REVIEW

Cardiovascular Research (2016) 111, 154–162 doi:10.1093/cvr/cvw107

Old dog, new tricks: novel cardiac targets and stress regulation by protein kinase G Peter P. Rainer1* and David A. Kass 2 1

Division of Cardiology, Medical University of Graz, Auenbruggerplatz 15, A-8036 Graz, Austria; and 2Division of Cardiology, Johns Hopkins Medical Institutions, Baltimore, MD, USA

Received 12 April 2016; revised 16 May 2016; accepted 18 May 2016; online publish-ahead-of-print 13 June 2016

Abstract

The second messenger cyclic guanosine 3′ 5′ monophosphate (cGMP) and its downstream effector protein kinase G (PKG) have been discovered more than 40 years ago. In vessels, PKG1 induces smooth muscle relaxation in response to nitric oxide signalling and thus lowers systemic and pulmonary blood pressure. In platelets, PKG1 stimulation by cGMP inhibits activation and aggregation, and in experimental models of heart failure (HF), PKG1 activation by inhibiting cGMP degradation is protective. The net effect of the above-mentioned signalling is cardiovascular protection. Yet, while modulation of cGMP-PKG has entered clinical practice for treating pulmonary hypertension or erectile dysfunction, translation of promising studies in experimental HF to clinical success has failed thus far. With the advent of new technologies, novel mechanisms of PKG regulation, including mechanosensing, redox regulation, protein quality control, and cGMP degradation, have been discovered. These novel, non-canonical roles of PKG1 may help understand why clinical translation has disappointed thus far. Addressing them appears to be a requisite for future, successful translation of experimental studies to the clinical arena.

----------------------------------------------------------------------------------------------------------------------------------------------------------Keywords

Protein kinase G † cGMP-dependent protein kinase type 1 † Cardiac mechanosensing † Proteasome † Redox regulation † Phosphodiesterase

1. Introduction Protein kinase G (PKG) is a major downstream effector of the nitric oxide (NO) and natriuretic peptide (NP) signalling pathways, each being linked to a respective guanylate cyclase (soluble—sGC, particulate—pGC) to generate cyclic guanosine 3′ 5′ monophosphate (cGMP), thereby activating the kinase (Figure 1). Levels of cGMP and thus PKG activation are also regulated by a select group of phosphodiesterases (PDEs) that control cGMP hydrolysis. This signalling system is highly compartmentalized, generating local pools of cGMP and indeed differential PKG activation which both shares and displays distinct signalling effects depending on how it was generated. In arterial smooth muscle, PKG1 activation induces relaxation and plays an important role in blood pressure regulation. In the heart, many studies have established its action as a brake on stress-response signalling. Much of this action relates to the capacity of PKG to suppress selective G-protein-coupled receptor agonism,1,2 phosphorylate proteins in the sarcomere,7,8,15 suppress transforming growth factor beta (TGFb) cascades,13 and antagonize members of transient receptor potential cation channels.10,16,17 Recent work has expanded this list to include mechanosensing11 and protein quality control,3 and has revealed novel PDE regulators that illuminate the compartmental nature of this signalling.6 In addition, post-translational oxidation of the PKG1a isoform impairs

its brake-like function, revealing a new mechanism whereby oxidative stress adversely impacts stress-response signalling.12 For this review, we have focused particularly on these recent findings. In addition, we discuss some of the continuing controversies that offer alternative perspectives on the role of cardiac PKG1a and its regulation by PDE5.

2. PKG regulation of transient receptor potential canonical channel 6: a new role in mechanosensing In 2006, several studies established that members of the transient receptor potential canonical channel (TRPC) family of non-selective cation channels are involved in myocardial hypertrophy.18 – 21 TRPCs consist of six transmembrane domains with intracytoplasmic N- and C-termini, forming homo- or heterotetramers.22 TRPC subtypes can respond to intracellular calcium depletion (termed store-operated) and/or be coupled to G-protein-coupled receptor agonism through stimulation by diacylglycerol (DAG). Two family members in particular, TRPC3 and TRPC6, appear involved. Both are up-regulated in cardiac disease such as hypertrophy from pressure overload,10,23 and calcium entering through either channel has been linked to the activation of calcineurin-nuclear factor of activated T cells (NFAT) signalling,

* Corresponding author. Tel: +43 316 385 12544; fax: +43 316 385 13733, Email: [email protected] Published on behalf of the European Society of Cardiology. All rights reserved. & The Author 2016. For permissions please email: [email protected].

Novel stress regulation by protein kinase G

155

Figure 1 cGMP-PKG signalling in the cardiomyocyte. cGMP is generated by NO stimulation of soluble guanylate cyclase (sGC) in the cytosol (left) or natriuretic peptide receptor (NPR) coupled particulate guanylate cyclase (pGC) activation at the cell membrane (right). PDE5, which is located at the z-discs, appears to mainly target sGC-synthetized cGMP. PKG attenuates G-protein-coupled signalling (GPCR) via phosphorylation of regulator of G-protein signalling (RGS). Studies suggest that this happens through both sGC- and pGC-activated PKG pools.1,2 NO-derived cGMP/PKG improves proteasomal function,3 dampens ischaemia- and doxorubicin-related damaging mitochondrial signalling,4,5 and reduces activation of the maladaptive transcription factors NFAT and myocyte enhancer factor-2 (MEF2).6 NO/sGC/cGMP-activated PKG also phosphorylates several myofilament proteins effecting enhanced lusitropy and attenuated beta-adrenergic inotropy.7 – 9 PKG stimulated by both cGMP generated via the pGC pathway at the cell membrane or PDE5 inhibition with sildenafil inhibits TRPC6.6 Calcium that enters the cell through TRPC6 partakes in myofilament activation and force generation (slow force response, SFR), and calcineurin (CaN)-NFAT signalling,10,11 and possibly mitochondrial calcium and permeability transition. In addition, pGC-cGMP-stimulated PKG selectively enhances transcription factors that have been linked to cell survival and enhanced adaption, GATAbinding protein 4 (GATA4), and cAMP response element-binding protein (CREB).6 NP-stimulated cGMP is preferentially targeted by PDE9, which localizes close to t-tubules. Oxidation (ROS) attenuates PKG activation and protective signalling in the cardiomyocyte.12 Whether oxidative activation differs according to the source of cGMP/PKG remains to be determined. PKG prevents activation and nuclear transport of mothers against decapentaplegic homologue 3 (SMAD3) and hence TGFb-related signalling in myofibroblasts.13,14 Whether this happens in cardiomyocytes and whether this is differentially impacted according to PKG generation are unclear. Please note that research into the cardiac targets of the PDE9-related cGMP-PKG signal is as yet limited, and many of the studies to date did not strictly differentiate between sGC- and pGC-regulated cGMP signalling. Future studies will reveal the amount of crosstalk between these functional compartments and the respective importance in varied pathophysiologies, and thus paint a more complete or modified picture of compartmentalized PKG-associated signalling.

contributing to fibrosis, hypertrophy, and cardiac dysfunction.18 – 21 Expression of dominant negative TRPC forms (3, 6, or 4) was protective against pathological cardiac stress.17 In 2012, Davis et al. further revealed that TRPC6 is pivotal to myofibroblast activation and thus scar formation in the heart and elsewhere.24 Expressing dominant negative channels may differ, however, from embryonic gene deletion, and indeed, slightly different results were reported in mice in which TRPC3 or TRPC6 was knocked out. In our work, we found that deletion of either channel alone did not suppress maladaptive hypertrophy from chronic pressure overload, whereas combined deletion was effective.23 Domes et al. found that deletion of TRPC3 but not TRPC6 blocked hypertrophy and fibrosis stimulated by angiotensin II (AII), though this effect was lost in TRPC3/6 doubleknockout (KO) animals.25 The cause for the differences between

studies, or why co-deleting TRPC6 eliminated the protection observed by knocking out TRPC3 in the latter study, remains unclear. Both channels may form heterotetramers with each other and other TRP channels, so interaction may be one factor. The reason we focus on these channels here is that both are phosphorylated by PKG in their N-terminus,26,27 which reduces channel conductance, and in the case of TRPC6, associated hypertrophy.10,16,28 Both NO- and NP-derived PKG stimulation has this effect (Figure 1), and it has been reported in cardiomyocytes,10,16 vascular smooth muscle,26 and the kidney.29 Studies have revealed that TRPC6 threonine 69 (70 in human) is one major target,10,16,28 while others have also found serine 321 (322 in human)10 highly relevant. Site mutagenesis of either was sufficient to reduce channel current, and block NFAT activation stimulated by Gaq-coupled agonists.10,16,27 Gaq-agonist-stimulated

