)InhaledY-27632 andfasudil

Decreased: mPAP

(Fagan et al.,2004)

CH-induced PH inmice

Y-27632(30 mg/kg)

Decreased: RVSP, RVhypertrophie

(Abe et al.,2004)

MCT-induced PH inrats

Fasudil (30–100 mg/kg)

Improved: survival, RVSP,RV hypertrophie, medialthickening, endothelialdysfunction

PH, pulmonary hypertension; CH, chronic hypoxia; PA, pulmonary artery; SMCs, smoothmuscle cells; MCT, monocrotaline; RV, right ventricle; RVSF, right ventricular systolicfunction; RVSP, right ventricular systemic pressure; mPAP, mean pulmonary arterialpressure; TPR, total pulmonary resistance; PVR, pulmonary vascular resistance.

nitric oxide (NO) (McNamara et al., 2008). In Fawn-Hooded Rat (FHR)with severe PHwhen raised for the first weeks of life in themild hypox-ia of Denver's altitude (Nagaoka et al., 2006), long-term treatment withfasudil reduced the development of PH and improved lung dysplasia inrat pups by decreasing alveolar size and increasing pulmonary vasculardensity.

Moreover, fasudil might be used in combination with other vaso-dilators in the treatment of PH. Tawara et al. (2007) showed that inMCT-induced PH in rats, when compared with monotherapy, thecombination of fasudil and prostacyclin analogue (beraprost sodium)significantly improved mPAP, RV hypertrophy, and pulmonary arteri-al medial thickness without any adverse effects. This result suggestedthat combined therapy between a Rho-kinase inhibitor with other va-sodilators might exert more beneficial effects in PH treatment due tothe additive and synergic effects of combined treatment.

In addition, fasudil when used by inhalation is as effective as in-haled Y-27632 in the treatment of PH. In CH- and MCT-induced PHin rats, inhaled fasudil reduced significantly mPAP, without any sig-nificant change in systemic arterial pressure and cardiac index orheart rate (Nagaoka et al., 2005). This route of administration is oneof the advantages of fasudil in the treatment of PH and it should beexplored in the future.

6.1.3. Other Rho-kinase inhibitors in the treatment of pulmonary hypertensionAlthough many other new Rho-kinase inhibitors have been devel-

oped, their role in the treatment of PH has not been completely studied.Recently, the therapeutic efficacy of azaindole-1, a potent ROCK inhibi-tor with highly specific ATP-competition, in MCT- and CH-induced PHin rodent has been demonstrated (Dahal et al., 2010). Oral administra-tion of azaindole-1 improved the hemodynamic, RV hypertrophy andpulmonary vascular remodeling. In this study, azaindole-1 had a potenteffect improving a hypoxic pulmonary vasoconstriction and a pulmo-nary arterial SMC proliferation.

Beside the effects of selective Rho-kinase inhibitors in the treat-ment of PH, the role of statin, a non selective Rho-kinase inhibitorwith pleiotropic effect, in the treatment of PH has been demonstrated(Girgis et al., 2007).

6.2. In humans

Successful results of Rho-kinase inhibitors in the treatment of PHin animal model encourage many researchers try to study it inhuman. The first clinical essays of Rho-kinase inhibitors in treatmentof PH have been initiated by Fukumoto et al. (2005) in a small num-ber of enrolled patients (9 patients, mean age of 53 years). The re-sults of this study showed that intravenous administration offasudil (30 mg for 30 minutes) decreased PAP and increased cardiacindex (CI). It also reduced PVR significantly without any side effecton systemic pressure. This result is similar to what reported byIshikura et al. in patients with idiopathic pulmonary arterial hyper-tension (iPAH) and associated PAH (Ishikura et al., 2006).

Recently, the acute beneficial effects of fasudil on hemodynamic in thetreatment of congenital heart diseasewith left-to-right shunt-induced PHin young patients have been demonstrated by a prospective study (12patients, mean age of 12.3 years) (Li et al., 2009). In this study, intrave-nous injection of fasudil (30 mg over 30 min) significantly decreasedPAP and PVR and increased cardiac output, whereas systemic arterialpressure were slightly reduced as compared to baseline value. Fujita etal. showed that the use of fasudil by inhalation in the treatment of PH(iPAH, aPAH, and PAH due to left heart dysfunction) decreased mPAPand the ratio of pulmonary vascular resistance/systemic vascular resis-tance (Fujita et al., 2010). However, the long term effect of fasudil inpatientswith PHasmonotherapy or in combinationwith other treatmenthas not been demonstrated (Table 3).

Table 3Effects of Rho-kinase inhibitors in patients with PAH.

Authors,Year

PH model Rho-kinase inhibitors Effect ofRho-kinaseinhibitors

(Fujita etal., 2010)

Patients with PAH(iPAH, aPAH, PAHdue to LHD)

Fasudil (inhalation: 30 mg/10 min)

Decreased:mPAP, PVR/SVR

(Li et al.,2009)

Left-to-rightshunt-induced PH

Fasudil (intravenousadministration: 30 mg/30 min)

Decreased:PASP, PVR, PVR/SVRIncreased: CI

(Ishikura etal., 2006)

Patients with PAH( iPAH, aPAH)

Fasudil (intravenousadministration: 30 mg/30 min)

Decreased:mPAP, TPRIncreased: CI

(Fukumotoet al.,2005)

Patients withsevere PAH

Fasudil (intravenousadministration: 30 mg/30 min)

Decreased:mPAP, PVRIncreased: CI

PAH, pulmonary arterial hypertension; iPAH, idiopathic PAH; aPAH, associatedPAH; LHD, leftheart disease; mPAP, mean pulmonary arterial pressure; TPR, total pulmonary resistance;PVR, pulmonary vascular resistance; SVR, systemic vascular resistance; PASP, pulmonaryarterial systolic pressure; CI, cardiac index.

361S. Duong-Quy et al. / Pharmacology & Therapeutics 137 (2013) 352–364

7. Conclusion

More than thirty years after the first meeting of PH in Geneva, upto now, PH is still an incurable disease with dramatic survival. De-spite the combination of available vasodilator drugs at early stageof disease, the prognostic of PH remains unfortunately the same assome progressed cancer. The new findings on pathophysiology andpathobiology of PH help physicians improving advanced knowledgeon pathogenesis of this disease. Recent discovery on the role ofRhoA/Rho-kinase signaling pathway in patients with PH and the ef-fects of Rho-kinase inhibitors in vivo promise a novel pharmacolog-ical treatment of PH. These selective Rho-kinase inhibitors areaimed to improve the pulmonary arterial relaxation and remodeling.However, the long-term outcome of clinical trials of Rho-kinase in-hibitors in patients with PHwill be necessary in the future to confirmits effects.

