11
Archives of Cardiovascular Disease (2015) 108, 519—529 Available online at ScienceDirect www.sciencedirect.com REVIEW Advancing knowledge of right ventricular pathophysiology in chronic pressure overload: Insights from experimental studies Physiopathologie du ventricule droit dans la surcharge de pression chronique : données expérimentales récentes Julien Guihaire a,b,, Pierre Emmanuel Noly a , Sonja Schrepfer c , Olaf Mercier a a Laboratory of Surgical Research, Marie-Lannelongue Hospital, Paris Sud University, 92350 Le Plessis Robinson, France b Thoracic and Cardiovascular Surgery, University Hospital of Rennes, 35033 Rennes, France c Transplant and Stem Cell Immunobiology Laboratory (TSI Lab), University of Hamburg, Hamburg, Germany Received 8 April 2015; received in revised form 25 May 2015; accepted 26 May 2015 Available online 13 July 2015 KEYWORDS Right ventricle; Pulmonary hypertension; Pulmonary circulation; Summary The right ventricle (RV) has to face major changes in loading conditions due to car- diovascular diseases and pulmonary vascular disorders. Clinical experience supports evidence that the RV better compensates for volume than for pressure overload, and for chronic than for acute changes. For a long time, right ventricular (RV) pathophysiology has been restricted to patterns extrapolated from left heart studies. However, the two ventricles are anatomi- cally, haemodynamically and functionally distinct. RV metabolic properties may also result in Abbreviations: AT, angiotensin; Ea, arterial elastance; Ees, end-systolic elastance; IVA, acceleration of the myocardium during isovolumic contraction; LV, left ventricle/ventricular; MHC, myosin heavy chain; PA, pulmonary arterial; PH, pulmonary hypertension; PVR, pulmonary vascular resistance; RAAS, renin-angiotensin-aldosterone system; RV, right ventricle/ventricular; RVF, right ventricular failure; RVFAC, right ventricular fractional area change; RVMPI, right ventricular myocardial performance index; SVI, stroke volume index; TAPSE, tricuspid annular plane systolic excursion. Corresponding author at: Laboratory of Surgical Research, Marie-Lannelongue Hospital, Paris Sud University, 133, avenue de la Résistance, 92350 Le Plessis-Robinson, France. E-mail address: [email protected] (J. Guihaire). http://dx.doi.org/10.1016/j.acvd.2015.05.008 1875-2136/© 2015 Elsevier Masson SAS. All rights reserved.

Advancing knowledge of right ventricular pathophysiology ...However, the two ventricles are anatomi-cally, haemodynamically and functionally distinct. RV metabolic properties may also

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Advancing knowledge of right ventricular pathophysiology ...However, the two ventricles are anatomi-cally, haemodynamically and functionally distinct. RV metabolic properties may also

Archives of Cardiovascular Disease (2015) 108, 519—529

Available online at

ScienceDirectwww.sciencedirect.com

REVIEW

Advancing knowledge of right ventricularpathophysiology in chronic pressureoverload: Insights from experimentalstudiesPhysiopathologie du ventricule droit dans la surcharge de pression chronique :données expérimentales récentes

Julien Guihairea,b,∗, Pierre Emmanuel Nolya,Sonja Schrepferc, Olaf Merciera

a Laboratory of Surgical Research, Marie-Lannelongue Hospital, Paris Sud University, 92350 LePlessis Robinson, Franceb Thoracic and Cardiovascular Surgery, University Hospital of Rennes, 35033 Rennes, Francec Transplant and Stem Cell Immunobiology Laboratory (TSI Lab), University of Hamburg,Hamburg, Germany

Received 8 April 2015; received in revised form 25 May 2015; accepted 26 May 2015Available online 13 July 2015

KEYWORDSRight ventricle;

Summary The right ventricle (RV) has to face major changes in loading conditions due to car-diovascular diseases and pulmonary vascular disorders. Clinical experience supports evidence

Pulmonaryhypertension;Pulmonarycirculation;

that the RV better compensates for volume than for pressure overload, and for chronic thanfor acute changes. For a long time, right ventricular (RV) pathophysiology has been restrictedto patterns extrapolated from left heart studies. However, the two ventricles are anatomi-cally, haemodynamically and functionally distinct. RV metabolic properties may also result in

Abbreviations: AT, angiotensin; Ea, arterial elastance; Ees, end-systolic elastance; IVA, acceleration of the myocardium during isovolumiccontraction; LV, left ventricle/ventricular; MHC, myosin heavy chain; PA, pulmonary arterial; PH, pulmonary hypertension; PVR, pulmonaryvascular resistance; RAAS, renin-angiotensin-aldosterone system; RV, right ventricle/ventricular; RVF, right ventricular failure; RVFAC, rightventricular fractional area change; RVMPI, right ventricular myocardial performance index; SVI, stroke volume index; TAPSE, tricuspidannular plane systolic excursion.

∗ Corresponding author at: Laboratory of Surgical Research, Marie-Lannelongue Hospital, Paris Sud University, 133, avenue de la Résistance,92350 Le Plessis-Robinson, France.

E-mail address: [email protected] (J. Guihaire).

http://dx.doi.org/10.1016/j.acvd.2015.05.0081875-2136/© 2015 Elsevier Masson SAS. All rights reserved.

Page 2: Advancing knowledge of right ventricular pathophysiology ...However, the two ventricles are anatomi-cally, haemodynamically and functionally distinct. RV metabolic properties may also

520 J. Guihaire et al.

Ventricular-arterialcoupling;Experimentalresearch

a different behaviour in response to pathological conditions compared with the left ventricle.In this review, current knowledge of RV pathophysiology is reported in the setting of chronicpressure overload, including recent experimental findings and emerging concepts. After a time-varying compensated period with preserved cardiac output despite overload conditions, RVfailure finally occurs, leading to death. The underlying mechanisms involved in the transitionfrom compensatory hypertrophy to maladaptive remodelling are not completely understood.© 2015 Elsevier Masson SAS. All rights reserved.

MOTS CLÉSVentricule droit ;Hypertensionpulmonaire ;Circulationpulmonaire ;Couplageventriculo-artériel ;Rechercheexpérimentale

Résumé Le ventricule droit (VD) fait face à d’importantes variations de ses conditions decharge en réponse aux maladies cardiovasculaires et pathologies vasculaires pulmonaires. LeVD supporte mieux une surcharge de volume qu’une surcharge de pression, de même il s’adaptemieux aux variations progressives qu’aux changements aigus. La physiopathologie du VD a pen-dant longtemps été résumée à des extrapolations de la physiopathologie du ventricule gauche.Cependant les deux ventricules sont différents, tant sur le plan anatomique, hémodynamiqueque fonctionnel. Les caractéristiques métaboliques singulières du VD peuvent également êtreà l’origine d’une adaptation différente aux conditions pathologiques. Après une longue périodecompensatrice avec préservation du débit cardiaque, la dysfonction du VD s’installe finale-ment face à la surcharge chronique persistante. Les mécanismes impliqués dans la transitiondepuis l’hypertrophie compensatrice jusqu’au remodelage inadapté sont méconnus. Dans cetterevue, les connaissances actuelles de la physiopathologie du VD dans l’hypertension pulmonairechronique sont reportées à partir des résultats récents issus de la recherche expérimentale.© 2015 Elsevier Masson SAS. Tous droits réservés.