156 myocyte hypertrophy was greater if TRPC6 was mutated so that it could not be phosphorylated at these sites, whereas phosphomimetic forms were protected.10 More recently, this same signalling interaction between PKG and TRPC6 was discovered to play an important role in myocyte mechanosensing. Upon an acute rise in systolic myocardial load (for example increasing muscle or cell length, while allowing for systolic shortening against a fixed afterload—auxotonic contraction), cardiac muscle augments its developed force. The initial response, known as the Frank – Starling mechanism, is calcium independent and related to myofilament calcium sensitivity.30 PKG impacts this dependence by phosphorylation of troponin I (TnI) that desensitizes myofilaments to calcium,7,31,32 an effect that is most notable when beta-adrenergic co-stimulation is present. This is achieved primarily via the NO-sGC pathway, and by inhibiting PDE5A which targets cGMP generated by sGC.8 Models in which hearts express the skeletal isoform of TnI that lacks the S23, S24 PKG phosphorylation sites have confirmed their importance.7,8,33 PKG1a also modifies myocardial passive stiffness by phosphorylating titin in its spring region (N2B, N2BA).15 This may also impact contractile force. Lastly, recent evidence supports PKG modulation of myosin-binding protein C, and this likely influences the kinetics of myocardial contraction.34 PKG also impacts a calcium-dependent contractility response that occurs in myocardium subjected to a sustained afterload increase. This is historically known as the Anrep effect35 and has been linked to stretch-activated G-protein-coupled receptors such as the AII type 1 receptor36 and subsequent mitogen-activated protein (MAP) kinase stimulation of the sodium – hydrogen exchanger (NHE-1).37 The latter then triggers reverse-mode sodium – calcium exchange (NCX)38 to increase cytosolic calcium.39 – 41 Other studies found that stretch-activated cation channels (SACs) are crucial and that this response is inhibited by blocking SACs using compounds such as gadolinium or the spider toxin GsMTx.42 – 44 In support of this, we found that PKG stimulation virtually eliminated loading stress-induced contractile responses, an effect that required expression of TRPC6.11 Myocytes and cardiac muscle lacking TRPC6 display a 30% decline in stress-stimulated contractility, whereas PKG stimulation alone reduced stress-stimulated contractility by 90%. Intriguingly, genetic deletion of TRPC6 eliminated PKG modulation.11 In a mouse model of Duchenne muscular dystrophy, this mechano-stimulated response including force, calcium, and arrhythmia was largely increased. Selective TRPC3/6 antagonism or TRPC6 gene deletion restored this mechano-response to normal levels, and PKG activation fully blocked it.11 Few studies have yet examined the interplay between PKG modulation and TRPC3 or TRPC6 in vivo. Domes et al. used a rescue model, where PKG1 is deleted from all tissues except from smooth muscle, and found that this greatly amplified hypertrophy and fibrosis induced by AII in mice that also lacked TRPC3/6. This was worse than with PKG1 deletion alone, so some interaction seems present.25,45 In contrast, Klaiber et al. found that mice lacking the NP-coupled guanylate cyclase (GC-A) in myocytes exhibited worse hypertrophy from AII infusion, and this was ameliorated if TRPC3/6 genes were also deleted.46 From our work,11 the interaction of PKG on TRPC6 showed more potent mechano-modulatory effects than gene deletion of TRPC6 alone, so coupling to other channel subtypes with phosphorylated TRPC6 acting like a dominant negative seems likely. TRPC1, another mechanosensitive channel, is also modified by PKG phosphorylation,47 resulting in inhibition of smooth muscle hyperpolarization. Whether this impacts cardiac mechanosensing remains to be explored.

P.P. Rainer and D.A. Kass

3. PKG and proteasomal function Protein synthesis and degradation represent a finely tuned equilibrium. The removal of dysfunctional proteins prevents their accumulation and aggregation and concurrent cell damage.48 There are two main pathways for clearance of misfolded or aggregated proteins, the ubiquitin proteasome system (UPS) and autophagy. Ranek et al. found that overexpressing constitutively active PKG1a or activating PKG by PDE5 inhibition stimulates proteasome peptidase activities and reduces misfolded and ubiquitinated protein accumulation. This was demonstrated in cultured neonatal rat ventricular myocytes (NRVMs) and a transgenic mouse model of desmin-related cardiomyopathy that exhibits intracellular protein aggregates (aBcrystallineR120G overexpressors). Interestingly, the increase in proteolytic activity did not affect all known targets of the UPS but demonstrated substrate specificity. Evidence for PKG phosphorylation of proteasome subunits remains indirect, with data showing an acidic shift of the isoelectric points of 20S (b5) and 19S (Rpt6) proteasome subunits.3 In smooth muscle, stimulation with 8-bromo-cGMP decreases PKG1a (but not PKG1b) expression by inducing ubiquitination and degradation. PKG1 inhibition with the specific inhibitor DT-2 or mutation of the PKG1a autophosphorylation site serine 64 abolished this response, suggesting that PKG1 catalytic activity is required for this feedback loop.49 PKG also partakes in the endoplasmic reticulum stress response (ERSR). PDE5 inhibition with sildenafil reduced ER stress in isoproterenol-treated rats and pressure-overloaded (transverse aortic constriction—TAC) mice in a PKG-dependent manner.50 The mechanism remains uncertain. A similar effect was observed with vasonatrin, a synthetic chimaera of atrial (ANP) and C-type natriuretic peptides (CNPs) in diabetic rats exposed to ischaemia/reperfusion. This was linked to a fall in the expression of the chaperone 78 kDa glucoseregulated protein (GRP78) and the transcription factor CCAAT/ enhancer binding protein (CHOP). The protection was mimicked by cGMP stimulation and prevented by PKG inhibition. Silencing PKG1a in cardiomyoblast H9c2 cells exposed to hypoxia blunted protection from vasonatrin on ER stress, whereas overexpressing PKG1a did the opposite.51

4. Redox regulation of PKG localization and activity Oxidant stress modifies many upstream components of the cGMP-PKG signalling pathway. NO synthase (NOS) oxidation induces enzyme uncoupling where less NO and more superoxide are generated,52 and oxidation of sGC reduces NO responsiveness.53 First described in vascular tissue, these oxidant targets play a role in myocyte pathophysiology as well.54,55 In 2007, Eaton et al. revealed that PKG1a itself is targeted by oxidation,56 proposing this as a cGMP-independent activation mechanism relevant to vascular smooth muscle hyperpolarization and vasorelaxation.57 Oxidation was shown to occur at cysteine 42 residues between the two homodimers in the N-terminus, just above the leucine zipper motif that mediates PKG1a dimerization, localization, and interaction with substrates.56,58 – 61 PKG1a responsiveness to H2O2, the direct transnitrosylating NO donor nitrocysteine,62 or H2S63 renders it a redox sensor. Oxidative activation through disulfide bond formation may

Novel stress regulation by protein kinase G

occur in the absence of cGMP;56 indeed, low levels of cGMP favour this modification.64 Increasing cGMP levels reduces H2O2-induced activation likely related to its impact on PKG N-terminus configuration that separates the C42 residues.56,64 As mentioned, PKG1a oxidation facilitates microvascular dilation, regulating blood pressure in intact mice as revealed in animals harbouring a C42 serine substitution (redox-dead PKG1aC42S).57 These animals display blunted blood pressure reduction to nitroglycerine administration65 and are resistant to hypotension induced by septic shock. 66 H2O2 promotes the translocation of PKG1a from the cytoplasm to the cell membrane where it phosphorylates membrane proteins.12,67 In smooth muscle (Figure 2A), this impacts largeconductance Ca2+-activated potassium (BKCa) channels, resulting in PKG-dependent activation of hyperpolarizing K+ currents and coronary vasodilation by H2O2.68 In the normal heart, PKG1a oxidation is less present, but it increases in ischaemic heart disease in humans, and in canine and mouse models of heart failure (HF).12 However, whereas PKG1a oxidation is a gain of function in vessels, in the heart, it is a loss of function with