Conflict of interest statement

The undersigned authors, Sy Duong‐Quy, Yihua Bei, Zhongmin Liu,and Anh Tuan Dinh‐Xuan declare that there are no conflict of interestrelated to this work.

References

Abe, K., Shimokawa, H., Morikawa, K., Uwatoku, T., Oi, K., Matsumoto, et al. (2004).Long-term treatment with a Rho-kinase inhibitor improves monocrotaline-inducedfatal pulmonary hypertension in rats. Circ Res 94, 385–393.

Abe, K., Tawara, S., Oi, K., Hizume, T., Uwatoku, T., Fukumoto, Y., et al. (2006). Long-term in-hibition of Rho-kinase ameliorates hypoxia-induced pulmonary hypertension in mice.J Cardiovasc Pharmacol 48, 280–285.

Alberts, A. S., Bouquin, N., Johnston, L. H., & Treisman, R. (1998). Analysis of RhoA-bindingproteins reveals an interaction domain conserved in heterotrimeric G protein beta sub-units and the yeast response regulator protein Skn7. J Biol Chem 273, 8616–8622.

Amano, M., Chihara, K., Nakamura, N., Kaneko, T., Matsuura, Y., & Kaibuchi, K. (1999).The COOH terminus of Rho-kinase negatively regulates rho-kinase activity. J BiolChem 274, 32418–32424.

Amano, M., Fukata, Y., & Kaibuchi, K. (2000). Regulation and functions of Rho-associatedkinase. Exp Cell Res 261, 44–51.

Assoian, R. K., & Marcantonio, E. E. (1996). The extracellular matrix as a cell cycle con-trol element in atherosclerosis and restenosis. J Clin Invest 98, 2436–2439.

Aznar, S., & Lacal, J. C. (2001). Rho signals to cell growth and apoptosis. Cancer Lett 165, 1–10.Balabanian, K., Foussat, A., Dorfmüller, P., Durand-Gasselin, I., Capel, F., Bouchet-Delbos, L.,

et al. (2002). CX(3)C chemokine fractalkine in pulmonary arterial hypertension. Am JRespir Crit Care Med 165, 1419–1425.

Barman, S. A. (2007). Vasoconstrictor effect of endothelin-1 on hypertensive pulmo-nary arterial smooth muscle involves Rho-kinase and protein kinase C. Am J PhysiolLung Cell Mol Physiol 293, L472–L479.

Batchelor, T. J., Sadaba, J. R., Ishola, A., Pacaud, P., Munsch, C. M., & Beech, D. J. (2001).Rho-kinase inhibitors prevent agonist-induced vasospasm in human internalmammaryartery. Br J Pharmacol 132, 302–308.

Begum, N., Sandu, O. A., & Duddy, N. (2002). Negative regulation of rho signaling by insulinand its impact on actin cytoskeleton organization in vascular smooth muscle cells: Roleof nitric oxide and cyclic guanosine monophosphate signaling pathways. Diabetes 51,2256–2263.

Bilan, P. J., Moyers, J. S., & Kahn, C. R. (1998). The ras-related protein rad associateswith the cytoskeleton in a non-lipid-dependent manner. Exp Cell Res 242,391–400.

Channick, R. N., Simonneau, G., Sitbon, O., Robbins, I. M., Frost, A., Tapson, V. F., et al. (2001).Effects of the dual endothelin-receptor antagonist bosentan in patients with pulmonaryhypertension: A randomised placebo-controlled study. Lancet 358, 1119–1123.

Chevrier, V., Piel, M., Collomb, N., Saoudi, Y., Frank, R., Paintrand, M., et al. (2002). TheRho-associated protein kinase p160ROCK is required for centrosome positioning.J Cell Biol 157, 807–817.

Chitaley, K., & Webb, R. C. (2002). Nitric oxide induces dilation of rat aorta via inhibi-tion of rho-kinase signaling. Hypertension 39, 438–442.

Christman, B. W., McPherson, C. D., Newman, J. H., King, G. A., Bernard, G. R., Groves, B. M.,et al. (1992). An imbalance between the excretion of thromboxane and prostacyclinmetabolites in pulmonary hypertension. N Engl J Med 327, 70–75.

D'Alonzo, G. E., Barst, R. J., Ayres, S. M., Bergofsky, E. H., Brundage, B. H., Detre, K. M.,et al. (1991). Survival in patients with primary pulmonary hypertension. Resultsfrom a national prospective registry. Ann Intern Med 115, 343–349.

Dahal, B. K., Kosanovic, D., Pamarthi, P. K., Sydykov, A., Lai, Y. -J., Kast, R., et al. (2010).Therapeutic efficacy of azaindole-1 in experimental pulmonary hypertension. EurRespir J 36, 808–818.

Dai, Z. -K., Wu, B. -N., Chen, I. -C., Chai, C. -Y., Wu, J. -R., Chou, S. -H., et al. (2011). At-tenuation of pulmonary hypertension secondary to left ventricular dysfunction inthe rat by Rho-kinase inhibitor fasudil. Pediatr Pulmonol 46, 45–59.

Davies, S. P., Reddy, H., Caivano, M., & Cohen, P. (2000). Specificity and mechanism ofaction of some commonly used protein kinase inhibitors. Biochem J 351, 95–105.

Desbuards, N., Antier, D., Rochefort, G. Y., Apfeldorfer, C. S., Schenck, E., Hanton, G., et al.(2009). Dexfenfluramine discontinuous treatment does not worsen hypoxia-inducedpulmonary vascular remodeling but activates RhoA/ROCK pathway: Consequences onpulmonary hypertension. Eur J Pharmacol 602, 355–363.

Dingemans, K. P., & Wagenvoort, C. A. (1978). Pulmonary arteries and veins in experi-mental hypoxia. An ultrastructural study. Am J Pathol 93, 353–368.

Do, e Z., Fukumoto, Y., Takaki, A., Tawara, S., Ohashi, J., Nakano, M., et al. (2009). Evi-dence for Rho-kinase activation in patients with pulmonary arterial hypertension.Circ J 73, 1731–1739.

Doran, J. D., Liu, X., Taslimi, P., Saadat, A., & Fox, T. (2004). New insights into thestructure-function relationships of Rho-associated kinase: A thermodynamic and hy-drodynamic study of the dimer-to-monomer transition and its kinetic implications.Biochem J 384, 255–262.

Dorfmüller, P., Perros, F., Balabanian, K., & Humbert, M. (2003). Inflammation in pul-monary arterial hypertension. Eur Respir J 22, 358–363.

Eddahibi, S., Chaouat, A., Morrell, N., Fadel, E., Fuhrman, C., Bugnet, A. -S., et al. (2003).Polymorphism of the serotonin transporter gene and pulmonary hypertension inchronic obstructive pulmonary disease. Circulation 108, 1839–1844.