B

Rlf(eilataa(tinacvmR

Ipntatc

dhRfoec[aminotaotmeod

vdmR

ackground

ight heart failure is commonly related to left ventricu-ar (LV) dysfunction. Ischaemic myocardial injury or, lessrequently, arrhythmogenic dysplasia of the right ventricleRV), congenital heart disease and chronic respiratory dis-ase, including pulmonary vascular disorders, can also resultn right ventricular failure (RVF). RVF is also a current prob-em in the post-operative course of heart transplantationnd LV assist device implantation [1,2]. In pulmonary hyper-ension (PH), pulmonary vascular damage will inevitablyffect the whole cardiopulmonary unit [3]. It has beenppreciated over the last two decades that right ventricularRV) dysfunction is the most important determinant of long-erm outcomes in PH patients. Actually, prolonged survivals related more to RV function than to pulmonary haemody-amics per se. In studies addressing haemodynamic variablesnd survival in PH, high mean right atrial pressures and lowardiac output are consistently associated with poorer sur-ival [4—6]. Despite major improvements in pharmacologicalanagement over the last 15 years, PH patients still die from

VF.RV pathophysiology has been overlooked for many years.

n the past, several experiments relegated the RV to aassive conduit, suggesting that the RV pump might beeglected. For example, Starr et al. showed that elec-

rocautery ablation of the RV free wall in dogs was notssociated with significant changes in haemodynamics, andhat all animals survived [7]. For a long time, the Fontan pro-edure also supported the clinical evidence that RV absence

srti

id not compromise overall heart function [8]. Physiciansave recently shown clinical interest in the right heart, asV function appears to be a prognostic factor in left heartailure, PH and other chronic respiratory disorders. Mostf mechanisms involved in left heart diseases were firstxtrapolated to RV pathophysiology, despite the two ventri-les differing in their embryology, geometry and physiology9,10]. Recently, a new focus on RVF has emerged, whichims to improve both understanding and clinical manage-ent. In PH, there are different phenotypic abnormalities

n the RV. Surprisingly, some patients share the same phe-otype, whereas the RV is not exposed to the same degreef pressure overload [11]. Considering the central role ofhe RV in cardiopulmonary diseases, Mehra et al. suggested

comprehensive nomenclature of right heart failure, basedn aetiology, anatomical injury, pathophysiology and func-ional status [12]. Experimentally, major efforts have beenade to improve our understanding of RV remodelling. Sev-

ral animal models of chronic RVF have thus been reportedver the last decade to reproduce the main features of RVysfunction [13].

The mechanisms of RVF as well as the clinically relevantariables for measuring RV function in PH are still beingebated. In this paper, after a brief description of the nor-al RV, we first seek to overview the functional evaluation of

V contractility and reserve in the setting of chronic pres-

ure overload. We then present current knowledge of RVemodelling related to PH. Based on experimental findings,he cellular mechanisms and molecular pathways involvedn the transition from compensated RV hypertrophy to
Page 3: Advancing knowledge of right ventricular pathophysiology ...However, the two ventricles are anatomi-cally, haemodynamically and functionally distinct. RV metabolic properties may also

ad

rabmklciplicatctevv

Ai

ActilRnippahsqsEpvstt

mcfcmPiEEsp

Right ventricular pathophysiology in chronic pressure overlo

dysfunction and failure are discussed. Finally, recent thera-peutic approaches for the failing RV are considered. Isolatedex vivo studies of the RV will not be considered in this review.

The normal RV: embryology, morphologyand physiology

The RV does not originate from the primary heart field,unlike the left ventricle (LV). Recent experiments haveshown that the RV does not share the same origin as the LV,emphasizing the need for a specific comprehensive approachto each ventricle [14—17]. The RV actually originates fromthe secondary heart field, while the atrium and the LVdevelop from the primary heart field. Three separate cham-bers can be described in the RV: the sinus or inlet, whichconsists of the tricuspid valve, chordae tendineae and pap-illary muscles; the trabeculated apical myocardium; and theinfundibulum or conus [10]. Compared with the LV, the RVhas a crescent shape, with a higher capacity and a lowermyocardial mass.

In utero the RV wall is thick because the pulmonaryvascular resistance (PVR) is high. At birth, PVR decreasesdramatically, leading to high compliance, resulting from pro-gressive thinning of the RV free wall [10]. RV contractioncan be understood as a three-step motion. The tricuspidannulus first moves toward the apex, then the apex movestoward the infundibulum. The radial contraction of the RVfree wall towards the interventricular septum is the thirdand minor component of RV contractility; it results frommarked shrinking of the deep muscle fibres. This myofibrelayer is continuous between the LV and the RV; it repre-sents one of the three anatomical elements of ventricularinterdependence, along with the pericardium and the inter-ventricular septum, explaining why the LV is involved in RVcontraction (from 20% to 40% of RV contractile performance)[10].

The RV is exposed to frequent changes in venous return,influenced by respiratory variations, by the supine or ortho-static position of the body and by pathological disordersaffecting filling conditions. The RV is highly distensiblebecause of its larger shape and four times thinner free wallcompared with the LV. The RV is therefore known to adaptmuch better to chronic volume overload than to pressureoverload [18]. An increase in preload has been demonstratedto improve myocardial contraction in the left heart [19].The Frank—Starling mechanism characterizes the increase inventricular stroke work in response to wall stress enhance-ment. Because of a weak relationship between surface areaand ventricular volume, the preload recruitable stroke workis moderate in the physiological RV. The Frank—Starlingmechanism may only improve ventricular contraction in caseof significant enlargement of the RV [18]. On the other hand,excessive volume loading may be detrimental to the RV,because of acute impairment of coronary flow within theRV free wall [20].

RV afterload is defined by pulmonary vascular load. The

pulmonary vasculature is characterized by a low resistanceand high compliance profile. PVR is usually linked to RVafterload in normal conditions because of easy assessmentin clinical practice. However, PVR only accounts for the

ctab

521

esistive component of the pulmonary vascular load; as result, PVR underestimates RV afterload by neglectinglood flow pulsatility and wave reflections through the pul-onary vasculature. Most pulmonary vascular diseases are

nown to disturb both components of the pulmonary vascu-ar load. Pulmonary arterial impedance is probably the mostomplete measure of the pulmonary vascular load. Inputmpedance actually includes both static and dynamic com-onents of RV afterload, and it may be a better predictor ofong-term outcomes in PH than PVR alone [21]; its definitionn the frequency domain is, however, restrictive for widelinical use. Sunagawa et al. characterized the systemicrterial load in the time domain as effective arterial elas-ance (Ea) [22]. Ea is defined as the change in pressure for ahange in volume, and can therefore be linked to the effec-ive arterial load that the ventricle has to overcome duringjection. Pulmonary Ea can be determined using pressure-olume loops analysis or estimated as the ratio of strokeolume to end-systolic pressure.

ssessment of RV pump function: themportance of RV-PA coupling

s RV function is the main determinant of survival in severalardiopulmonary diseases, there is major interest in inves-igating the right heart. Echocardiography is the mainstayn the assessment of RV function. The main limitation is theoad dependency of most of these noninvasive indices. TheV is optimally matched to pulmonary vascular load in theormal state. Afterload mismatch appears to be the mostmportant prognostic factor for RV systolic dysfunction inressure overload conditions [23]. Ventricular-arterial cou-ling is an important determinant of cardiac performancend energetics; it reflects maximal efficiency betweenydraulic work production and myocardial oxygen con-umption. RV-pulmonary arterial (RV-PA) coupling can beuantified in real time, using pressure-volume loops analy-is, as the ratio of the RV end-systolic elastance (Ees) to thea (Fig. 1). Ees is defined as the slope of the end-systolicressure-volume linear relationship obtained by changes inentricular filling [24,25]. To be load-independent, this end-ystolic relationship is studied during transient occlusion ofhe inferior vena cava, so that Ees is currently consideredo be the most reliable measure of ventricular contractility.