157 respect to its capacity to counter pathological stress remodelling. C42S PKG1a knockin mice exposed to sustained pressure overload developed less hypertrophy and fibrosis. Membrane translocation of PKG1a upon Gaq-protein or pressure-load stimulation is also observed in cardiomyocytes and has been proposed to enhance suppression of Gaq-coupled signalling.2 When oxidized, however, PKG1a returns to the cytosol, reducing its effective targeting of membranelocalized contributors to the hypertrophic response. One example was its suppression of TRPC6-related signalling, which is reduced when PKGa oxidation is present (Figure 2B). In contrast, the permanently reduced form remained at the membrane where it displayed TRPC6 antagonism.12 Another redox-dependent effect on PKG1a is S-guanylation (covalent adduction of cGMP) at cysteine 195.69 This occurs in the presence of 8-nitro-cGMP, a nitrated derivative of cGMP that is endogenously generated under conditions associated with excess reactive oxygen species (ROS) and NO production. The result is also activation of the kinase. Of note, ROS may also directly impact PKG effector molecules, such as TRPC6.70

Figure 2 Oxidative regulation of PKG in smooth and cardiac muscle cells. In smooth muscle cells, oxidant activation of PKG (cysteine 42 disulfide formation) induces PKG translocation to the cell membrane where it phosphorylates Ca2+-activated potassium channels (BKCa) and induces hyperpolarization and vasorelaxation. Preventing oxidant PKG activation by substituting cysteine 42 with serine (C42S, redox-dead PKG) impairs this process. cGMP activation of PKG is still present; however, it appears that oxidant and cGMP activation impede each other (NTG: nitroglycerine). In contrast, in cardiomyocytes, oxidant activation of PKG, such as happens with neurohumoral activation (Gaq) or pressure overload (TAC) retains oxidized PKG (PKGox) in the cytosol. Redox-dead PKGC42S, which may still be activated by cGMP, localizes to the cell membrane where it attenuates TRPC6-related calcium entry and associated maladaptive signalling.

158

5. Enhancing the myocardial stress response by inhibiting PDE9 vs. PDE5: new approaches for treating heart disease Cyclic AMP- and GMP-degrading PDEs consist of 21 genes and are grouped into 11 gene families according to their substrate affinity, selectivity, and regulation. They degrade cyclic nucleotides by hydrolysing them to their 5′ -monophosphate form. PDEs either hydrolyse cAMP (PDEs 4, 7, 8), cGMP (PDEs 5, 6, 9), cAMP cGMP-sensitively (PDEs 3, 10), or both cAMP and cGMP (PDEs 1, 2, 11).71,72 These major species are further modified by alternative splicing to yield over 100 different isoforms and reside in specific microdomains to confer fine-tuned cyclic nucleotide signalling and target specificity.72 Of the three cGMP-selective PDEs, PDE5, PDE6, and PDE9, only the first and last are thought to be expressed in the heart. PDE5 was the first cGMP-selective enzyme discovered, and its inhibition has provided successful clinical therapy for erectile dysfunction and pulmonary hypertension.73,74 Although many investigations find PDE5 expressed in the myocardium with amplified expression in forms of cardiac disease such as hypertrophy and dilated HF,75,76 not all studies have observed this. Indeed, controversies with respect to the amount and functional impact of PDE5 in the myocardium endure.45,77 – 79 The growing body of data in adult myocytes and hearts, using genetic PDE5 deletion as well as small molecule inhibitors, however, clearly supports its role. In the effort to explore cardiac PDE5 regulation, some studies have questioned the role of PKG1a, too.45,80 These reports used a rescue mouse model, in which PKG1a is re-expressed in vascular smooth muscle, while otherwise globally deleted. The model improves the severe early mortality of the global KO81 but still has 50% mortality at 1 year.82 This may be important as two alternative models that do not display such a severe underlying phenotype have confirmed PKG1a’s role in the heart. In one study, PDE5A was overexpressed solely in myocytes to block PKG activation, revealing exacerbated disease after pressure overload or infarction.83 In another study, PDE5A gene expression was reversible (tetracycline-sensitive promoter), and this led to enhanced PKG activity and improvement of pre-established disease.84 Frantz et al. used another approach with sGC1 (PKG1a and 1b) selectively knocked out in cardiac myocytes only.85 This resulted in worsened maladaptive responses to AII and pressure-overload stimulation. Animal models of heart disease ranging from muscular dystrophies,11,86 – 88 pressure-overload hypertrophy,84,89 ischaemia/infarction,90 – 92 and doxorubicin cardiotoxicity4,93 have reported amelioration of myocardial disease from PDE5 inhibition and cardioprotection by PKG94,95 (for review96). Efforts at human translation continue, and single-centre trials support chronic treatment that improves clinical status, cardiac morphology and function, and exercise capacity in HF patients with depressed systolic function and concomitant pulmonary hypertension.97 – 99 PDE5 inhibition was not useful, however, in the only reported multicentre trial to test PDE5 inhibition in human HF. This trial focused on HF with preserved ejection fraction (HFpEF), the form of disease where the ejection fraction (EF) is in a normal range.100 In HFpEF, both myocardial cGMP and corresponding PKG activity appear to be very low.101 So, unless PDE5 up-regulation was responsible for low cGMP levels, an inhibitor is unlikely to help. To date, there are no data showing that PDE5 is up-regulated in patients

P.P. Rainer and D.A. Kass

studied in this trial, many of which had normal blood pressure, no ventricular hypertrophy, and even mild diastolic disease. In a follow-up subgroup analysis, Borlaug et al. reported that PDE5 inhibition might have depressed left ventricular function and limited exercise reserve.102 This study, however, was too small to make definitive conclusions, and there was no significant difference in exercise capacity between groups. It is possible that anti-adrenergic effects of sildenafil as observed in normal healthy subjects103 or prior animal models104,105 play a role. Two additional under-powered single-centre studies in Duchenne and Becker muscular dystrophies did not show benefits from PDE5 inhibition despite prior encouraging animal data.106,107 Overly advanced cardiomyopathy in the former and absence of heart disease in the latter may explain these findings. While there are many potential reasons for the difficulty in translating PDE5 inhibition benefit, one that has become a prominent focus is the requirement for NO-sGC-derived cGMP for PDE5 regulation to be effective.108,109 The same diseases studied have abnormal NO signalling, and this could impact the efficacy of this approach. Earlier studies established the requirement for NOS3 and consequent cGMP synthesis for PDE5 inhibition to blunt beta-adrenergic inotropy.103 – 105 NOS3 knockout mice or mice with NOS inhibited (L-NAME) do not display anti-adrenergic effects with sildenafil.105 In myocytes, in which cGMP signals at the membrane or cytosol were detected, PDE5 inhibition did not augment cGMP content generated by NP stimulation at the plasma membrane but did increase cGMP stimulated by an NO donor.109 Another example where NO-dependent signalling is compromised and in turn limits the efficacy of PDE5 inhibition as a counter-stress brake is females that lack sex hormones. Female mice with either a Gaq-overexpression genotype or subjected to sustained pressure overload developed cardiac dysfunction and hypertrophy. This was ameliorated by co-administration of a PDE5 inhibitor. However, if they underwent ovariectomy, the benefit from PDE5 inhibition was completely lost and recovered by treatment with exogenous oestrogen. This signalling couples oestrogen to NO-cGMP stimulation and again reveals the tight link between NO-cGMP and PDE5 modulation.110 All these data led to an important question: Is there a way to modulate cGMP signalling by a PDE that does not depend on its synthesis by the NO-sGC pathway? The alternative synthetic path is coupled to NP receptors. Prior work had highlighted PDE2 as a potential NP-targeting PDE.109,111 However, in vivo, the data on PDE2 have shown that it primarily modulates cAMP levels, being stimulated by cGMP to do so.112 – 114 Another possibility is PDE1, a Ca2+-calmodulin (CaM)activated PDE that hydrolyses both cAMP and cGMP.115,116 Data for its role in the heart remain limited; however,117 and a selective impact on cGMP is uncertain. There is another PDE that was known to be highly selective for cGMP—namely PDE9A. PDE9 was discovered in the late 1990s, and its expression is found not only in the brain, gut, and kidneys but also in the heart.118 It has the highest selectivity for cGMP over cAMP.118,119 PDE9A protein expression is found in both human and mouse myocardium with extremely low levels of expression in the normal state, but with elevated expression in disease conditions. Notably, in human HFpEF and HF with reduced ejection fraction (HFrEF), expression is increased.6 Both genetic and pharmacologic inhibition of PDE9A suppressed experimental hypertrophic heart disease, fibrosis, and dysfunction induced by sustained pressure overload. It also blunted hypertrophic signalling cascades in isolated myocytes subjected to endothelin or phenylephrine stimulation.