Eddahibi, S., Humbert, M., Fadel, E., Raffestin, B., Darmon, M., Capron, F., et al. (2001). Sero-tonin transporter overexpression is responsible for pulmonary artery smooth musclehyperplasia in primary pulmonary hypertension. J Clin Invest 108, 1141–1150.

Emerson, M., Momi, S., Paul, W., Alberti, P. F., Page, C., & Gresele, P. (1999). Endogenousnitric oxide acts as a natural antithrombotic agent in vivo by inhibiting platelet ag-gregation in the pulmonary vasculature. Thromb Haemost 81, 961–966.

Eto, M., Senba, S., Morita, F., & Yazawa, M. (1997). Molecular cloning of a novelphosphorylation-dependent inhibitory protein of protein phosphatase-1 (CPI17) insmooth muscle: Its specific localization in smooth muscle. FEBS Lett 410, 356–360.

Fagan, K. A., Fouty, B. W., Tyler, R. C., Morris, K. G., Jr., Hepler, L. K., Sato, K., et al. (1999).The pulmonary circulation of homozygous or heterozygous eNOS-null mice ishyperresponsive to mild hypoxia. J Clin Invest 103, 291–299.

Fagan, K. A., Oka, M., Bauer, N. R., Gebb, S. A., Ivy, D. D., Morris, K. G., et al. (2004). At-tenuation of acute hypoxic pulmonary vasoconstriction and hypoxic pulmonaryhypertension in mice by inhibition of Rho-kinase. Am J Physiol Lung Cell Mol Physiol287, L656–L664.

Feng, J., Ito, M., Ichikawa, K., Isaka, N., Nishikawa, M., Hartshorne, D. J., et al. (1999a). Inhib-itory phosphorylation site for Rho-associated kinase on smooth muscle myosin phos-phatase. J Biol Chem 274, 37385–37390.

Feng, J., Ito, M., Kureishi, Y., Ichikawa, K., Amano, M., Isaka, N., et al. (1999b). Rho-associatedkinase of chicken gizzard smooth muscle. J Biol Chem 274, 3744–3752.

Foletta, V. C., Lim, M. A., Soosairajah, J., Kelly, A. P., Stanley, E. G., Shannon, M., et al.(2003). Direct signaling by the BMP type II receptor via the cytoskeletal regulatorLIMK1. J Cell Biol 162, 1089–1098.

Fujisawa, K., Madaule, P., Ishizaki, T., Watanabe, G., Bito, H., Saito, Y., et al. (1998). Dif-ferent regions of Rho determine Rho-selective binding of different classes of Rhotarget molecules. J Biol Chem 273, 18943–18949.

Fujita, H., Fukumoto, Y., Saji, K., Sugimura, K., Demachi, J., Nawata, J., et al. (2010). Acutevasodilator effects of inhaled fasudil, a specific Rho-kinase inhibitor, in patientswith pulmonary arterial hypertension. Heart Vessels 25, 144–149.

Fukata, Y., Amano, M., & Kaibuchi, K. (2001). Rho-Rho-kinase pathway in smooth musclecontraction and cytoskeletal reorganization of non-muscle cells. Trends Pharmacol Sci22, 32–39.

Fukumoto, Y., Matoba, T., Ito, A., Tanaka, H., Kishi, T., Hayashidani, S., et al. (2005).Acute vasodilator effects of a Rho-kinase inhibitor, fasudil, in patients with severepulmonary hypertension. Heart 91, 391–392.

362 S. Duong-Quy et al. / Pharmacology & Therapeutics 137 (2013) 352–364

Furuya, Y., Satoh, T., & Kuwana, M. (2010). Interleukin-6 as a potential therapeutic tar-get for pulmonary arterial hypertension. Int J Rheumatol 2010, 720305.

Galiè, N., Hoeper, M. M., Humbert, M., Torbicki, A., Vachiery, J. -L., Barbera, J. A., et al. (2009).Guidelines for the diagnosis and treatment of pulmonary hypertension. Eur Respir J 34,1219–1263.

Galié, N., Manes, A., & Branzi, A. (2004). The endothelin system in pulmonary arterialhypertension. Cardiovasc Res 61, 227–237.

Giaid, A., & Saleh, D. (1995). Reduced expression of endothelial nitric oxide synthase inthe lungs of patients with pulmonary hypertension. N Engl J Med 333, 214–221.

Gien, J., Seedorf, G. J., Balasubramaniam, V., Tseng, N., Markham, N., & Abman, S. H.(2008). Chronic intrauterine pulmonary hypertension increases endothelial cellRho kinase activity and impairs angiogenesis in vitro. Am J Physiol Lung Cell MolPhysiol 295, L680–L687.

Girgis, R. E., Li, D., Zhan, X., Garcia, J. G. N., Tuder, R. M., Hassoun, P. M., et al. (2003).Attenuation of chronic hypoxic pulmonary hypertension by simvastatin. Am J Phys-iol Heart Circ Physiol 285, H938–H945.

Girgis, R. E., Mozammel, S., Champion, H. C., Li, D., Peng, X., Shimoda, L., et al. (2007). Regres-sion of chronic hypoxic pulmonary hypertension by simvastatin. Am J Physiol Lung CellMol Physiol 292, L1105–L1110.

Guilluy, C., Eddahibi, S., Agard, C., Guignabert, C., Izikki, M., Tu, L., et al. (2009). RhoAand Rho kinase activation in human pulmonary hypertension: role of 5-HT signal-ing. Am J Respir Crit Care Med 179, 1151–1158.

Guilluy, C., Sauzeau, V., Rolli-Derkinderen, M., Guérin, P., Sagan, C., Pacaud, P., et al.(2005). Inhibition of RhoA/Rho kinase pathway is involved in the beneficial effectof sildenafil on pulmonary hypertension. Br J Pharmacol 146, 1010–1018.

Hassoun, P. M., Mouthon, L., Barberà, J. A., Eddahibi, S., Flores, S. C., Grimminger, F., et al.(2009). Inflammation, growth factors, and pulmonary vascular remodeling. J AmColl Cardiol 54(1 Suppl.), S10–S19.

Hatano, S., & Strasser, T. (1975). Primary pulmonary hypertension. Report on a WHOMeeting. October 15–17, 1973 (pp. 1–45). Geneva: World Health Organization.

Hemnes, A. R., Zaiman, A., & Champion, H. C. (2008). PDE5A inhibition attenuatesbleomycin-induced pulmonary fibrosis and pulmonary hypertension through inhi-bition of ROS generation and RhoA/Rho kinase activation. Am J Physiol Lung Cell MolPhysiol 294, L24–L33.