Despite significant interspecies differences in basal pul-onary haemodynamics, Wauthy et al. showed that Ees/Ea

hanges similarly, secondary to RV pressure overload, in dif-erent animal species [26]. Furthermore, RV adaptation tohanges in loading conditions is relatively preserved throughammalian spontaneous evolution [26]. Ranges of usual RV-

A coupling values among animal species are summarizedn Table 1. Impairment of RV-PA coupling is reflected in anes/Ea ratio of < 1 in large animals, whereas the normales/Ea ratio ranges from 0.5 to 0.8 in rodents. At the earlytage of RV pressure overload, Ees is often increased to com-ensate for the elevated afterload [27,28]. However, RV-PA

oupling may be decreased at that time, without RV dysfunc-ion at rest. This is related more to an increase in Ea than to

significant decrease in Ees. When contractile performanceecomes insufficient to overcome the elevated afterload,

Page 4: Advancing knowledge of right ventricular pathophysiology ...However, the two ventricles are anatomi-cally, haemodynamically and functionally distinct. RV metabolic properties may also

522 J. Guihaire et al.

Table 1 Right ventricular and pulmonary arterial elastances and ventricular-arterial coupling values among animalspecies in normal loading conditions.

Animals Weight (kg) Methodsa Ees (mmHg/mL) Ea (mmHg/mL) Ees/Ea

Rat [75] 0.20—0.25 Conductance 82 ± 53 150 ± 60 0.53 ± 0.2Mouse [76] 0.02—0.04 Admittance 1.8 ± 0.5 2.7 ± 1.2 0.71 ± 0.27Dog [77] 25 Single-beat 1.11 ± 0.07 1.0 ± 0.06 1.13 ± 0.08Goat [26] 24 Single-beat 2.0 ± 0.4 1.3 ± 0.1 1.7 ± 0.3Piglet [78] 5.7—6.1 Single-beat 1.45 ± 0.08 0.91 ± 0.07 1.62 ± 0.09Pig [79] 20—25 Single-beat 1.4 ± 0.2 0.9 ± 0.1 1.4 ± 0.4

Ea: pulmonary arterial elastance; Ees: right ventricular end-systolic elastance.a Methods of recording pressure-volume loops were specified as conductance or admittance catheterization, or as single-beat elastance.

Figure 1. Pressure-volume loops in the right ventricle.A. Schematic conductance catheterization of the right ventri-cle (RV). B. Example of pressure-volume loops in the normal RV;transient occlusion of the inferior vena cava reduces right ventric-ular preload, leading to progressive decrease of right ventricularstroke volume. Ea: arterial elastance; Ees: end-systolic elastance;RA: right artery.

RdR

miefviRtafs(pmlectirambitmtsihatvtd

fPmpla

V stroke volume and RV ejection fraction decrease. RVilatation and adverse remodelling then occur, leading toVF [27,29].

The concept of ventricular-arterial coupling is not com-only used in clinical practice, mainly because it requires

nvasive and time-consuming measurement. Considerablefforts have been made to estimate the contractile per-ormance of the RV from measurement of RV pressure andolume. Single-beat elastance is a promising method val-dated recently in clinical practice for the assessment ofV elastance without dramatic changes in loading condi-ions, using cardiac magnetic resonance imaging for volumessessment [30—32]. Several echocardiography indices of RVunction have been further reported to characterize theystole, such as tricuspid annular plane systolic excursionTAPSE), RV fractional area change (RVFAC), RV myocardialerformance index (RVMPI), stroke volume index (SVI) and,ore recently, acceleration of the myocardium during isovo-

umic contraction (IVA) [33—36]. Of all these indices, Vogelt al. showed that IVA may be the most relevant for RVontractility [33—36]. There were two main limitations tohis study. First, IVA (as well as TAPSE, RVFAC and RVMPI)s strongly load dependent, and thus may not accuratelyeflect RV pump function. Second, this study validated IVAs a contractility index for the RV in a ‘healthy’ animalodel, not in the setting of chronic RV remodelling causedy either pressure or volume overload. We recently showedn a piglet model of chronic pressure overload for the RV,hat usual noninvasive measures of RV systolic function wereore strongly associated with RV-PA coupling than with con-

ractility (Ees). Despite a marked improvement in Ees, RVystolic dysfunction was observed, with a marked decreasen TAPSE, RVFAC or RVMPI. This is in accordance with ourypothesis that commonly used indices of RV function do notppear to be specific for RV contractility, but rather reflecthe interaction between RV pump function and pulmonaryascular load [28]. However, the best way to assess contrac-ile performance for the overloaded RV is still a matter ofebate.

Clinical series in PH have highlighted that changes in RVunction — rather than changes in pulmonary pressure orVR — affect long-term survival dramatically [37]. Impair-

ent of RV contractility in both longitudinal and transverselanes is often observed at an advanced stage in the over-oaded RV. This functional assessment is routinely performedt rest, whereas exercise capacity is a strong predictor
Page 5: Advancing knowledge of right ventricular pathophysiology ...However, the two ventricles are anatomi-cally, haemodynamically and functionally distinct. RV metabolic properties may also

ad 523

Table 2 Cellular and molecular changes involved inright ventricular maladaptive remodelling.

Cardiac myocyte ↑ Length and cross-sectional area↑ Apoptosis↑ Necrosis

Extracellularmatrix

↑ Collagen synthesis; ↓ collagendegradation↓ Capillary density↑ Rate of mast cells; ↑ rate ofTNF-�

Contractility ↓ �-MHC; ↑ �-MHC; ↑ �-SMA↓ SERCA-2

Mitochondrialmetabolism

Glycolytic shift

Oxidative stress Failure in antioxidative defenceNeurohormonal

modulation↓ �-adrenergic receptor density↑ AT II receptor type 1 densityImpairment of AT II receptor type 1signalling pathway

AT: angiotensin; MHC: myosin heavy chain; SERCA-2: sarcoplas-mic endoplasmic reticular calcium adenosine triphosphatase 2;

ebt

nasmfiddtttiott[imt

eutiRmsro

Right ventricular pathophysiology in chronic pressure overlo

of long-term outcome in cardiopulmonary diseases. RVcontractile reserve has been poorly reported. Grunig et al.recently suggested that exercise-induced pulmonary arterysystolic pressure increase could estimate RV contractilereserve, and may be a prognostic factor in PH [38]. Pharma-cological stress using dobutamine infusion is better adaptedto heart failure patients than exercise testing, especiallyfor echocardiography Doppler measurements during stress.Sharma et al. recently studied RV reserve using low-dosedobutamine stress echocardiography in PH patients [39].Contractile reserve for the RV was quantified as any signifi-cant changes in TAPSE and tricuspid annular systolic velocity(S′). The protocol was well tolerated in PH patients. Thisstudy showed that RV reserve was impaired in PH patients,even in those who displayed normal RV function at rest.Dobutamine stress may reveal early RV dysfunction in thesetting of chronic pressure overload. These findings arein accordance with our experimental results. We recentlyreported that dobutamine-induced relative changes in RVstroke volume index (SVI) and Ees were lower in pigletsexposed to chronic pressure overload. RV reserve, definedas any change in SVI during pharmacological stress, wasstrongly associated with resting ventricular-arterial couplingin this study [40]. It could be a promising predictive measurefor estimating RV response to therapy, as well as a prognos-tic marker, as long-term survival is strongly associated withthe ability of the RV to durably face the elevated afterload.