Novel stress regulation by protein kinase G

The particularly intriguing aspect of PDE9A is that it targets cGMP generated by the NP pathway but is largely insensitive to cGMP produced by sGC. This meant that intact mice with NOS inhibited by L-NAME provided in the drinking water were still susceptible to improvement of pressure-overload stress by PDE9A inhibition, whereas PDE5A inhibition did not provide benefit under these conditions. In addition, phospho-kinome analysis by mass spectrometry and transcription factor analysis revealed that suppression of PDE9A or PDE5A yields distinctive molecular foot prints, indicating that these are not redundant pathways, but individualistic—sharing some but also exhibiting different downstream targets (Figure 1). They are localized to different regions of the muscle cell—PDE5A at the z-disc, and PDE9A at the T-tubular membrane, and this likely contributes to these compartmentalized signalling effects.

6. Concluding remarks Knowledge about cGMP-PKG signalling has vastly expanded since the discovery of cGMP and PKG in the late 1960s.120 Despite the exponential increase in knowledge and the unravelling of detailed signalling properties, translation of successful experimental studies into efficacious clinical HF therapies has failed thus far. With discovering new layers of complexity and PKG regulation, such as signalling in intracellular microdomains, redox regulation, and cGMP degradation, we gain some insight into why these studies may have failed. The NO/ sGC, NP/pGC – cGMP – PDE5/9 molecules are all druggable and potentially excellent candidates for therapeutic interventions. Many pharmacologic compounds targeting this signalling are already in clinical use for other diseases; further research and a precise understanding of the signalling network will identify conditions and patient subsets which will likely benefit from modulation and thus probably enable successful translation to the clinic. The new therapeutic compound LCZ696,121 a neprilysin inhibitor, which prevents degradation of NPs and thus enhances pGC-GMP signalling, demonstrated that there are still significant benefits to be gained by devising new therapies based on GC-cGMP-PKG signalling. Conflict of interest: D.A.K. received research support from Pfizer and serves as a consultant to Merck and Ironwood Pharmaceuticals.

Funding This work was supported by the Medical University of Graz, the Austrian Academy of Sciences, and the Austrian Society of Cardiology (to P.P.R.) and the National Heart, Lung, and Blood Institute (NHLBI: HL119012 and HL107153 to D.A.K.).

References 1. Tokudome T, Kishimoto I, Horio T, Arai Y, Schwenke DO, Hino J, Okano I, Kawano Y, Kohno M, Miyazato M, Nakao K, Kangawa K. Regulator of G-protein signaling subtype 4 mediates antihypertrophic effect of locally secreted natriuretic peptides in the heart. Circulation 2008;117:2329 –2339. 2. Takimoto E, Koitabashi N, Hsu S, Ketner EA, Zhang M, Nagayama T, Bedja D, Gabrielson KL, Blanton R, Siderovski DP, Mendelsohn ME, Kass DA. Regulator of G protein signaling 2 mediates cardiac compensation to pressure overload and antihypertrophic effects of PDE5 inhibition in mice. J Clin Invest 2009;119:408 –420. 3. Ranek MJ, Terpstra EJ, Li J, Kass DA, Wang X. Protein kinase G positively regulates proteasome-mediated degradation of misfolded proteins. Circulation 2013;128: 365 –376. 4. Fisher PW, Salloum F, Das A, Hyder H, Kukreja RC. Phosphodiesterase-5 inhibition with sildenafil attenuates cardiomyocyte apoptosis and left ventricular dysfunction in a chronic model of doxorubicin cardiotoxicity. Circulation 2005;111:1601 – 1610.

159 5. Das A, Xi L, Kukreja RC. Phosphodiesterase-5 inhibitor sildenafil preconditions adult cardiac myocytes against necrosis and apoptosis. Essential role of nitric oxide signaling. J Biol Chem 2005;280:12944– 12955. 6. Lee DI, Zhu G, Sasaki T, Cho GS, Hamdani N, Holewinski R, Jo SH, Danner T, Zhang M, Rainer PP, Bedja D, Kirk JA, Ranek MJ, Dostmann WR, Kwon C, Margulies KB, Van Eyk JE, Paulus WJ, Takimoto E, Kass DA. Phosphodiesterase 9A controls nitric-oxide-independent cGMP and hypertrophic heart disease. Nature 2015;519:472 –476. 7. Layland J, Li JM, Shah AM. Role of cyclic GMP-dependent protein kinase in the contractile response to exogenous nitric oxide in rat cardiac myocytes. J Physiol 2002; 540:457 –467. 8. Lee DI, Vahebi S, Tocchetti CG, Barouch LA, Solaro RJ, Takimoto E, Kass DA. PDE5A suppression of acute beta-adrenergic activation requires modulation of myocyte beta-3 signaling coupled to PKG-mediated troponin I phosphorylation. Basic Res Cardiol 2010;105:337 – 347. 9. Jin CZ, Jang JH, Kim HJ, Wang Y, Hwang IC, Sadayappan S, Park BM, Kim SH, Jin ZH, Seo EY, Kim KH, Kim YJ, Kim SJ, Zhang YH. Myofilament Ca2+ desensitization mediates positive lusitropic effect of neuronal nitric oxide synthase in left ventricular myocytes from murine hypertensive heart. J Mol Cell Cardiol 2013;60: 107 – 115. 10. Koitabashi N, Aiba T, Hesketh GG, Rowell J, Zhang M, Takimoto E, Tomaselli GF, Kass DA. Cyclic GMP/PKG-dependent inhibition of TRPC6 channel activity and expression negatively regulates cardiomyocyte NFAT activation Novel mechanism of cardiac stress modulation by PDE5 inhibition. J Mol Cell Cardiol 2010;48: 713 – 724. 11. Seo K, Rainer PP, Lee DI, Hao S, Bedja D, Birnbaumer L, Cingolani OH, Kass DA. Hyperactive adverse mechanical stress responses in dystrophic heart are coupled to transient receptor potential canonical 6 and blocked by cGMP-protein kinase G modulation. Circ Res 2014;114:823–832. 12. Nakamura T, Ranek MJ, Lee DI, Shalkey Hahn V, Kim C, Eaton P, Kass DA. Prevention of PKG1alpha oxidation augments cardioprotection in the stressed heart. J Clin Invest 2015;125:2468 –2472. 13. Li P, Wang D, Lucas J, Oparil S, Xing D, Cao X, Novak L, Renfrow MB, Chen YF. Atrial natriuretic peptide inhibits transforming growth factor beta-induced Smad signaling and myofibroblast transformation in mouse cardiac fibroblasts. Circ Res 2008;102: 185– 192. 14. Chen J, Shearer GC, Chen Q, Healy CL, Beyer AJ, Nareddy VB, Gerdes AM, Harris WS, O’Connell TD, Wang D. Omega-3 fatty acids prevent pressure overload-induced cardiac fibrosis through activation of cyclic GMP/protein kinase G signaling in cardiac fibroblasts. Circulation 2011;123:584 –593. 15. Kruger M, Kotter S, Grutzner A, Lang P, Andresen C, Redfield MM, Butt E, dos Remedios CG, Linke WA. Protein kinase G modulates human myocardial passive stiffness by phosphorylation of the titin springs. Circ Res 2009;104:87–94. 16. Kinoshita H, Kuwahara K, Nishida M, Jian Z, Rong X, Kiyonaka S, Kuwabara Y, Kurose H, Inoue R, Mori Y, Li Y, Nakagawa Y, Usami S, Fujiwara M, Yamada Y, Minami T, Ueshima K, Nakao K. Inhibition of TRPC6 channel activity contributes to the antihypertrophic effects of natriuretic peptides-guanylyl cyclase-A signaling in the heart. Circ Res 2010;106:1849 – 1860. 17. Wu X, Eder P, Chang B, Molkentin JD. TRPC channels are necessary mediators of pathologic cardiac hypertrophy. Proc Natl Acad Sci USA 2010;107:7000 –7005. 18. Bush EW, Hood DB, Papst PJ, Chapo JA, Minobe W, Bristow MR, Olson EN, McKinsey TA. Canonical transient receptor potential channels promote cardiomyocyte hypertrophy through activation of calcineurin signaling. J Biol Chem 2006;281: 33487– 33496. 19. Kuwahara K, Wang Y, McAnally J, Richardson JA, Bassel-Duby R, Hill JA, Olson EN. TRPC6 fulfills a calcineurin signaling circuit during pathologic cardiac remodeling. J Clin Invest 2006;116:3114 –3126. 20. Nakayama H, Wilkin BJ, Bodi I, Molkentin JD. Calcineurin-dependent cardiomyopathy is activated by TRPC in the adult mouse heart. FASEB J 2006;20:1660 –1670. 21. Onohara N, Nishida M, Inoue R, Kobayashi H, Sumimoto H, Sato Y, Mori Y, Nagao T, Kurose H. TRPC3 and TRPC6 are essential for angiotensin II-induced cardiac hypertrophy. EMBO J 2006;25:5305 –5316. 22. Eder P, Molkentin JD. TRPC channels as effectors of cardiac hypertrophy. Circ Res 2011;108:265 –272. 23. Seo K, Rainer PP, Shalkey Hahn V, Lee DI, Jo SH, Andersen A, Liu T, Xu X, Willette RN, Lepore JJ, Marino JP Jr, Birnbaumer L, Schnackenberg CG, Kass DA. Combined TRPC3 and TRPC6 blockade by selective small-molecule or genetic deletion inhibits pathological cardiac hypertrophy. Proc Natl Acad Sci USA 2014;111: 1551 –1556. 24. Davis J, Burr AR, Davis GF, Birnbaumer L, Molkentin JD. A TRPC6-dependent pathway for myofibroblast transdifferentiation and wound healing in vivo. Dev Cell 2012; 23:705 –715. 25. Domes K, Patrucco E, Loga F, Dietrich A, Birnbaumer L, Wegener JW, Hofmann F. Murine cardiac growth, TRPC channels, and cGMP kinase I. Pflugers Arch 2015;467: 2229 –2234. 26. Takahashi S, Lin H, Geshi N, Mori Y, Kawarabayashi Y, Takami N, Mori MX, Honda A, Inoue R. Nitric oxide-cGMP-protein kinase G pathway negatively