Hess, D. T., Matsumoto, A., Nudelman, R., & Stamler, J. S. (2001). S-nitrosylation: spec-trum and specificity. Nat Cell Biol 3, E46–E49.

Higenbottam, T. W., & Laude, E. A. (1998). Endothelial dysfunction providing the basis forthe treatment of pulmonary hypertension: Giles F. Filley lecture. Chest 114(1 Suppl.),72S–79S.

Hoeper, M. M., & Dinh-Xuan, A. T. (2004). Combination therapy for pulmonary arterialhypertension: still more questions than answers. Eur Respir J 24, 339–340.

Homma, N., Nagaoka, T., Karoor, V., Imamura, M., Taraseviciene-Stewart, L., Walker,L. A., et al. (2008). Involvement of RhoA/Rho kinase signaling in protectionagainst monocrotaline-induced pulmonary hypertension in pneumonectomizedrats by dehydroepiandrosterone. Am J Physiol Lung Cell Mol Physiol 295, L71–L78.

Homma, N., Nagaoka, T., Morio, Y., Ota, H., Gebb, S. A., Karoor, V., et al. (2007).Endothelin-1 and serotonin are involved in activation of RhoA/Rho kinase sig-naling in the chronically hypoxic hypertensive rat pulmonary circulation.J Cardiovasc Pharmacol 50, 697–702.

Humbert, M., Monti, G., Brenot, F., Sitbon, O., Portier, A., Grangeot-Keros, L., et al.(1995). Increased interleukin-1 and interleukin-6 serum concentrations in severeprimary pulmonary hypertension. Am J Respir Crit Care Med 151, 1628–1631.

Humbert, M., Morrell, N. W., Archer, S. L., Stenmark, K. R., MacLean, M. R., Lang, I. M.,et al. (2004). Cellular and molecular pathobiology of pulmonary arterial hyperten-sion. J Am Coll Cardiol 43(12 Suppl. S), 13S–24S.

Isern, R. A., Yaneva, M., Weiner, E., Parke, A., Rothfield, N., Dantzker, D., et al. (1992).Autoantibodies in patients with primary pulmonary hypertension: associationwith anti-Ku. Am J Med 93, 307–312.

Ishikura, K., Yamada, N., Ito, M., Ota, S., Nakamura, M., Isaka, N., et al. (2006). Beneficialacute effects of rho-kinase inhibitor in patients with pulmonary arterial hyperten-sion. Circ J 70, 174–178.

Ishizaki, T., Maekawa, M., Fujisawa, K., Okawa, K., Iwamatsu, A., Fujita, A., et al. (1996).The small GTP-binding protein Rho binds to and activates a 160 kDa Ser/Thr pro-tein kinase homologous to myotonic dystrophy kinase. EMBO J 15, 1885–1893.

Ishizaki, T., Uehata, M., Tamechika, I., Keel, J., Nonomura, K., Maekawa, M., et al. (2000).Pharmacological properties of Y-27632, a specific inhibitor of rho-associated ki-nases. Mol Pharmacol 57, 976–983.

Ito, K., Ichiki, T., Ohi, K., Egashira, K., Ohta, M., Taguchi, K., et al. (2003). Pulmonary cap-illary hemangiomatosis with severe pulmonary hypertension. Circ J 67, 793–795.

Jasmin, J. F., Lucas, M., Cernacek, P., & Dupuis, J. (2001). Effectiveness of a nonselectiveET(A/B) and a selective ET(A) antagonist in rats with monocrotaline-induced pul-monary hypertension. Circulation 103, 314–318.

Jeffery, T. K., & Wanstall, J. C. (2001). Pulmonary vascular remodeling: A target for ther-apeutic intervention in pulmonary hypertension. Pharmacol Ther 92, 1–20.

Jiang, B. H., Tawara, S., Abe, K., Takaki, A., Fukumoto, Y., & Shimokawa, H. (2007). Acutevasodilator effect of fasudil, a Rho-kinase inhibitor, in monocrotaline-induced pul-monary hypertension in rats. J Cardiovasc Pharmacol 49, 85–89.

Jones, R., Jacobson, M., & Steudel, W. (1999). alpha-smooth-muscle actin and microvas-cular precursor smooth-muscle cells in pulmonary hypertension. Am J Respir CellMol Biol 20, 582–594.

Kamiyama, M., Utsunomiya, K., Taniguchi, K., Yokota, T., Kurata, H., Tajima, N., et al. (2003).Contribution of Rho A and Rho kinase to platelet-derived growth factor-BB-inducedproliferation of vascular smooth muscle cells. J Atheroscler Thromb 10, 117–123.

Kanda, T., Hayashi, K., Wakino, S., Homma, K., Yoshioka, K., Hasegawa, K., et al. (2005).Role of Rho-kinase and p27 in angiotensin II-induced vascular injury. Hypertension45, 724–729.

Kawabata, S., Usukura, J., Morone, N., Ito, M., Iwamatsu, A., Kaibuchi, K., et al. (2004).Interaction of Rho-kinase with myosin II at stress fibres. Genes Cells 9, 653–660.

Keil, A., Blom, I. E., Goldschmeding, R., & Rupprecht, H. D. (2002). Nitric oxidedown-regulates connective tissue growth factor in rat mesangial cells. KidneyInt 62, 401–411.

Khan, I., Oriowo, M. A., Chandrasekhar, B., & Kadavil, E. A. (2005). Attenuatednoradrenaline-induced contraction of pulmonary arteries from rats treatedwith monocrotaline: Role of rho kinase. J Vasc Res 42, 433–440.

Knock, G. A., Snetkov, V. A., Shaifta, Y., Connolly, M., Drndarski, S., Noah, A., et al.(2009). Superoxide constricts rat pulmonary arteries via Rho-kinase-mediatedCa(2+) sensitization. Free Radic Biol Med 46, 633–642.

Koyama, M., Ito, M., Feng, J., Seko, T., Shiraki, K., Takase, K., et al. (2000). Phosphoryla-tion of CPI-17, an inhibitory phosphoprotein of smooth muscle myosin phospha-tase, by Rho-kinase. FEBS Lett 475, 197–200.

Krepinsky, J. C., Ingram, A. J., Tang, D., Wu, D., Liu, L., & Scholey, J. W. (2003). Nitricoxide inhibits stretch-induced MAPK activation in mesangial cells through RhoAinactivation. J Am Soc Nephrol 14, 2790–2800.

Lane, K. B., Machado, R. D., Pauciulo, M. W., Thomson, J. R., Phillips, J. A., III, Loyd, J. E.,et al. (2000). Heterozygous germline mutations in BMPR2, encoding a TGF-betareceptor, cause familial primary pulmonary hypertension. Nat Genet 26, 81–84.