Molecular and cellular correlates of RVremodelling in chronic pressure overload

Examination of animal models provides insight into RV pro-gressive remodelling, from compensated hypertrophy tomaladaptive enlargement and failure. Most pathways havebeen observed previously in LV failure. A better understand-ing of RVF pathophysiology, especially during the transitionfrom adaptive hypertrophy to the decompensated state,is needed to find new therapeutic targets. Cellular andmolecular changes suggested to determine RV maladaptiveremodelling are summarized in Table 2.

Local and systemic activities of the renin-angiotensin-aldosterone system (RAAS) are involved in RV structural andfunctional changes. Even if neurohormonal modulation isbeneficial at the early stage, persistent adrenergic over-stimulation and upregulation of the RAAS are associatedwith disease progression and mortality in PH patients [41].Angiotensin II (AT II) significantly impacts on myocardialremodelling. Rouleau et al. observed that RV chronic pres-sure overload resulted in AT II receptors uncoupling in acompensated RV hypertrophy rabbit model. This impairmentof the RAAS signalling pathway led to adverse contractilityperformance, but could be efficiently reversed after admin-istration of the angiotensin-converting enzyme inhibitorramipril [42]. Systemic and myocardial elevated rates ofAT II may induce cardiomyocyte hypertrophy and contrac-tile dysfunction. Fibrosis secondary to inhibition of collagen

degradation and, to a lesser extent, extracellular matrixinflammation have also been related to an increased level ofAT II [43]. These adverse effects are supported by the cardio-protective action of RAAS blockage. Angiotensin-converting

dapi

SMA: smooth muscle actin; TNF: tumour necrosis factor.

nzyme inhibitors and AT II type 1 receptor blockers haveeen demonstrated to decrease RV expression of collagenype I and type III in rodents [44,45].

Transition from RV compensated state to RVF resultsot only from cardiomyocyte dysfunction, but also fromdverse remodelling of the extracellular matrix. Under per-istent overload conditions, extracellular matrix changesay lead to RV dysfunction. Increasing rates of myocardialbrosis have been reported in RVF models, as previouslyemonstrated in PH patients, whereas only a minimalegree of fibrosis is observed in compensated RV hyper-rophy [43,46]. Increased collagen synthesis occurs early inhe setting of RV wall stress enhancement. This upregula-ion at both transcriptional and post-transcriptional levelsncreases dramatically during the first week of pressureverload conditions, and decreases thereafter. However,he collagen production rate still remains higher than inhe normal RV because of a decreased degradation process47]. In monocrotalline rats, Lamberts et al. suggested thatncreased collagen content in the myocardium may be theost important structural change before RV dysfunction in

he setting of chronic pressure overload [48].Recent animal and human studies support the growing

vidence that inflammation plays a key role in heart fail-re pathophysiology. An increased rate of production ofumour necrosis factor alpha can impair systolic functionn left heart failure [49]. If neutrophils are found in theV myocardium early after acute increase in afterload,acrophages may be involved in progressive remodelling

econdary to chronic PH [50]. We recently showed in aat model of RV dysfunction that macrophage infiltrationf the myocardium was strongly associated with systolic

ysfunction characterized by significant decreases in TAPSEnd RVFAC [51]. The precise role of macrophages in theathophysiology of right heart failure needs to be furthernvestigated.
Page 6: Advancing knowledge of right ventricular pathophysiology ...However, the two ventricles are anatomi-cally, haemodynamically and functionally distinct. RV metabolic properties may also

5

temiohf(cnmfccagtefttccewcttdtFibet

isdsseWpfaiwhsg9ddRieldps

Rvibd

R

CImaati[pmciaofeoohPoRtn

aipmctTsVwmobefti

nhhlpT

24

Reversion to foetal phenotype occurs during RV hyper-rophy in response to chronic pressure overload. Thompsont al. found foetal gene re-expression within the RVyocardium in response to pulmonary artery banding

n cats. Upregulation of Nkx2-5 transcription factor wasbserved, which is usually only expressed in the growingeart. However, the precise function of this transcriptionactor is unknown [52]. A decrease in ˛-myosin heavy chainMHC) gene expression and protein synthesis results in aompensatory increase in �-MHC [46]. This adaptive phe-otype provides a lower energetic cost profile to the failingyocardium, as �-MHC is usually involved in stronger and

aster contractions, whereas �-MHC accounts for lowerontractile potential. We reported in our piglet model ofhronic PH that ventricular-arterial uncoupling was stronglyssociated with upregulation of �-MHC expression. We sug-ested that dynamic changes in myosin expression overime may determine RV work efficiency [53]. Foetal �-MHCxpression may be associated with a metabolic shift fromatty acid to glucose oxidation [54]. As described in solidumours, suppression of the mitochondrial oxidative func-ion has been shown in the RV myocardium exposed tohronic pressure overload [55]. If mitochondrial structuralhanges are observed in RV remodelling (defects, differ-nt shape and size), they have not been well correlatedith the severity of RV dysfunction [43]. Hibernation mito-hondrial metabolism seems, however, to be involved in theransition from compensated RV hypertrophy to maladap-ive remodelling. The glycolytic phenotype resulting in RVysfunction is associated with impaired mitochondrial elec-rical potential, as illustrated by an increased uptake of-18 fluorodeoxyglucose on positron emission tomographymaging [56]. This maladaptive metabolism can be reversedy a pyruvate dehydrogenase kinase inhibitor, dichloroac-tate, leading to an increase in pyruvate concentration inhe mitochondria [56].

RV ischaemia is, furthermore, known to play a key rolen the haemodynamic collapse related to acute severe pres-ure overload. Ischaemia has been reported to induce RVysfunction in experimental acute PH because of compres-ion of coronary vessels, resulting from elevated myocardialtretch. A loss of coronary vasodilatation reserve could alsoxacerbate RV contractility decline in these conditions [57].hether ischaemia is involved in the transition from com-

ensated RV hypertrophy to maladaptive remodelling andailure is still a matter of debate in the overloaded RV. Atn early stage, RV pressure overload may result in signif-cant changes in coronary perfusion because of increasedall stress, especially during systole [58,59]. RV ischaemiaas been investigated in PH patients without coronary arterytenosis. Significant perfusion defects were found in a smallroup of primary PH patients using myocardial technetium-9m scintigraphy, but they were not correlated with RVysfunction severity [60]. Many authors think that myocar-ial ischaemia could be involved in the development ofVF. Recent experimental studies demonstrated that RV cap-llary rarefaction, with decreased expression of vascularndothelial growth factor, could be observed after pro-

onged elevated wall stress in different models of RV chronicysfunction [61—63]. The findings of Bogaard et al. also sup-ort the evidence that RV chronic pressure overload is notufficient per se to induce RVF. Oxidative stress within the

t�ph

J. Guihaire et al.

V myocardium may be significantly increased in pulmonaryascular disease. The authors suggested that a failing antiox-dant defence, influenced by unknown mediators releasedy the damaged pulmonary vasculature, could lead to RVysfunction [64].