160

27.

28.

29.

30. 31. 32. 33.

34.

35. 36.

37.

38.

39.

40.

41.

42.

43.

44.

45.

46.

47.

48. 49.

50.

regulates vascular transient receptor potential channel TRPC6. J Physiol 2008;586: 4209 – 4223. Kwan HY, Huang Y, Yao X. Regulation of canonical transient receptor potential isoform 3 (TRPC3) channel by protein kinase G. Proc Natl Acad Sci USA 2004;101: 2625 –2630. Nishida M, Watanabe K, Sato Y, Nakaya M, Kitajima N, Ide T, Inoue R, Kurose H. Phosphorylation of TRPC6 channels at Thr69 is required for anti-hypertrophic effects of phosphodiesterase 5 inhibition. J Biol Chem 2010;285:13244 –13253. Hall G, Rowell J, Farinelli F, Gbadegesin RA, Lavin P, Wu G, Homstad A, Malone A, Lindsey T, Jiang R, Spurney R, Tomaselli GF, Kass DA, Winn MP. Phosphodiesterase 5 inhibition ameliorates angiontensin II-induced podocyte dysmotility via the protein kinase G-mediated downregulation of TRPC6 activity. Am J Physiol Renal Physiol 2014; 306:F1442– F1450. Hibberd MG, Jewell BR. Calcium- and length-dependent force production in rat ventricular muscle. J Physiol 1982;329:527 –540. Shah AM, Spurgeon HA, Sollott SJ, Talo A, Lakatta EG. 8-bromo-cGMP reduces the myofilament response to Ca2+ in intact cardiac myocytes. Circ Res 1994;74:970 –978. Seddon M, Shah AM, Casadei B. Cardiomyocytes as effectors of nitric oxide signalling. Cardiovasc Res 2007;75:315 –326. Yasuda S, Coutu P, Sadayappan S, Robbins J, Metzger JM. Cardiac transgenic and gene transfer strategies converge to support an important role for troponin I in regulating relaxation in cardiac myocytes. Circ Res 2007;101:377 –386. Thoonen R, Giovanni S, Govindan S, Lee DI, Wang GR, Calamaras TD, Takimoto E, Kass DA, Sadayappan S, Blanton RM. Molecular screen identifies cardiac myosinbinding protein-C as a protein kinase G-Ialpha substrate. Circ Heart Fail 2015;8: 1115 –1122. von Anrep G. On the part played by the suprarenals in the normal vascular reactions of the body. J Physiol 1912;45:307 –317. Zou Y, Akazawa H, Qin Y, Sano M, Takano H, Minamino T, Makita N, Iwanaga K, Zhu W, Kudoh S, Toko H, Tamura K, Kihara M, Nagai T, Fukamizu A, Umemura S, Iiri T, Fujita T, Komuro I. Mechanical stress activates angiotensin II type 1 receptor without the involvement of angiotensin II. Nat Cell Biol 2004;6:499 –506. Cingolani HE, Perez NG, Aiello EA, Ennis IL, Garciarena CD, Villa-Abrille MC, Dulce RA, Caldiz CI, Yeves AM, Correa MV, Nolly MB, Chiappe de Cingolani G. Early signals after stretch leading to cardiac hypertrophy. Key role of NHE-1. Front Biosci 2008;13:7096 –7114. Perez NG, de Hurtado MC, Cingolani HE. Reverse mode of the Na+-Ca2+ exchange after myocardial stretch: underlying mechanism of the slow force response. Circ Res 2001;88:376 –382. Villa-Abrille MC, Caldiz CI, Ennis IL, Nolly MB, Casarini MJ, Chiappe de Cingolani GE, Cingolani HE, Perez NG. The Anrep effect requires transactivation of the epidermal growth factor receptor. J Physiol 2010;588:1579 –1590. von Lewinski D, Stumme B, Fialka F, Luers C, Pieske B. Functional relevance of the stretch-dependent slow force response in failing human myocardium. Circ Res 2004; 94:1392 –1398. Kockskamper J, Khafaga M, Grimm M, Elgner A, Walther S, Kockskamper A, von Lewinski D, Post H, Grossmann M, Dorge H, Gottlieb PA, Sachs F, Eschenhagen T, Schondube FA, Pieske B. Angiotensin II and myosin light-chain phosphorylation contribute to the stretch-induced slow force response in human atrial myocardium. Cardiovasc Res 2008;79:642 – 651. Dyachenko V, Husse B, Rueckschloss U, Isenberg G. Mechanical deformation of ventricular myocytes modulates both TRPC6 and Kir2.3 channels. Cell Calcium 2009;45: 38 –54. Ward ML, Williams IA, Chu Y, Cooper PJ, Ju YK, Allen DG. Stretch-activated channels in the heart: contributions to length-dependence and to cardiomyopathy. Prog Biophys Mol Biol 2008;97:232 –249. Spassova MA, Hewavitharana T, Xu W, Soboloff J, Gill DL. A common mechanism underlies stretch activation and receptor activation of TRPC6 channels. Proc Natl Acad Sci USA 2006;103:16586– 16591. Lukowski R, Rybalkin SD, Loga F, Leiss V, Beavo JA, Hofmann F. Cardiac hypertrophy is not amplified by deletion of cGMP-dependent protein kinase I in cardiomyocytes. Proc Natl Acad Sci USA 2010;107:5646 –5651. Klaiber M, Kruse M, Volker K, Schroter J, Feil R, Freichel M, Gerling A, Feil S, Dietrich A, Londono JE, Baba HA, Abramowitz J, Birnbaumer L, Penninger JM, Pongs O, Kuhn M. Novel insights into the mechanisms mediating the local antihypertrophic effects of cardiac atrial natriuretic peptide: role of cGMP-dependent protein kinase and RGS2. Basic Res Cardiol 2010;105:583 –595. Zhang P, Ma Y, Wang Y, Ma X, Huang Y, Li RA, Wan S, Yao X. Nitric oxide and protein kinase G act on TRPC1 to inhibit 11,12-EET-induced vascular relaxation. Cardiovasc Res 2014;104:138 – 146. Willis MS, Patterson C. Proteotoxicity and cardiac dysfunction—Alzheimer’s disease of the heart? N Engl J Med 2013;368:455 –464. Dey NB, Busch JL, Francis SH, Corbin JD, Lincoln TM. Cyclic GMP specifically suppresses type-Ialpha cGMP-dependent protein kinase expression by ubiquitination. Cell Signal 2009;21:859 –866. Gong W, Duan Q, Cai Z, Chen C, Ni L, Yan M, Wang X, Cianflone K, Wang DW. Chronic inhibition of cGMP-specific phosphodiesterase 5 suppresses endoplasmic reticulum stress in heart failure. Br J Pharmacol 2013;170:1396 –1409.