Laufs, U., Endres, M., & Liao, J. K. (1999a). Regulation of endothelial NO production byRho GTPase. Med Klin (Munich) 94, 211–218.

Laufs, U., & Liao, J. K. (1998). Post-transcriptional regulation of endothelial nitric oxidesynthase mRNA stability by Rho GTPase. J Biol Chem 273, 24266–24271.

Laufs, U., Marra, D., Node, K., & Liao, J. K. (1999b). 3-Hydroxy-3-methylglutaryl-CoA re-ductase inhibitors attenuate vascular smooth muscle proliferation by preventingrho GTPase-induced down-regulation of p27(Kip1). J Biol Chem 274, 21926–21931.

Leung, T., Chen, X. Q., Manser, E., & Lim, L. (1996). The p160 RhoA-binding kinase ROKalpha is a member of a kinase family and is involved in the reorganization of thecytoskeleton. Mol Cell Biol 16, 5313–5327.

Leung, T., Manser, E., Tan, L., & Lim, L. (1995). A novel serine/threonine kinase bindingthe Ras-related RhoA GTPase which translocates the kinase to peripheral mem-branes. J Biol Chem 270, 29051–29054.

Li, X. H., Peng, J., Tan, N., Wu, W. H., Li, T. T., Shi, R. Z., et al. (2010). Involvement ofasymmetric dimethylarginine and Rho kinase in the vascular remodeling inmonocrotaline-induced pulmonary hypertension. Vascul Pharmacol 53, 223–229.

Li, F., Xia, W., Li, A., Zhao, C., & Sun, R. (2007). Long-term inhibition of Rho kinase withfasudil attenuates high flow induced pulmonary artery remodeling in rats.Pharmacol Res 55, 64–71.

Li, F., Xia, W., Yuan, S., & Sun, R. (2009). Acute inhibition of Rho-kinase attenuates pulmo-nary hypertension in patients with congenital heart disease. Pediatr Cardiol 30, 363–366.

Liao, J. K., & Laufs, U. (2005). Pleiotropic effects of statins. Annu Rev Pharmacol Toxicol 45,89–118.

Loirand, G., Guérin, P., & Pacaud, P. (2006). Rho kinases in cardiovascular physiologyand pathophysiology. Circ Res 98, 322–334.

Machado, R. D., Aldred, M. A., James, V., Harrison, R. E., Patel, B., Schwalbe, E. C., et al.(2006). Mutations of the TGF-beta type II receptor BMPR2 in pulmonary arterialhypertension. Hum Mutat 27, 121–132.

Machado, R. D., Eickelberg, O., Elliott, C. G., Geraci, M. W., Hanaoka, M., Loyd, J. E., et al.(2009). Genetics and genomics of pulmonary arterial hypertension. J Am CollCardiol 54(1 Suppl.), S32–S42.

Mahapatra, S., Nishimura, R. A., Oh, J. K., & McGoon, M. D. (2006a). The prognostic valueof pulmonary vascular capacitance determined by Doppler echocardiography inpatients with pulmonary arterial hypertension. J Am Soc Echocardiogr 19,1045–1050.

Mahapatra, S., Nishimura, R. A., Sorajja, P., Cha, S., & McGoon, M. D. (2006b). Relation-ship of pulmonary arterial capacitance and mortality in idiopathic pulmonary arte-rial hypertension. J Am Coll Cardiol 47, 799–803.

Matsui, T., Amano, M., Yamamoto, T., Chihara, K., Nakafuku, M., Ito, M., et al. (1996).Rho-associated kinase, a novel serine/threonine kinase, as a putative target forsmall GTP binding protein Rho. EMBO J 15, 2208–2216.

McNamara, P. J., Murthy, P., Kantores, C., Teixeira, L., Engelberts, D., van Vliet, T., et al.(2008). Acute vasodilator effects of Rho-kinase inhibitors in neonatal rats with pul-monary hypertension unresponsive to nitric oxide. Am J Physiol Lung Cell Mol Phys-iol 294, L205–L213.

Moncada, S., Palmer, R. M., & Higgs, E. A. (1989). Biosynthesis of nitric oxide fromL-arginine. A pathway for the regulation of cell function and communication.Biochem Pharmacol 38, 1709–1715.

Montani, D., Price, L. C., Dorfmuller, P., Achouh, L., Jaïs, X., Yaïci, A., et al. (2009). Pulmo-nary veno-occlusive disease. Eur Respir J 33, 189–200.

Moosmang, S., Schulla, V., Welling, A., Feil, R., Feil, S., Wegener, J. W., et al. (2003).Dominant role of smooth muscle L-type calcium channel Cav1.2 for blood pressureregulation. EMBO J 22, 6027–6034.

Morgan, D. O. (1995). Principles of CDK regulation. Nature 374, 131–134.Morrell, N. W., Adnot, S., Archer, S. L., Dupuis, J., Jones, P. L., MacLean, M. R., et al.

(2009). Cellular and molecular basis of pulmonary arterial hypertension. J AmColl Cardiol 54(1 Suppl.), S20–S31.

Mouchaers, K. T. B., Schalij, I., de Boer, M. A., Postmus, P. E., van Hinsbergh, V. W. M., vanNieuw Amerongen, G. P., et al. (2010). Fasudil reduces monocrotaline-induced pul-monary arterial hypertension: Comparison with bosentan and sildenafil. Eur RespirJ 36, 800–807.

Naeije, R., & Huez, S. (2007). Expert opinion on available options treating pulmonaryarterial hypertension. Expert Opin Pharmacother 8, 2247–2265.

Nagaoka, T., Fagan, K. A., Gebb, S. A., Morris, K. G., Suzuki, T., Shimokawa, H., et al.(2005). Inhaled Rho kinase inhibitors are potent and selective vasodilators in ratpulmonary hypertension. Am J Respir Crit Care Med 171, 494–499.

363S. Duong-Quy et al. / Pharmacology & Therapeutics 137 (2013) 352–364

Nagaoka, T., Gebb, S. A., Karoor, V., Homma, N., Morris, K. G., McMurtry, I. F., et al.(2006). Involvement of RhoA/Rho kinase signaling in pulmonary hypertension ofthe fawn-hooded rat. J Appl Physiol 100, 996–1002.

Nagaoka, T., Morio, Y., Casanova, N., Bauer, N., Gebb, S., McMurtry, I., et al. (2004).Rho/Rho kinase signaling mediates increased basal pulmonary vascular tone inchronically hypoxic rats. Am J Physiol Lung Cell Mol Physiol 287, L665–L672.

Nakagawa, O., Fujisawa, K., Ishizaki, T., Saito, Y., Nakao, K., & Narumiya, S. (1996).ROCK-I and ROCK-II, two isoforms of Rho-associated coiled-coil forming proteinserine/threonine kinase in mice. FEBS Lett 392, 189–193.