V response to therapy

urrent drugs and emerging therapies are listed in Table 3.n PH, three main categories of medication are com-only used: prostanoids, endothelin receptor antagonists

nd phosphodiesterase type 5 inhibitors. A recent meta-nalysis based on pharmacological randomized controlledrials suggested an improvement in both exercise capac-ty and survival in PH patients receiving targeted therapies65]. Improvement of RV function is poorly reported as arimary endpoint in these trials, whereas RVF remains theain cause of hospitalization and mortality in patients with

hronic cardiopulmonary disease. Currently, there is no ded-cated medical cure for RVF. The therapies applied to the LVre usually less beneficial for the dysfunctional RV. More-ver, the beneficial effects of approved PH drugs on RVunction are mainly related to decreased afterload. Directffects on the RV are controversial and might be deleteri-us. Nagendran et al. showed that myocardial expressionf the endothelin axis was upregulated in the setting of RVypertrophy, in both humans and rats exposed to chronicH. These results suggested that endothelin receptor antag-nists may worsen RV contractility, leading to irreversibleV dysfunction [66]. Long-term effects of prostacyclins andype 5 phosphodiesterase inhibitors on RV remodelling areot known.

RV contractility usually compensates for the increasedfterload, and RV-PA uncoupling occurs when RV afterloadncreases much more than RV contractility. However, therostanoid epoprostenol has been associated with increasedortality at 6 months, despite early improvement in exer-

ise capacity [67]. The increase in cardiac output after PHherapy is mainly due to the increase in RV contractility.his may actually result in high myocardial oxygen con-umption and may therefore be detrimental [68]. van deeerdonk et al. reported a poor prognosis for PH patientsith impaired RV systolic function despite significant pul-onary vascular response to therapy [69]. The true effects

f current medications on ventricular-arterial coupling haveeen poorly investigated. It would be of great interest ifmerging therapies improved RV-PA coupling rather thanocussing only on RV contractility. The optimal characteris-ics of a targeted therapy for RV dysfunction are suggestedn Table 4.

Adrenergic stimulation is the main endogenous mecha-ism for increasing cardiac work in the setting of chroniceart failure. Beta-receptor sensitivity and density are,owever, reduced in the hypertrophied RV [70]. Ventricu-ar chronic pressure overload is actually known to be therimary determinant of �-adrenoreceptor density [71,72].he recent findings of Bogaard et al. supported experimen-

al benefits for RV remodelling in response to treatment with-adrenergic blocking agents, such as carvedilol and meto-rolol. In two different rat models of chronic pulmonaryypertension, 4 weeks of treatment with beta-receptor
Page 7: Advancing knowledge of right ventricular pathophysiology ...However, the two ventricles are anatomi-cally, haemodynamically and functionally distinct. RV metabolic properties may also

Right ventricular pathophysiology in chronic pressure overload 525

Table 3 Direct and afterload-dependent effects of pharmacological treatments in right ventricular remodelling: insightsfrom preclinical animal models.

Drug Afterload-independent effects Afterload-dependent effects

PH-targeted therapiesPhosphodiesterase-5inhibitors [80—83]

↑ Mitochondrial activity↓ Ventricular dilatation↑ Contractility

↓ Myocardial fibrosis↑ Capillary density↑ Ventricular-arterial coupling

Prostacyclin analogues[84,85]

↑ Capillary density ↓ Myocardial fibrosis↑ Ventricular-arterial coupling

Endothelin-1 receptorantagonists [66,86]

↓ Contractility ↓ Myocardial fibrosis

Drugs used for left ventricular dysfunctionLevosimendan [77] ↑ Contractility ↑ Ventricular-arterial coupling

(marked decreased in pulmonaryarterial elastance)

Beta-blockers [61] ↓ Myocardial fibrosis↑ Capillary density↑ Systolic function↓ Ventricular dilatation

Any effect on pulmonary circulation

Angiotensin-convertingenzyme inhibitors [44]

↓ Myocardial fibrosis ↓ Myocardial hypertrophy

Statins [87] ↓ ROS production↓ Myocardial inflammation↑ Nitric oxide production

Unknown

Blockers of the renin-angiotensin-aldosteroneaxis [41]

↓ Myocardial fibrosis Unknown

Emerging therapiesPyruvate dehydrogenasekinase inhibitor(dichloroacetate) [56]

↑ Mitochondrial oxidative activity ↑ Exercise capacity

Antioxidant (EUK-134) [88] ↓ Oxidative stress↓ Proapoptotic signalling↓ Myocardial fibrosis↑ Systolic function

Any effect on pulmonary circulation

Multikinase inhibitors(sunitinib and sorafenib)[73]

↓ Cellular hypertrophy↓ Myocardial fibrosis↑ Systolic function

↓ Cellular hypertrophy↓ Myocardial fibrosis↑ Systolic function

Rho-kinase inhibitor(fasudil) [89]

Unknown ↓ Ventricular dilatation↑ Systolic function

Inhibitors of histonedeacetylases [90]

↓ Cellular hypertrophy↓ Capillary density

↓ Endothelial and smooth musclecell hyperplasia

Selective pulmonarySERCA-2a gene transfer[74]

Unknown ↓ Cellular hypertrophy↓ Myocardial fibrosis

PH: pulmonary hypertension; ROS: reactive oxygen species; SERCA-2a: sarcoplasmic endoplasmic reticular calcium adenosinetriphosphatase 2a.

Table 4 Characteristics of the optimal drug to treat the failing right ventricle.

Promotion of cardiomyocyte survivalImprovement in myocardial capillary densityRestoration of mitochondrial activityPreservation of ventricular-arterial coupling without increasing myocardial oxygen consumptionOptimization of right ventricular preload and decreasing of right ventricular afterloadPrevention of myocardial fibrosisControl of inflammatory response

Page 8: Advancing knowledge of right ventricular pathophysiology ...However, the two ventricles are anatomi-cally, haemodynamically and functionally distinct. RV metabolic properties may also

5

be

uiitpaoope(tinntctt

C

Goeoatttn

D

Tc

St1IS

R

[

[

[

[

[

[

[

[

[

[

[

[

[

26

lockers was associated with improvement in exercisendurance and RV systolic performance [61].

Among the new therapeutic approaches, metabolic mod-lation might be the most promising for restoration ofmpaired mitochondrial activity in chronic RV dysfunction,ncluding the use of dichloroacetate. This orphan drug, ini-ially employed in cancer therapy, reduces the inhibitoryhosphorylation of the pyruvate dehydrogenase complex,nd thus restores mitochondrial dysfunction [56]. The usef multikinase inhibitors in RV remodelling has been appliednly in experimental models of compensated RV hypertro-hy, and not yet in RVF [73]. Gene transfer of sarcoplasmicndoplasmic reticular calcium adenosine triphosphatase 2aSERCA-2a) has recently been demonstrated to have in vivoherapeutic efficacy in a rodent model of chronic PH, asllustrated by a marked decrease in pulmonary haemody-amics and myocardial hypertrophy and fibrosis [74]. Otheron-pharmacological approaches have been suggested toarget the failing RV, including myocardial bioengineering,ardiac resynchronization and RV mechanical support. Fur-her experimental investigations need to be carried out inhe setting of isolated RV dysfunction.

onclusion

iven the importance of RV function in the prognosisf chronic cardiopulmonary diseases, major experimentalfforts have been made over the last two decades to improveur knowledge of RV pathophysiology. A comprehensivepproach in animal models has led to new pharmacologicalreatments, but basic research has yet to develop a targetedherapy for the failing RV. Moreover, the question of protec-ive phenotypes with long-term compensated hypertrophyeeds to be addressed.

isclosure of interest

he authors declare that they have no conflicts of interestoncerning this article.

ources of funding: J.G. received a travel grant fromhe Fédération Francaise de Cardiologie in 2012 for a-year research fellowship in the Transplant and Stem Cellmmunobiology Laboratory, Hamburg, Germany (Professoronja Schrepfer).

eferences

[1] Klotz S, Naka Y, Oz MC, Burkhoff D. Biventricular assist device-induced right ventricular reverse structural and functionalremodeling. J Heart Lung Transplant 2005;24:1195—201.