P.P. Rainer and D.A. Kass

51. Shi Z, Fu F, Yu L, Xing W, Su F, Liang X, Tie R, Ji L, Zhu M, Yu J, Zhang H. Vasonatrin peptide attenuates myocardial ischemia-reperfusion injury in diabetic rats and underlying mechanisms. Am J Physiol Heart Circ Physiol 2015;308:H281–H290. 52. Karbach S, Wenzel P, Waisman A, Munzel T, Daiber A. eNOS uncoupling in cardiovascular diseases—the role of oxidative stress and inflammation. Curr Pharm Des 2014; 20:3579 –3594. 53. Stasch JP, Schmidt PM, Nedvetsky PI, Nedvetskaya TY, Ak HS, Meurer S, Deile M, Taye A, Knorr A, Lapp H, Muller H, Turgay Y, Rothkegel C, Tersteegen A, Kemp-Harper B, Muller-Esterl W, Schmidt HH. Targeting the heme-oxidized nitric oxide receptor for selective vasodilatation of diseased blood vessels. J Clin Invest 2006;116:2552 –2561. 54. Takimoto E, Champion HC, Li M, Ren S, Rodriguez ER, Tavazzi B, Lazzarino G, Paolocci N, Gabrielson KL, Wang Y, Kass DA. Oxidant stress from nitric oxide synthase-3 uncoupling stimulates cardiac pathologic remodeling from chronic pressure load. J Clin Invest 2005;115:1221 –1231. 55. Tsai EJ, Liu Y, Koitabashi N, Bedja D, Danner T, Jasmin JF, Lisanti MP, Friebe A, Takimoto E, Kass DA. Pressure-overload-induced subcellular relocalization/oxidation of soluble guanylyl cyclase in the heart modulates enzyme stimulation. Circ Res 2012; 110:295 –303. 56. Burgoyne JR, Madhani M, Cuello F, Charles RL, Brennan JP, Schroder E, Browning DD, Eaton P. Cysteine redox sensor in PKGIa enables oxidant-induced activation. Science 2007;317:1393 –1397. 57. Prysyazhna O, Rudyk O, Eaton P. Single atom substitution in mouse protein kinase G eliminates oxidant sensing to cause hypertension. Nat Med 2012;18: 286 – 290. 58. Surks HK, Mochizuki N, Kasai Y, Georgescu SP, Tang KM, Ito M, Lincoln TM, Mendelsohn ME. Regulation of myosin phosphatase by a specific interaction with cGMP-dependent protein kinase Ialpha. Science 1999;286:1583 –1587. 59. Kato M, Blanton R, Wang GR, Judson TJ, Abe Y, Myoishi M, Karas RH, Mendelsohn ME. Direct binding and regulation of RhoA protein by cyclic GMP-dependent protein kinase Ialpha. J Biol Chem 2012;287:41342 –41351. 60. Yuasa K, Michibata H, Omori K, Yanaka N. A novel interaction of cGMP-dependent protein kinase I with troponin T. J Biol Chem 1999;274:37429 –37434. 61. Michael SK, Surks HK, Wang Y, Zhu Y, Blanton R, Jamnongjit M, Aronovitz M, Baur W, Ohtani K, Wilkerson MK, Bonev AD, Nelson MT, Karas RH, Mendelsohn ME. High blood pressure arising from a defect in vascular function. Proc Natl Acad Sci USA 2008;105:6702 –6707. 62. Burgoyne JR, Eaton P. Transnitrosylating nitric oxide species directly activate type I protein kinase A, providing a novel adenylate cyclase-independent cross-talk to beta-adrenergic-like signaling. J Biol Chem 2009;284:29260 –29268. 63. Stubbert D, Prysyazhna O, Rudyk O, Scotcher J, Burgoyne JR, Eaton P. Protein kinase G Ialpha oxidation paradoxically underlies blood pressure lowering by the reductant hydrogen sulfide. Hypertension 2014;64:1344 –1351. 64. Burgoyne JR, Prysyazhna O, Rudyk O, Eaton P. cGMP-Dependent activation of protein kinase G precludes disulfide activation: implications for blood pressure control. Hypertension 2012;60:1301 –1308. 65. Rudyk O, Prysyazhna O, Burgoyne JR, Eaton P. Nitroglycerin fails to lower blood pressure in redox-dead Cys42Ser PKG1alpha knock-in mouse. Circulation 2012;126: 287– 295. 66. Rudyk O, Phinikaridou A, Prysyazhna O, Burgoyne JR, Botnar RM, Eaton P. Protein kinase G oxidation is a major cause of injury during sepsis. Proc Natl Acad Sci USA 2013;110:9909 –9913. 67. Dou D, Zheng X, Liu J, Xu X, Ye L, Gao Y. Hydrogen peroxide enhances vasodilatation by increasing dimerization of cGMP-dependent protein kinase type Ialpha. Circ J 2012;76:1792 –1798. 68. Zhang DX, Borbouse L, Gebremedhin D, Mendoza SA, Zinkevich NS, Li R, Gutterman DD. H2O2-Induced dilation in human coronary arterioles: role of protein kinase G dimerization and large-conductance Ca2+-activated K+ channel activation. Circ Res 2012;110:471 –480. 69. Akashi S, Ahmed KA, Sawa T, Ono K, Tsutsuki H, Burgoyne JR, Ida T, Horio E, Prysyazhna O, Oike Y, Rahaman MM, Eaton P, Fujii S, Akaike T. Persistent activation of cGMP-dependent protein kinase by a nitrated cyclic nucleotide via site specific protein S-guanylation. Biochemistry 2016;55:751 –761. 70. Graham S, Ding M, Ding Y, Sours-Brothers S, Luchowski R, Gryczynski Z, Yorio T, Ma H, Ma R. Canonical transient receptor potential 6 (TRPC6), a redox-regulated cation channel. J Biol Chem 2010;285:23466 –23476. 71. Conti M, Beavo J. Biochemistry and physiology of cyclic nucleotide phosphodiesterases: essential components in cyclic nucleotide signaling. Annu Rev Biochem 2007; 76:481 – 511. 72. Maurice DH, Ke H, Ahmad F, Wang Y, Chung J, Manganiello VC. Advances in targeting cyclic nucleotide phosphodiesterases. Nat Rev Drug Discov 2014;13: 290 – 314. 73. Sastry BK, Narasimhan C, Reddy NK, Raju BS. Clinical efficacy of sildenafil in primary pulmonary hypertension: a randomized, placebo-controlled, double-blind, crossover study. J Am Coll Cardiol 2004;43:1149 –1153. 74. Goldstein I, Lue TF, Padma-Nathan H, Rosen RC, Steers WD, Wicker PA. Oral sildenafil in the treatment of erectile dysfunction. Sildenafil Study Group. N Engl J Med 1998;338:1397 –1404.