Newman, J. H., Trembath, R. C., Morse, J. A., Grunig, E., Loyd, J. E., Adnot, S., et al. (2004).Genetic basis of pulmonary arterial hypertension: current understanding and fu-ture directions. J Am Coll Cardiol 43(12 Suppl. S), 33S–39S.

Newman, J. H., Wheeler, L., Lane, K. B., Loyd, E., Gaddipati, R., Phillips, J. A., III, et al.(2001). Mutation in the gene for bone morphogenetic protein receptor II as acause of primary pulmonary hypertension in a large kindred. N Engl J Med 345,319–324.

Ni, W., Egashira, K., Kataoka, C., Kitamoto, S., Koyanagi, M., Inoue, S., et al. (2001).Antiinflammatory and antiarteriosclerotic actions of HMG-CoA reductase inhibi-tors in a rat model of chronic inhibition of nitric oxide synthesis. Circ Res 89,415–421.

Nishimura, T., Vaszar, L. T., Faul, J. L., Zhao, G., Berry, G. J., Shi, L., et al. (2003). Simvastat-in rescues rats from fatal pulmonary hypertension by inducing apoptosis ofneointimal smooth muscle cells. Circulation 108, 1640–1645.

Oka, M., Homma, N., Taraseviciene-Stewart, L., Morris, K. G., Kraskauskas, D., Burns, N.,et al. (2007). Rho kinase-mediated vasoconstriction is important in severe occlu-sive pulmonary arterial hypertension in rats. Circ Res 100, 923–929.

Olschewski, H., Simonneau, G., Galiè, N., Higenbottam, T., Naeije, R., Rubin, L. J., et al.(2002). Inhaled iloprost for severe pulmonary hypertension. N Engl J Med 347,322–329.

Palevsky, H. I., Schloo, B. L., Pietra, G. G., Weber, K. T., Janicki, J. S., Rubin, E., et al. (1989).Primary pulmonary hypertension. Vascular structure, morphometry, and respon-siveness to vasodilator agents. Circulation 80, 1207–1221.

Perros, F., Dorfmüller, P., Souza, R., Durand-Gasselin, I., Godot, V., Capel, F., et al. (2007a).Fractalkine-induced smooth muscle cell proliferation in pulmonary hypertension. EurRespir J 29, 937–943.

Perros, F., Dorfmüller, P., Souza, R., Durand-Gasselin, I., Mussot, S., Mazmanian, M., et al.(2007b). Dendritic cell recruitment in lesions of human and experimental pulmo-nary hypertension. Eur Respir J 29, 462–468.

Piddini, E., Schmid, J. A., de Martin, R., & Dotti, C. G. (2001). The Ras-like GTPase Gem isinvolved in cell shape remodelling and interacts with the novel kinesin-like pro-tein KIF9. EMBO J 20, 4076–4087.

Pierson, D. J. (2000). Pathophysiology and clinical effects of chronic hypoxia. RespirCare 45, 39–51 (discussion 51–53).

Pietra, G. G., Capron, F., Stewart, S., Leone, O., Humbert, M., Robbins, I. M., et al. (2004). Path-ologic assessment of vasculopathies in pulmonary hypertension. J Am Coll Cardiol 43(12Suppl. S), 25S–32S.

Rabinovitch, M., Gamble, W., Nadas, A. S., Miettinen, O. S., & Reid, L. (1979). Rat pulmo-nary circulation after chronic hypoxia: Hemodynamic and structural features. Am JPhysiol 236, H818–H827.

Riento, K., Guasch, R. M., Garg, R., Jin, B., & Ridley, A. J. (2003). RhoE binds to ROCK I andinhibits downstream signaling. Mol Cell Biol 23, 4219–4229.

Riento, K., & Ridley, A. J. (2003). Rocks: multifunctional kinases in cell behaviour. NatRev Mol Cell Biol 4, 446–456.

Rikitake, Y., Kim, H. -H., Huang, Z., Seto, M., Yano, K., Asano, T., et al. (2005). Inhibitionof Rho kinase (ROCK) leads to increased cerebral blood flow and stroke protection.Stroke 36, 2251–2257.

Rikitake, Y., & Liao, J. K. (2005). Rho GTPases, statins, and nitric oxide. Circ Res 97,1232–1235.

Rubin, L. J., Badesch, D. B., Barst, R. J., Galie, N., Black, C. M., Keogh, A., et al. (2002).Bosentan therapy for pulmonary arterial hypertension. N Engl J Med 346, 896–903.

Sakao, S., Taraseviciene-Stewart, L., Cool, C. D., Tada, Y., Kasahara, Y., Kurosu, K., et al.(2007). VEGF-R blockade causes endothelial cell apoptosis, expansion of survivingCD34+ precursor cells and transdifferentiation to smooth muscle-like andneuronal-like cells. FASEB J 21, 3640–3652.

Sakurada, S., Okamoto, H., Takuwa, N., Sugimoto, N., & Takuwa, Y. (2001). Rho activa-tion in excitatory agonist-stimulated vascular smooth muscle. Am J Physiol CellPhysiol 281, C571–C578.

Sanchez, O., Marcos, E., Perros, F., Fadel, E., Tu, L., Humbert, M., et al. (2007). Role ofendothelium-derived CC chemokine ligand 2 in idiopathic pulmonary arterial hy-pertension. Am J Respir Crit Care Med 176, 1041–1047.

Sastry, B. K. S., Narasimhan, C., Reddy, N. K., & Raju, B. S. (2004). Clinical efficacy of sil-denafil in primary pulmonary hypertension: A randomized, placebo-controlled,double-blind, crossover study. J Am Coll Cardiol 43, 1149–1153.

Sata, M. (2006). Role of circulating vascular progenitors in angiogenesis, vascularhealing, and pulmonary hypertension: Lessons from animal models. ArteriosclerThromb Vasc Biol 26, 1008–1014.

Satoh, S., Utsunomiya, T., Tsurui, K., Kobayashi, T., Ikegaki, I., Sasaki, Y., et al. (2001).Pharmacological profile of hydroxy fasudil as a selective rho kinase inhibitor on is-chemic brain damage. Life Sci 69, 1441–1453.

Sauzeau, V., Le Jeune, H., Cario-Toumaniantz, C., Smolenski, A., Lohmann, S. M.,Bertoglio, J., et al. (2000). Cyclic GMP-dependent protein kinase signaling pathwayinhibits RhoA-induced Ca2+ sensitization of contraction in vascular smooth mus-cle. J Biol Chem 275, 21722–21729.