[2] Klotz S, Wenzelburger F, Stypmann J, Welp H, Drees G, SchmidC, et al. Reversible pulmonary hypertension in heart transplantcandidates: to transplant or not to transplant. Ann Thorac Surg

2006;82:1770—3.

[3] Humbert M. Pulmonary arterial hypertension and chronicthromboembolic pulmonary hypertension: pathophysiology.Eur Respir Rev 2010;19:59—63.

[

J. Guihaire et al.

[4] Benza RL, Miller DP, Gomberg-Maitland M, Frantz RP, ForemanAJ, Coffey CS, et al. Predicting survival in pulmonary arterialhypertension: insights from the Registry to Evaluate Early andLong-Term Pulmonary Arterial Hypertension Disease Manage-ment (REVEAL). Circulation 2010;122:164—72.

[5] Hoeper MM, Mayer E, Simonneau G, Rubin LJ. Chronicthromboembolic pulmonary hypertension. Circulation2006;113:2011—20.

[6] Humbert M, Sitbon O, Chaouat A, Bertocchi M, HabibG, Gressin V, et al. Survival in patients with idiopathic,familial, and anorexigen-associated pulmonary arterial hyper-tension in the modern management era. Circulation 2010;122:156—63.

[7] Starr IJW, Meade RH. The absence of conspicuous increments ofvenous pressure after severe damage to the RV of the dog, withthe discussion with the relation between clinical congestiveheart failure and heart disease. Am Heart J 1943;26:291—301.

[8] Fontan F, Baudet E. Surgical repair of tricuspid atresia. Thorax1971;26:240—8.

[9] Buckingham M, Meilhac S, Zaffran S. Building the mammalianheart from two sources of myocardial cells. Nat Rev Genet2005;6:826—35.

10] Haddad F, Hunt SA, Rosenthal DN, Murphy DJ. Right ventricularfunction in cardiovascular disease, part I: anatomy, physiol-ogy, aging, and functional assessment of the right ventricle.Circulation 2008;117:1436—48.

11] Simon MA, Deible C, Mathier MA, Lacomis J, Goitein O, ShroffSG, et al. Phenotyping the right ventricle in patients with pul-monary hypertension. Clin Transl Sci 2009;2:294—9.

12] Mehra MR, Park MH, Landzberg MJ, Lala A, Waxman AB. Rightheart failure: toward a common language. J Heart Lung Trans-plant 2014;33:123—6.

13] Guihaire J, Bogaard HJ, Flecher E, Noly PE, Mercier O, HaddadF, et al. Experimental models of right heart failure: a windowfor translational research in pulmonary hypertension. SeminRespir Crit Care Med 2013;34:689—99.

14] Kelly RG. Building the right ventricle. Circ Res 2007;100:943—5.

15] Kelly RG, Brown NA, Buckingham ME. The arterial pole of themouse heart forms from Fgf10-expressing cells in pharyngealmesoderm. Dev Cell 2001;1:435—40.

16] Mjaatvedt CH, Nakaoka T, Moreno-Rodriguez R, Norris RA,Kern MJ, Eisenberg CA, et al. The outflow tract of theheart is recruited from a novel heart-forming field. Dev Biol2001;238:97—109.

17] Viragh S, Challice CE. Origin and differentiation of car-diac muscle cells in the mouse. J Ultrastruct Res 1973;42:1—24.

18] Greyson CR. Pathophysiology of right ventricular failure. CritCare Med 2008;36:S57—65.

19] Bishop VS, Horwitz LD. Quantitative assessment of cardiacpump performance. J Physiol 1977;269:355—70.

20] Dyke CM, Brunsting LA, Salter DR, Murphy CE, Abd-ElfattahA, Wechsler AS. Preload dependence of right ventricularblood flow: I. The normal right ventricle. Ann Thorac Surg1987;43:478—83.

21] Hunter KS, Lee P-F, Lanning CJ, Ivy DD, Kirby KS, Claussen LR,et al. Pulmonary vascular input impedance is a combined mea-sure of pulmonary vascular resistance and stiffness and predictsclinical outcomes better than pulmonary vascular resistancealone in pediatric patients with pulmonary hypertension. AmHeart J 2008;155:166—74.

22] Sunagawa K, Maughan WL, Sagawa K. Optimal arterial resis-tance for the maximal stroke work studied in isolated canineleft ventricle. Circ Res 1985;56:586—95.

23] Ghio S, Gavazzi A, Campana C, Inserra C, Klersy C, Sebas-tiani R, et al. Independent and additive prognostic valueof right ventricular systolic function and pulmonary artery

Page 9: Advancing knowledge of right ventricular pathophysiology ...However, the two ventricles are anatomi-cally, haemodynamically and functionally distinct. RV metabolic properties may also

ad

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

Right ventricular pathophysiology in chronic pressure overlo

pressure in patients with chronic heart failure. J Am Coll Car-diol 2001;37:183—8.

[24] Sagawa K. The end-systolic pressure-volume relation of theventricle: definition, modifications and clinical use. Circulation1981;63:1223—7.

[25] Sunagawa K, Sagawa K. Models of ventricular contrac-tion based on time-varying elastance. Crit Rev Biomed Eng1982;7:193—228.

[26] Wauthy P, Pagnamenta A, Vassalli F, Naeije R, BrimioulleS. Right ventricular adaptation to pulmonary hypertension:an interspecies comparison. Am J Physiol Heart Circ Physiol2004;286:H1441—47.

[27] Aguero J, Ishikawa K, Hadri L, Santos-Gallego C, Fish K, Ham-moudi N, et al. Characterization of right ventricular remodelingand failure in a chronic pulmonary hypertension model. Am JPhysiol Heart Circ Physiol 2014;307:H1204—15.

[28] Guihaire J, Haddad F, Boulate D, Decante B, Denault AY, WuJ, et al. Noninvasive indices of right ventricular function aremarkers of ventricular-arterial coupling rather than ventric-ular contractility: insights from a porcine model of chronicpressure overload. Eur Heart j Cardiovasc Imaging 2013;14:1140—9.

[29] Guyton AC, Lindsey AW, Gilluly JJ. The limits of rightventricular compensation following acute increasein pulmonary circulatory resistance. Circ Res 1954;2:326—32.

[30] Brimioulle S, Wauthy P, Ewalenko P, Rondelet B, Vermeulen F,Kerbaul F, et al. Single-beat estimation of right ventricular end-systolic pressure-volume relationship. Am J Physiol Heart CircPhysiol 2003;284:H1625—30.

[31] Kuehne T, Saeed M, Gleason K, Turner D, Teitel D, HigginsCB, et al. Effects of pulmonary insufficiency on biventricularfunction in the developing heart of growing swine. Circulation2003;108:2007—13.

[32] Trip P, Kind T, van de Veerdonk MC, Marcus JT, de ManFS, Westerhof N, et al. Accurate assessment of load-independent right ventricular systolic function in patients withpulmonary hypertension. J Heart Lung Transplant 2013;32:50—5.

[33] Cheung MM, Smallhorn JF, Redington AN, Vogel M. Theeffects of changes in loading conditions and modulation ofinotropic state on the myocardial performance index: com-parison with conductance catheter measurements. Eur Heart J2004;25:2238—42.

[34] Mauritz GJ, Kind T, Marcus JT, Bogaard HJ, van de Veerdonk M,Postmus PE, et al. Progressive changes in right ventricular geo-metric shortening and long-term survival in pulmonary arterialhypertension. Chest 2012;141:935—43.