Novel stress regulation by protein kinase G

75. Shan X, Quaile MP, Monk JK, French B, Cappola TP, Margulies KB. Differential expression of PDE5 in failing and nonfailing human myocardium. Circ Heart Fail 2012; 5:79 – 86. 76. Nagendran J, Archer SL, Soliman D, Gurtu V, Moudgil R, Haromy A, St Aubin C, Webster L, Rebeyka IM, Ross DB, Light PE, Dyck JR, Michelakis ED. Phosphodiesterase type 5 is highly expressed in the hypertrophied human right ventricle, and acute inhibition of phosphodiesterase type 5 improves contractility. Circulation 2007;116: 238 –248. 77. Straubinger J, Schottle V, Bork N, Subramanian H, Dunnes S, Russwurm M, Gawaz M, Friebe A, Nemer M, Nikolaev VO, Lukowski R. Sildenafil does not prevent heart hypertrophy and fibrosis induced by cardiomyocyte angiotensin II type 1 receptor signaling. J Pharmacol Exp Ther 2015;354:406 –416. 78. Degen CV, Bishu K, Zakeri R, Ogut O, Redfield MM, Brozovich FV. The emperor’s new clothes: PDE5 and the heart. PLoS One 2015;10:e0118664. 79. Kass DA, Takimoto E. Regulation and role of myocyte cyclic GMP-dependent protein kinase-1. Proc Natl Acad Sci USA 2010;107:E98; author reply E99. 80. Patrucco E, Domes K, Sbroggio M, Blaich A, Schlossmann J, Desch M, Rybalkin SD, Beavo JA, Lukowski R, Hofmann F. Roles of cGMP-dependent protein kinase I (cGKI) and PDE5 in the regulation of Ang II-induced cardiac hypertrophy and fibrosis. Proc Natl Acad Sci USA 2014;111:12925 –12929. 81. Pfeifer A, Klatt P, Massberg S, Ny L, Sausbier M, Hirneiss C, Wang GX, Korth M, Aszodi A, Andersson KE, Krombach F, Mayerhofer A, Ruth P, Fassler R, Hofmann F. Defective smooth muscle regulation in cGMP kinase I-deficient mice. EMBO J 1998;17:3045 – 3051. 82. Weber S, Bernhard D, Lukowski R, Weinmeister P, Worner R, Wegener JW, Valtcheva N, Feil S, Schlossmann J, Hofmann F, Feil R. Rescue of cGMP kinase I knockout mice by smooth muscle specific expression of either isozyme. CircRes 2007;101: 1096 –1103. 83. Pokreisz P, Vandenwijngaert S, Bito V, Van den BA, Lenaerts I, Busch C, Marsboom G, Gheysens O, Vermeersch P, Biesmans L, Liu X, Gillijns H, Pellens M, Van Lommel A, Buys E, Schoonjans L, Vanhaecke J, Verbeken E, Sipido K, Herijgers P, Bloch KD, Janssens SP. Ventricular phosphodiesterase-5 expression is increased in patients with advanced heart failure and contributes to adverse ventricular remodeling after myocardial infarction in mice. Circulation 2009;119:408–416. 84. Zhang M, Takimoto E, Hsu S, Lee DI, Nagayama T, Danner T, Koitabashi N, Barth AS, Bedja D, Gabrielson KL, Wang Y, Kass DA. Myocardial remodeling is controlled by myocyte-targeted gene regulation of phosphodiesterase type 5. J Am Coll Cardiol 2010;56:2021 –2030. 85. Frantz S, Klaiber M, Baba HA, Oberwinkler H, Volker K, Gabetaner B, Bayer B, Abebetaer M, Schuh K, Feil R, Hofmann F, Kuhn M. Stress-dependent dilated cardiomyopathy in mice with cardiomyocyte-restricted inactivation of cyclic GMP-dependent protein kinase I. Eur Heart J 2013;34:1233 –1244. 86. Adamo CM, Dai DF, Percival JM, Minami E, Willis MS, Patrucco E, Froehner SC, Beavo JA. Sildenafil reverses cardiac dysfunction in the mdx mouse model of Duchenne muscular dystrophy. Proc Natl Acad Sci USA 2010;107:19079 –19083. 87. Khairallah M, Khairallah RJ, Young ME, Allen BG, Gillis MA, Danialou G, Deschepper CF, Petrof BJ, Des Rosiers C. Sildenafil and cardiomyocyte-specific cGMP signaling prevent cardiomyopathic changes associated with dystrophin deficiency. Proc Natl Acad Sci USA 2008;105:7028 – 7033. 88. Ascah A, Khairallah M, Daussin F, Bourcier-Lucas C, Godin R, Allen BG, Petrof BJ, Des Rosiers C, Burelle Y. Stress-induced opening of the permeability transition pore in the dystrophin-deficient heart is attenuated by acute treatment with sildenafil. Am J Physiol Heart Circ Physiol 2011;300:H144–H153. 89. Takimoto E, Champion HC, Li M, Belardi D, Ren S, Rodriguez ER, Bedja D, Gabrielson KL, Wang Y, Kass DA. Chronic inhibition of cyclic GMP phosphodiesterase 5A prevents and reverses cardiac hypertrophy. Nat Med 2005;11: 214 – 222. 90. Ockaili R, Salloum F, Hawkins J, Kukreja RC. Sildenafil (Viagra) induces powerful cardioprotective effect via opening of mitochondrial K(ATP) channels in rabbits. Am J Physiol Heart Circ Physiol 2002;283:H1263 –H1269. 91. Salloum F, Yin C, Xi L, Kukreja RC. Sildenafil induces delayed preconditioning through inducible nitric oxide synthase-dependent pathway in mouse heart. Circ Res 2003;92: 595 –597. 92. Das S, Maulik N, Das DK, Kadowitz PJ, Bivalacqua TJ. Cardioprotection with sildenafil, a selective inhibitor of cyclic 3′ ,5′ -monophosphate-specific phosphodiesterase 5. Drugs Exp Clin Res 2002;28:213 –219. 93. Koka S, Das A, Zhu SG, Durrant D, Xi L, Kukreja RC. Long-acting phosphodiesterase-5 inhibitor tadalafil attenuates doxorubicin-induced cardiomyopathy without interfering with chemotherapeutic effect. J Pharmacol Exp Ther 2010;334: 1023 –1030. 94. Das A, Samidurai A, Hoke NN, Kukreja RC, Salloum FN. Hydrogen sulfide mediates the cardioprotective effects of gene therapy with PKG-Ialpha. Basic Res Cardiol 2015; 110:42. 95. Bibli SI, Andreadou I, Chatzianastasiou A, Tzimas C, Sanoudou D, Kranias E, Brouckaert P, Coletta C, Szabo C, Kremastinos DT, Iliodromitis EK, Papapetropoulos A. Cardioprotection by H2S engages a cGMP-dependent protein kinase G/phospholamban pathway. Cardiovasc Res 2015;106:432 –442.