Sauzeau, V., Le Mellionnec, E., Bertoglio, J., Scalbert, E., Pacaud, P., & Loirand, G. (2001).Human urotensin II-induced contraction and arterial smooth muscle cell prolifera-tion are mediated by RhoA and Rho-kinase. Circ Res 88, 1102–1104.

Sawada, N., Itoh, H., Ueyama, K., Yamashita, J., Doi, K., Chun, T. H., et al. (2000). Inhibi-tion of rho-associated kinase results in suppression of neointimal formation ofballoon-injured arteries. Circulation 101, 2030–2033.

Seasholtz, T. M., Majumdar, M., Kaplan, D. D., & Brown, J. H. (1999). Rho and Rho kinasemediate thrombin-stimulated vascular smooth muscle cell DNA synthesis and mi-gration. Circ Res 84, 1186–1193.

Sebbagh,M., Hamelin, J., Bertoglio, J., Solary, E., & Bréard, J. (2005). Direct cleavage of ROCK IIby granzyme B induces target cell membrane blebbing in a caspase-independent man-ner. J Exp Med 201, 465–471.

Sebbagh, M., Renvoizé, C., Hamelin, J., Riché, N., Bertoglio, J., & Bréard, J. (2001).Caspase-3-mediated cleavage of ROCK I induces MLC phosphorylation and apopto-tic membrane blebbing. Nat Cell Biol 3, 346–352.

Sherr, C. J., & Roberts, J. M. (1995). Inhibitors of mammalian G1 cyclin-dependent ki-nases. Genes Dev 9, 1149–1163.

Shimokawa, H. (2002). Rho-kinase as a novel therapeutic target in treatment of cardio-vascular diseases. J Cardiovasc Pharmacol 39, 319–327.

Shimokawa, H., Hiramori, K., Iinuma, H., Hosoda, S., Kishida, H., Osada, H., et al. (2002).Anti-anginal effect of fasudil, a Rho-kinase inhibitor, in patients with stable effortangina: A multicenter study. J Cardiovasc Pharmacol 40, 751–761.

Shimokawa, H., Seto, M., Katsumata, N., Amano, M., Kozai, T., Yamawaki, T., et al. (1999).Rho-kinase-mediated pathway induces enhanced myosin light chain phosphorylationsin a swine model of coronary artery spasm. Cardiovasc Res 43, 1029–1039.

Shirao, S., Kashiwagi, S., Sato, M., Miwa, S., Nakao, F., Kurokawa, T., et al. (2002).Sphingosylphosphorylcholine is a novel messenger for Rho-kinase-mediatedCa2+ sensitization in the bovine cerebral artery: unimportant role for proteinkinase C. Circ Res 91, 112–119.

Simonneau, G., Galiè, N., Rubin, L. J., Langleben, D., Seeger, W., Domenighetti, G., et al.(2004). Clinical classification of pulmonary hypertension. J Am Coll Cardiol 43(12Suppl. S), 5S–12S.

Sin, W. C., Chen, X. Q., Leung, T., & Lim, L. (1998). RhoA-binding kinase alpha transloca-tion is facilitated by the collapse of the vimentin intermediate filament network.Mol Cell Biol 18, 6325–6339.

Sitbon, O., Humbert, M., Nunes, H., Parent, F., Garcia, G., Hervé, P., et al. (2002). Long-termintravenous epoprostenol infusion in primary pulmonary hypertension: prognostic fac-tors and survival. J Am Coll Cardiol 40, 780–788.

Somlyo, A. P., & Somlyo, A. V. (2003). Ca2+ sensitivity of smooth muscle and nonmusclemyosin II: Modulated by G proteins, kinases, and myosin phosphatase. Physiol Rev 83,1325–1358.

Sommer, N., Dietrich, A., Schermuly, R. T., Ghofrani, H. A., Gudermann, T., Schulz, R.,et al. (2008). Regulation of hypoxic pulmonary vasoconstriction: Basic mecha-nisms. Eur Respir J 32, 1639–1651.

Stamler, J. S., Lamas, S., & Fang, F. C. (2001). Nitrosylation. The prototypic redox-basedsignaling mechanism. Cell 106, 675–683.

Steiner, M. K., Syrkina, O. L., Kolliputi, N., Mark, E. J., Hales, C. A., & Waxman, A. B.(2009). Interleukin-6 overexpression induces pulmonary hypertension. Circ Res104, 236–244 (28 pp. following 244).

Sztrymf, B., Coulet, F., Girerd, B., Yaici, A., Jais, X., Sitbon, O., et al. (2008). Clinical out-comes of pulmonary arterial hypertension in carriers of BMPR2 mutation. Am JRespir Crit Care Med 177, 1377–1383.

Takahashi, N., Tuiki, H., Saya, H., & Kaibuchi, K. (1999). Localization of the gene codingfor ROCK II/Rho kinase on human chromosome 2p24. Genomics 55, 235–237.

Takemoto, M., Sun, J., Hiroki, J., Shimokawa, H., & Liao, J. K. (2002). Rho-kinase medi-ates hypoxia-induced downregulation of endothelial nitric oxide synthase. Circula-tion 106, 57–62.

Takizawa, N., Koga, Y., & Ikebe, M. (2002). Phosphorylation of CPI17 and myosin bind-ing subunit of type 1 protein phosphatase by p21-activated kinase. BiochemBiophys Res Commun 297, 773–778.

Tamby, M. C., Chanseaud, Y., Humbert, M., Fermanian, J., Guilpain, P., Garcia-de-la-Peña-Lefebvre, P., et al. (2005). Anti-endothelial cell antibodies in idio-pathic and systemic sclerosis associated pulmonary arterial hypertension.Thorax 60, 765–772.

Tawara, S., Fukumoto, Y., & Shimokawa, H. (2007). Effects of combined therapy with aRho-kinase inhibitor and prostacyclin on monocrotaline-induced pulmonary hy-pertension in rats. J Cardiovasc Pharmacol 50, 195–200.

Toshima, Y., Satoh, S., Ikegaki, I., & Asano, T. (2000). A new model of cerebralmicrothrombosis in rats and the neuroprotective effect of a Rho-kinase inhibitor.Stroke 31, 2245–2250.

Trembath, R. C., Thomson, J. R., Machado, R. D., Morgan, N. V., Atkinson, C., Winship, I.,et al. (2001). Clinical and molecular genetic features of pulmonary hypertension inpatients with hereditary hemorrhagic telangiectasia. N Engl J Med 345, 325–334.

Tuder, R. M., Abman, S. H., Braun, T., Capron, F., Stevens, T., Thistlethwaite, P. A., et al.(2009). Development and pathology of pulmonary hypertension. J Am Coll Cardiol54(1 Suppl.), S3–S9.