[35] Vogel M, Schmidt MR, Kristiansen SB, Cheung M, White PA,Sorensen K, et al. Validation of myocardial acceleration dur-ing isovolumic contraction as a novel noninvasive index ofright ventricular contractility: comparison with ventricularpressure-volume relations in an animal model. Circulation2002;105:1693—9.

[36] Yeo TC, Dujardin KS, Tei C, Mahoney DW, McGoon MD,Seward JB. Value of a Doppler-derived index combiningsystolic and diastolic time intervals in predicting outcomein primary pulmonary hypertension. Am J Cardiol 1998;81:1157—61.

[37] Vonk-Noordegraaf A, Haddad F, Chin KM, Forfia PR, Kawut SM,Lumens J, et al. Right heart adaptation to pulmonary arterialhypertension: physiology and pathobiology. J Am Coll Cardiol2013;62:D22—33.

[38] Grunig E, Tiede H, Enyimayew EO, Ehlken N, Seyfarth

HJ, Bossone E, et al. Assessment and prognostic rele-vance of right ventricular contractile reserve in patientswith severe pulmonary hypertension. Circulation 2013;128:2005—15.

[

527

39] Sharma T, Lau EM, Choudhary P, Torzillo PJ, Munoz PA, SimmonsLR, et al. Dobutamine stress for evaluation of right ventricu-lar reserve in pulmonary arterial hypertension. Eur Respir J2015;45:700—8.

40] Guihaire J, Haddad F, Noly PE, Boulate D, Decante B, DartevelleP, et al. Right ventricular reserve in a piglet model of chronicpulmonary hypertension. Eur Respir J 2015;45:709—17.

41] de Man FS, Tu L, Handoko ML, Rain S, Ruiter G, Francois C,et al. Dysregulated Renin-Angiotensin-aldosterone system con-tributes to pulmonary arterial hypertension. Am J Respir CritCare Med 2012;186:780—9.

42] Rouleau JL, Kapuku G, Pelletier S, Gosselin H, Adam A, GagnonC, et al. Cardioprotective effects of ramipril and losartan inright ventricular pressure overload in the rabbit: importance ofkinins and influence on angiotensin II type 1 receptor signalingpathway. Circulation 2001;104:939—44.

43] Bogaard HJ, Abe K, Vonk-Noordegraaf A, Voelkel NF. Theright ventricle under pressure: cellular and molecular mech-anisms of right heart failure in pulmonary hypertension. Chest2009;135:794—804.

44] Ahn BH, Park HK, Cho HG, Lee HA, Lee YM, Yang EK, et al.Estrogen and enalapril attenuate the development of rightventricular hypertrophy induced by monocrotaline in ovariec-tomized rats. J Korean Med Sci 2003;18:641—8.

45] Yoshiyama M, Takeuchi K, Hanatani A, Shimada T, Takemoto Y,Shimizu N, et al. Effect of cilazapril on ventricular remodelingassessed by Doppler-echocardiographic assessment and cardiacgene expression. Cardiovasc Drugs Ther 1998;12:57—70.

46] Lowes BD, Minobe W, Abraham WT, Rizeq MN, Bohlmeyer TJ,Quaife RA, et al. Changes in gene expression in the intacthuman heart. Downregulation of alpha-myosin heavy chain inhypertrophied, failing ventricular myocardium. J Clin Invest1997;100:2315—24.

47] Bishop JE, Rhodes S, Laurent GJ, Low RB, Stirewalt WS.Increased collagen synthesis and decreased collagen degra-dation in right ventricular hypertrophy induced by pressureoverload. Cardiovasc Res 1994;28:1581—5.

48] Lamberts RR, Caldenhoven E, Lansink M, Witte G, VaessenRJ, St Cyr JA, et al. Preservation of diastolic function inmonocrotaline-induced right ventricular hypertrophy in rats.Am J Physiol Heart Circ Physiol 2007;293:H1869—76.

49] Wrigley BJ, Lip GY, Shantsila E. The role of monocytes andinflammation in the pathophysiology of heart failure. Eur JHeart Fail 2011;13:1161—71.

50] Watts JA, Gellar MA, Obraztsova M, Kline JA, Zagorski J. Roleof inflammation in right ventricular damage and repair follow-ing experimental pulmonary embolism in rats. Int J Exp Pathol2008;89:389—99.

51] Guihaire J, Deuse T, Fadel E, Reichenspurner H, RobbinsRC, Schrepfer S. Macrophages infiltration is associated withmaladaptive remodeling of the right ventricle in an experi-mental model of pulmonary hypertension. J Heart Lung Transpl2014;33:S228.

52] Thompson JT, Rackley MS, O’Brien TX. Upregulation of the car-diac homeobox gene Nkx2-5 (CSX) in feline right ventricularpressure overload. Am J Physiol 1998;274:H1569—73.

53] Guihaire J, Haddad F, Boulate D, Capderou A, Decante B,Flecher E, et al. Right ventricular plasticity in a porcinemodel of chronic pressure overload. J Heart Lung Transplant2014;33:194—202.

54] Watts JA, Marchick MR, Kline JA. Right ventricular heart fail-ure from pulmonary embolism: key distinctions from chronicpulmonary hypertension. J Cardiac Fail 2010;16:250—9.

55] Michelakis ED, Webster L, Mackey JR. Dichloroacetate (DCA) asa potential metabolic-targeting therapy for cancer. Br J Cancer

2008;99:989—94.

56] Piao L, Fang YH, Cadete VJ, Wietholt C, Urboniene D, TothPT, et al. The inhibition of pyruvate dehydrogenase kinase

Page 10: Advancing knowledge of right ventricular pathophysiology ...However, the two ventricles are anatomi-cally, haemodynamically and functionally distinct. RV metabolic properties may also

5

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

28

improves impaired cardiac function and electrical remodelingin two models of right ventricular hypertrophy: resusci-tating the hibernating right ventricle. J Mol Med 2010;88:47—60.

57] Vlahakes GJ, Turley K, Hoffman JI. The pathophysiologyof failure in acute right ventricular hypertension: hemo-dynamic and biochemical correlations. Circulation 1981;63:87—95.

58] Morimont P, Lambermont B, Ghuysen A, Gerard P, Kolh P,Lancellotti P, et al. Effective arterial elastance as an indexof pulmonary vascular load. Am J Physiol Heart Circ Physiol2008;294:H2736—42.

59] Saouti N, Westerhof N, Postmus PE, Vonk-Noordegraaf A.The arterial load in pulmonary hypertension. Eur Respir Rev2010;19:197—203.

60] Gomez A, Bialostozky D, Zajarias A, Santos E, Palomar A,Martinez ML, et al. Right ventricular ischemia in patientswith primary pulmonary hypertension. J Am Coll Cardiol2001;38:1137—42.

61] Bogaard HJ, Natarajan R, Mizuno S, Abbate A, Chang PJ, ChauVQ, et al. Adrenergic receptor blockade reverses right heartremodeling and dysfunction in pulmonary hypertensive rats.Am J Respir Crit Care Med 2010;182:652—60.

62] Noly PE, Haddad F, Mercier O, Dorfmuller P, Decante B, Dartev-elle P, et al. Role of angiogenesis in the right ventricleremodeling in response to pulmonary hypertension. J HeartLung Transpl 2013;32:S78.

63] Rondelet B, Dewachter C, Kerbaul F, Kang X, Fesler P, BrimioulleS, et al. Prolonged overcirculation-induced pulmonary arterialhypertension as a cause of right ventricular failure. Eur HeartJ 2012;33:1017—26.