161 96. Inserte J, Garcia-Dorado D. The cGMP/PKG pathway as a common mediator of cardioprotection: translatability and mechanism. Br J Pharmacol 2015;172:1996 –2009. 97. Guazzi M, Vicenzi M, Arena R, Guazzi MD. Pulmonary hypertension in heart failure with preserved ejection fraction: a target of phosphodiesterase-5 inhibition in a 1-year study. Circulation 2011;124:164–174. 98. Guazzi M, Vicenzi M, Arena R, Guazzi MD. PDE5 inhibition with sildenafil improves left ventricular diastolic function, cardiac geometry, and clinical status in patients with stable systolic heart failure: results of a 1-year, prospective, randomized, placebocontrolled study. Circ Heart Fail 2011;4:8 –17. 99. Lewis GD, Shah R, Shahzad K, Camuso JM, Pappagianopoulos PP, Hung J, Tawakol A, Gerszten RE, Systrom DM, Bloch KD, Semigran MJ. Sildenafil improves exercise capacity and quality of life in patients with systolic heart failure and secondary pulmonary hypertension. Circulation 2007;116:1555 –1562. 100. Redfield MM, Chen HH, Borlaug BA, Semigran MJ, Lee KL, Lewis G, LeWinter MM, Rouleau JL, Bull DA, Mann DL, Deswal A, Stevenson LW, Givertz MM, Ofili EO, O’Connor CM, Felker GM, Goldsmith SR, Bart BA, McNulty SE, Ibarra JC, Lin G, Oh JK, Patel MR, Kim RJ, Tracy RP, Velazquez EJ, Anstrom KJ, Hernandez AF, Mascette AM, Braunwald E, Trial R. Effect of phosphodiesterase-5 inhibition on exercise capacity and clinical status in heart failure with preserved ejection fraction: a randomized clinical trial. JAMA 2013;309:1268 – 1277. 101. van Heerebeek L, Hamdani N, Falcao-Pires I, Leite-Moreira AF, Begieneman MP, Bronzwaer JG, van der Velden J, Stienen GJ, Laarman GJ, Somsen A, Verheugt FW, Niessen HW, Paulus WJ. Low myocardial protein kinase G activity in heart failure with preserved ejection fraction. Circulation 2012;126:830 –839. 102. Borlaug BA, Lewis GD, McNulty SE, Semigran MJ, LeWinter M, Chen H, Lin G, Deswal A, Margulies KB, Redfield MM. Effects of sildenafil on ventricular and vascular function in heart failure with preserved ejection fraction. Circ Heart Fail 2015;8: 533 –541. 103. Borlaug BA, Melenovsky V, Marhin T, Fitzgerald P, Kass DA. Sildenafil inhibits beta-adrenergic-stimulated cardiac contractility in humans. Circulation 2005;112: 2642 –2649. 104. Senzaki H, Smith CJ, Juang GJ, Isoda T, Mayer SP, Ohler A, Paolocci N, Tomaselli GF, Hare JM, Kass DA. Cardiac phosphodiesterase 5 (cGMP-specific) modulates beta-adrenergic signaling in vivo and is down-regulated in heart failure. FASEB J 2001;15:1718 –1726. 105. Takimoto E, Champion HC, Belardi D, Moslehi J, Mongillo M, Mergia E, Montrose DC, Isoda T, Aufiero K, Zaccolo M, Dostmann WR, Smith CJ, Kass DA. cGMP catabolism by phosphodiesterase 5A regulates cardiac adrenergic stimulation by NOS3dependent mechanism. Circ Res 2005;96:100 –109. 106. Leung DG, Herzka DA, Thompson WR, He B, Bibat G, Tennekoon G, Russell SD, Schuleri KH, Lardo AC, Kass DA, Thompson RE, Judge DP, Wagner KR. Sildenafil does not improve cardiomyopathy in Duchenne/Becker muscular dystrophy. Ann Neurol 2014;76:541 – 549. 107. Witting N, Kruuse C, Nyhuus B, Prahm KP, Citirak G, Lundgaard SJ, von Huth S, Vejlstrup N, Lindberg U, Krag TO, Vissing J. Effect of sildenafil on skeletal and cardiac muscle in Becker muscular dystrophy. Ann Neurol 2014;76:550 – 557. 108. Takimoto E, Belardi D, Tocchetti CG, Vahebi S, Cormaci G, Ketner EA, Moens AL, Champion HC, Kass DA. Compartmentalization of cardiac beta-adrenergic inotropy modulation by phosphodiesterase type 5. Circulation 2007;115:2159 –2167. 109. Castro LR, Verde I, Cooper DM, Fischmeister R. Cyclic guanosine monophosphate compartmentation in rat cardiac myocytes. Circulation 2006;113:2221 –2228. 110. Sasaki H, Nagayama T, Blanton RM, Seo K, Zhang M, Zhu G, Lee DI, Bedja D, Hsu S, Tsukamoto O, Takashima S, Kitakaze M, Mendelsohn ME, Karas RH, Kass DA, Takimoto E. PDE5 inhibitor efficacy is estrogen dependent in female heart disease. J Clin Invest 2014;124:2464 – 2471. 111. Bubb KJ, Trinder SL, Baliga RS, Patel J, Clapp LH, MacAllister RJ, Hobbs AJ. Inhibition of phosphodiesterase 2 augments cGMP and cAMP signaling to ameliorate pulmonary hypertension. Circulation 2014;130:496–507. 112. Zoccarato A, Surdo NC, Aronsen JM, Fields LA, Mancuso L, Dodoni G, Stangherlin A, Livie C, Jiang H, Sin YY, Gesellchen F, Terrin A, Baillie GS, Nicklin SA, Graham D, Szabo-Fresnais N, Krall J, Vandeput F, Movsesian M, Furlan L, Corsetti V, Hamilton G, Lefkimmiatis K, Sjaastad I, Zaccolo M. Cardiac hypertrophy is inhibited by a local pool of cAMP regulated by phosphodiesterase 2. Circ Res 2015;117: 707– 719. 113. Mongillo M, Tocchetti CG, Terrin A, Lissandron V, Cheung YF, Dostmann WR, Pozzan T, Kass DA, Paolocci N, Houslay MD, Zaccolo M. Compartmentalized phosphodiesterase-2 activity blunts beta-adrenergic cardiac inotropy via an NO/ cGMP-dependent pathway. Circ Res 2006;98:226 –234. 114. Mehel H, Emons J, Vettel C, Wittkopper K, Seppelt D, Dewenter M, Lutz S, Sossalla S, Maier LS, Lechene P, Leroy J, Lefebvre F, Varin A, Eschenhagen T, Nattel S, Dobrev D, Zimmermann WH, Nikolaev VO, Vandecasteele G, Fischmeister R, El-Armouche A. Phosphodiesterase-2 is up-regulated in human failing hearts and blunts beta-adrenergic responses in cardiomyocytes. J Am Coll Cardiol 2013;62:1596–1606. 115. Yan C, Zhao AZ, Bentley JK, Beavo JA. The calmodulin-dependent phosphodiesterase gene PDE1C encodes several functionally different splice variants in a tissue-specific manner. J Biol Chem 1996;271:25699 –25706. 116. Nagel DJ, Aizawa T, Jeon KI, Liu W, Mohan A, Wei H, Miano JM, Florio VA, Gao P, Korshunov VA, Berk BC, Yan C. Role of nuclear Ca2+/calmodulin-stimulated

162 phosphodiesterase 1A in vascular smooth muscle cell growth and survival. Circ Res 2006;98:777–784. 117. Miller CL, Oikawa M, Cai Y, Wojtovich AP, Nagel DJ, Xu X, Xu H, Florio V, Rybalkin SD, Beavo JA, Chen YF, Li JD, Blaxall BC, Abe J, Yan C. Role of Ca2+/ calmodulin-stimulated cyclic nucleotide phosphodiesterase 1 in mediating cardiomyocyte hypertrophy. Circ Res 2009;105:956 –964. 118. Fisher DA, Smith JF, Pillar JS, St Denis SH, Cheng JB. Isolation and characterization of PDE9A, a novel human cGMP-specific phosphodiesterase. J Biol Chem 1998;273: 15559– 15564.

P.P. Rainer and D.A. Kass

119. Soderling SH, Bayuga SJ, Beavo JA. Identification and characterization of a novel family of cyclic nucleotide phosphodiesterases. J Biol Chem 1998;273: 15553– 15558. 120. Kots AY, Martin E, Sharina IG, Murad F. A short history of cGMP, guanylyl cyclases, and cGMP-dependent protein kinases. Handb Exp Pharmacol 2009;1 –14. 121. McMurray JJ, Packer M, Desai AS, Gong J, Lefkowitz MP, Rizkala AR, Rouleau JL, Shi VC, Solomon SD, Swedberg K, Zile MR, Investigators P-H, Committees. Angiotensin-neprilysin inhibition versus enalapril in heart failure. N Engl J Med 2014; 371:993 –1004.

Old dog, new tricks: novel cardiac targets and stress regulation by protein kinase G.

The second messenger cyclic guanosine 3'5' monophosphate (cGMP) and its downstream effector protein kinase G (PKG) have been discovered more than 40 y...
423KB Sizes 1 Downloads 13 Views