Tuder, R. M., Cool, C. D., Geraci, M. W., Wang, J., Abman, S. H., Wright, L., et al. (1999).Prostacyclin synthase expression is decreased in lungs from patients with severepulmonary hypertension. Am J Respir Crit Care Med 159, 1925–1932.

Tuder, R. M., Groves, B., Badesch, D. B., & Voelkel, N. F. (1994). Exuberant endothelialcell growth and elements of inflammation are present in plexiform lesions of pul-monary hypertension. Am J Pathol 144, 275–285.

Tuder, R. M., & Voelkel, N. F. (1998). Pulmonary hypertension and inflammation. J LabClin Med 132, 16–24.

Tuder, R. M., & Zaiman, A. L. (2004). Pathology of pulmonary vascular disease. In A. Peacock,& L. J. Rubin (Eds.), Pulmonary circulation (pp. 25–32). (2nd ed.). London: Arnold.

Uehata, M., Ishizaki, T., Satoh, H., Ono, T., Kawahara, T., Morishita, T., et al. (1997). Cal-cium sensitization of smooth muscle mediated by a Rho-associated protein kinasein hypertension. Nature 389, 990–994.

364 S. Duong-Quy et al. / Pharmacology & Therapeutics 137 (2013) 352–364

VanBavel, E., Sorop, O., Andreasen, D., Pfaffendorf, M., & Jensen, B. L. (2002). Role ofT-type calcium channels in myogenic tone of skeletal muscle resistance arteries.Am J Physiol Heart Circ Physiol 283, H2239–H2243.

Vanderpool, R. R., Kim, A. R., Molthen, R., & Chesler, N. C. (2011). Effects of acute Rhokinase inhibition on chronic hypoxia-induced changes in proximal and distal pul-monary arterial structure and function. J Appl Physiol 110, 188–198.

Velasco, G., Armstrong, C., Morrice, N., Frame, S., & Cohen, P. (2002). Phosphorylation ofthe regulatory subunit of smooth muscle protein phosphatase 1M at Thr850 in-duces its dissociation from myosin. FEBS Lett 527, 101–104.

Voelkel, N. F., Cool, C., Lee, S. D., Wright, L., Geraci, M. W., & Tuder, R. M. (1998). Primarypulmonary hypertension between inflammation and cancer. Chest 114(3 Suppl.),225S–230S.

Wagenvoort, C. A., & Wagenvoort, N. (1970). Primary pulmonary hypertension a path-ologic study of the lung vessels in 156 clinically diagnosed cases. Circulation 42,1163–1184.

Ward, Y., Yap, S. F., Ravichandran, V., Matsumura, F., Ito, M., Spinelli, B., et al. (2002).The GTP binding proteins Gem and Rad are negative regulators of the Rho-Rho ki-nase pathway. J Cell Biol 157, 291–302.

Wibberley, A., Chen, Z., Hu, E., Hieble, J. P., & Westfall, T. D. (2003). Expression andfunctional role of Rho-kinase in rat urinary bladder smooth muscle. Br J Pharmacol138, 757–766.

Wilson, D. W., Segall, H. J., Pan, L. C., & Dunston, S. K. (1989). Progressive inflammatoryand structural changes in the pulmonary vasculature of monocrotaline-treatedrats. Microvasc Res 38, 57–80.

Wilson, D. P., Susnjar, M., Kiss, E., Sutherland, C., & Walsh, M. P. (2005). ThromboxaneA2-induced contraction of rat caudal arterial smooth muscle involves activation ofCa2+ entry and Ca2+ sensitization: Rho-associated kinase-mediated phosphory-lation of MYPT1 at Thr-855, but not Thr-697. Biochem J 389, 763–774.

Wilson, D. P., Sutherland, C., & Walsh, M. P. (2002). Ca2+ activation of smooth musclecontraction: evidence for the involvement of calmodulin that is bound to the tritoninsoluble fraction even in the absence of Ca2+. J Biol Chem 277, 2186–2192.

Wolfrum, S., Dendorfer, A., Rikitake, Y., Stalker, T. J., Gong, Y., Scalia, R., et al. (2004). Inhibi-tion of Rho-kinase leads to rapid activation of phosphatidylinositol 3-kinase/protein ki-nase Akt and cardiovascular protection. Arterioscler Thromb Vasc Biol 24, 1842–1847.

Xu, E. Z., Kantores, C., Ivanovska, J., Engelberts, D., Kavanagh, B. P., McNamara, P. J., et al.(2010). Rescue treatment with a Rho-kinase inhibitor normalizes right ventricularfunction and reverses remodeling in juvenile rats with chronic pulmonary hyper-tension. Am J Physiol Heart Circ Physiol 299, H1854–H1864.

Yamakawa, T., Tanaka, S., Numaguchi, K., Yamakawa, Y., Motley, E. D., Ichihara, S., et al.(2000). Involvement of Rho-kinase in angiotensin II-induced hypertrophy of ratvascular smooth muscle cells. Hypertension 35, 313–318.

Yi, E. S., Kim, H., Ahn, H., Strother, J., Morris, T., Masliah, E., et al. (2000). Distribution ofobstructive intimal lesions and their cellular phenotypes in chronic pulmonary hy-pertension. A morphometric and immunohistochemical study. Am J Respir Crit CareMed 162, 1577–1586.

Yuan, J. X., Aldinger, A. M., Juhaszova, M., Wang, J., Conte, J. V., Jr., Gaine, S. P., et al. (1998).Dysfunctional voltage-gated K+ channels in pulmonary artery smooth muscle cells ofpatients with primary pulmonary hypertension. Circulation 98, 1400–1406.

Yuan, J. X. J., & Rubin, L. J. (2005). Pathogenesis of pulmonary arterial hypertension: theneed for multiple hits. Circulation 111, 534–538.

Zhu, P., Huang, L., Ge, X., Yan, F., Wu, R., & Ao, Q. (2006). Transdifferentiation of pulmo-nary arteriolar endothelial cells into smooth muscle-like cells regulated bymyocardin involved in hypoxia-induced pulmonary vascular remodelling. Int JExp Pathol 87, 463–474.

Ziino, A. J. A., Ivanovska, J., Belcastro, R., Kantores, C., Xu, E. Z., Lau, M., et al. (2010). Ef-fects of rho-kinase inhibition on pulmonary hypertension, lung growth, and struc-ture in neonatal rats chronically exposed to hypoxia. Pediatr Res 67, 177–182.

Zuckerbraun, B. S., Stoyanovsky, D. A., Sengupta, R., Shapiro, R. A., Ozanich, B. A., Rao, J.,et al. (2007). Nitric oxide-induced inhibition of smooth muscle cell proliferationinvolves S-nitrosation and inactivation of RhoA. Am J Physiol Cell Physiol 292,C824–C831.


Recommended