64] Bogaard HJ, Natarajan R, Henderson SC, Long CS, KraskauskasD, Smithson L, et al. Chronic pulmonary artery pressure ele-vation is insufficient to explain right heart failure. Circulation2009;120:1951—60.

65] Galie N, Rubin L, Simonneau G. Developing a heart score: nextsteps. Am J Cardiol 2012;110:49S—51S.

66] Nagendran J, Sutendra G, Paterson I, Champion HC, WebsterL, Chiu B, et al. Endothelin axis is upregulated in humanand rat right ventricular hypertrophy. Circ Res 2013;112:347—54.

67] Califf RM, Adams KF, McKenna WJ, Gheorghiade M, Uret-sky BF, McNulty SE, et al. A randomized controlled trial ofepoprostenol therapy for severe congestive heart failure: theFlolan International Randomized Survival Trial (FIRST). AmHeart J 1997;134:44—54.

68] Gomez-Arroyo J, Sandoval J, Simon MA, Dominguez-Cano E,Voelkel NF, Bogaard HJ. Treatment for pulmonary arterialhypertension-associated right ventricular dysfunction. Ann AmThorac Soc 2014;11:1101—15.

69] van de Veerdonk MC, Kind T, Marcus JT, Mauritz GJ, HeymansMW, Bogaard HJ, et al. Progressive right ventricular dysfunctionin patients with pulmonary arterial hypertension responding totherapy. J Am Coll Cardiol 2011;58:2511—9.

70] Pela G, Missale C, Raddino R, Condorelli E, Spano PF, Visioli O.Beta 1- and beta 2-receptors are differentially desensitized inan experimental model of heart failure. J Cardiovasc Pharma-col 1990;16:839—46.

71] Bristow MR, Minobe W, Rasmussen R, Larrabee P, Skerl L, KleinJW, et al. Beta-adrenergic neuroeffector abnormalities in thefailing human heart are produced by local rather than systemicmechanisms. J Clin Invest 1992;89:803—15.

72] Bristow MR, Minobe WA, Raynolds MV, Port JD, RasmussenR, Ray PE, et al. Reduced beta 1 receptor messenger

RNA abundance in the failing human heart. J Clin Invest1993;92:2737—45.

73] Kojonazarov B, Sydykov A, Pullamsetti SS, Luitel H, Dahal BK,Kosanovic D, et al. Effects of multikinase inhibitors on pressure

[

J. Guihaire et al.

overload-induced right ventricular remodeling. Int J Cardiol2013;167:2630—7.

74] Hadri L, Kratlian RG, Benard L, Maron BA, Dorfmuller P,Ladage D, et al. Therapeutic efficacy of AAV1.SERCA2a inmonocrotaline-induced pulmonary arterial hypertension. Cir-culation 2013;128:512—23.

75] Hessel MH, Steendijk P, den Adel B, Schutte CI, vander Laarse A. Characterization of right ventricular func-tion after monocrotaline-induced pulmonary hypertension inthe intact rat. Am J Physiol Heart Circ Physiol 2006;291:H2424—30.

76] Tabima DM, Hacker TA, Chesler NC. Measuring right ventricularfunction in the normal and hypertensive mouse hearts usingadmittance-derived pressure-volume loops. Am J Physiol HeartCirc Physiol 2010;299:H2069—75.

77] Kerbaul F, Gariboldi V, Giorgi R, Mekkaoui C, Guieu R, Fes-ler P, et al. Effects of levosimendan on acute pulmonaryembolism-induced right ventricular failure. Crit Care Med2007;35:1948—54.

78] Rondelet B, Kerbaul F, Motte S, van Beneden R, Rem-melink M, Brimioulle S, et al. Bosentan for the preventionof overcirculation-induced experimental pulmonary arterialhypertension. Circulation 2003;107:1329—35.

79] Lambermont B, Segers P, Ghuysen A, Tchana-Sato V, Morimont P,Dogne JM, et al. Comparison between single-beat and multiple-beat methods for estimation of right ventricular contractility.Crit Care Med 2004;32:1886—90.

80] Borgdorff MA, Bartelds B, Dickinson MG, Boersma B, Weij M,Zandvoort A, et al. Sildenafil enhances systolic adaptation, butdoes not prevent diastolic dysfunction, in the pressure-loadedright ventricle. Eur J Heart Fail 2012;14:1067—74.

81] Mouchaers KT, Schalij I, Versteilen AM, Hadi AM, van NieuwAmerongen GP, van Hinsbergh VW, et al. Endothelin recep-tor blockade combined with phosphodiesterase-5 inhibitionincreases right ventricular mitochondrial capacity in pul-monary arterial hypertension. Am J Physiol Heart Circ Physiol2009;297:H200—7.

82] Nagendran J, Archer SL, Soliman D, Gurtu V, Moudgil R, HaromyA, et al. Phosphodiesterase type 5 is highly expressed in thehypertrophied human right ventricle, and acute inhibition ofphosphodiesterase type 5 improves contractility. Circulation2007;116:238—48.

83] Schafer S, Ellinghaus P, Janssen W, Kramer F, Lustig K, Milting H,et al. Chronic inhibition of phosphodiesterase 5 does not pre-vent pressure overload-induced right ventricular remodelling.Cardiovasc Res 2009;82:30—9.

84] Schermuly RT, Kreisselmeier KP, Ghofrani HA, Samidurai A,Pullamsetti S, Weissmann N, et al. Antiremodeling effects ofiloprost and the dual-selective phosphodiesterase 3/4 inhibitortolafentrine in chronic experimental pulmonary hypertension.Circ Res 2004;94:1101—8.

85] van Albada ME, Berger RM, Niggebrugge M, van Veghel R,Cromme-Dijkhuis AH, Schoemaker RG. Prostacyclin therapyincreases right ventricular capillarisation in a model forflow-associated pulmonary hypertension. Eur J Pharmacol2006;549:107—16.

86] Yuyama H, Koakutsu A, Fujiyasu N, Tanahashi M, Fujimori A,Sato S, et al. Effects of selective endothelin ET(A) recep-tor antagonists on endothelin-1-induced potentiation of cancerpain. Eur J Pharmacol 2004;492:177—82.

87] Jasinska-Stroschein M, Owczarek J, Wesolowska A, Orszulak-Michalak D. Rosuvastatin, sildenafil and their combination inmonocrotaline-induced pulmonary hypertension in rat. ActaPharm 2014;64:345—53.

88] Redout EM, van der Toorn A, Zuidwijk MJ, van de Kolk CW, van

Echteld CJ, Musters RJ, et al. Antioxidant treatment attenuatespulmonary arterial hypertension-induced heart failure. Am JPhysiol Heart Circ Physiol 2010;298:H1038—47.
Page 11: Advancing knowledge of right ventricular pathophysiology ...However, the two ventricles are anatomi-cally, haemodynamically and functionally distinct. RV metabolic properties may also

ad

[

Right ventricular pathophysiology in chronic pressure overlo

[89] Mouchaers KT, Schalij I, de Boer MA, Postmus PE, van

Hinsbergh VW, van Nieuw Amerongen GP, et al. Fasudilreduces monocrotaline-induced pulmonary arterial hyperten-sion: comparison with bosentan and sildenafil. Eur Respir J2010;36:800—7.

529

90] Bogaard HJ, Mizuno S, Hussaini AA, Toldo S, Abbate A,

Kraskauskas D, et al. Suppression of histone deacetylasesworsens right ventricular dysfunction after pulmonary arterybanding in rats. Am J Respir Crit Care Med 2011;183:1402—10.