Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Jun 1.
Published in final edited form as: J Cardiovasc Pharmacol. 2020 Jun;75(6):483–493. doi: 10.1097/FJC.0000000000000773

Myocardial Phosphodiesterases and their Role in cGMP Regulation

Brittany Dunkerly-Eyring 1, David A Kass 1
PMCID: PMC7585454  NIHMSID: NIHMS1635716  PMID: 31651671

Abstract

Cyclic nucleotide phosphodiesterases comprise an 11-member superfamily yielding near 100 isoform variants that hydrolyze cAMP or cGMP to their respective 5’-monophosphate form. Each plays a role in compartmentalized cyclic nucleotide signaling, with varying selectivity for each substrate, and conveying cell and intracellular specific localized control. This review focuses on the five PDEs expressed in the cardiac myocyte capable of hydrolyzing cGMP and that have been shown to play a role in cardiac physiological and pathological processes. PDE1, PDE2, and PDE3 catabolize cAMP as well, whereas PDE5 and PDE9 are cGMP selective. PDE3 and PDE5 are already in clinical use, the former for heart failure, and PDE1, PDE9, and PDE5 are all being actively studied for this indication in patients. Research in just the past few years has revealed many novel cardiac influences of each isoform, expanding the therapeutic potential from their selective pharmacological blockade or in some instances, activation. PDE1C inhibition was found to confer cell survival protection and enhance cardiac contractility, while PDE2 inhibition or activation induces beneficial effects in hypertrophied or failing hearts, respectively. PDE3 inhibition is already clinically employed to treat acute decompensated heart failure, though toxicity has precluded its long-term use. However, newer approaches including isoform specific allosteric modulation may change this. Lastly, inhibition of PDE5A and PDE9A counter pathological remodeling of the heart and are both being pursued in clinical trials. Here we discuss recent research advances in each of these PDEs, their impact on the myocardium, and cardiac therapeutic potential.

Introduction

Phosphodiesterases (PDEs) are a superfamily of enzymes that hydrolyze the cyclic nucleotides adenosine 3’,5’-cyclic monophosphate (cAMP) and/or cyclic guanosine 3’,5’-cyclic monophosphate (cGMP). Both cyclic nucleotides are generated within intracellular nanodomains by corresponding cyclases, and are in turn catabolized by members of the PDE superfamily. Both synthesis and catabolism of cAMP and cGMP are altered by physiological and pathological stress, and this plays essential homeostatic roles as well as contributes to heart disease. Therapeutic benefits from direct stimulation of either cardiac cyclic nucleotide, as by beta-adrenergic agonism, organo-nitrates or nitic oxide donors, soluble guanylyl cyclase activators, or natriuretic peptides, are clinically used to trigger associated signaling. One disadvantage of these approaches is their diffuse impact on many cells, such that cardiomyocyte regulation often takes a back seat to changes in blood pressure, heart rate, and other changes.

The alternative to enhancing cyclic nucleotide synthesis is to selectively block their hydrolysis by inhibiting the relevant PDEs. Despite there being only two primary cyclic nucleotides, there are >100 different PDE members/isoforms to modulate them. These differ primarily in their N-terminus regulatory domain1 to control localization and regulation, with the catalytic domain conferring substrate specificity 2, 3. PDEs are very amenable to family-member selective potent small molecule inhibition, and many such inhibitors are being used or studied as therapeutics (Supplemental Table 1). This selectivity has its limitations, most notably that isoform and splice variants in a given species are equally susceptible as they share common catalytic domains. Another is that the relevant cyclic nucleotide must first be synthesized in order for a particular PDE inhibitor to have an impact. This is not required when this synthesis is itself being stimulated.

There are seven PDEs so far reported to be expressed in myocardium. PDE1, 2, and 3 are dual substrate esterases, PDE5 and PDE9 are selective for cGMP, and PDE4 and PDE8 are selective for cAMP. Preclinical studies support a role for each of these species in the heart, while existing clinical data pertain to PDE3 and PDE5. Each are expressed in myocytes, and many are also expressed in fibroblasts, vascular smooth muscle, and in some cases endothelial cells (see Supplemental Table 2). Importantly, many of these PDEs can contribute to cyclic nucleotide dysregulation in diseased heart, and so have become therapeutic targets. In this review, we focus on PDEs capable of hydrolyzing cGMP, PDEs 1-3, 5, and 9, highlighting recent research revealing novel roles to normal physiology and contributions to heart disease.

Cardiac role of cyclic nucleotides and their associated protein kinases

Cyclic AMP and cGMP control a broad range of myocardial properties including heart rate, cell growth and survival, interstitial fibrosis, vascular tone, endothelial permeability and proliferation, and muscle contractility and lusitropy. Cyclic AMP is generated by adenylate cyclase (AC type 5 and type 6 in the heart) and activates one of three cognate proteins: protein kinase A (PKA) expressed as one of two isoforms PKA-I and PKA-II, or the exchange protein directly activated by cAMP (Epac). PKA-I is primarily engaged in adrenergic stimulated phosphorylation of proteins that control excitation-contraction coupling and sarcomere function. These include troponin I 4, titin 5, myosin binding protein C 6, 7, phospholamban 8, the ryanodine receptor (RyR2) 9, and the L-type calcium channel 10. Epac is a guanine nucleotide exchange factor (GEF) protein that activates calcium-calmodulin activated kinase II (CaMKII) to influence calcium cycling and gene transcription 11.

Cyclic GMP is generated by either one of two guanylyl cyclases; GC-1, which is stimulated by nitric oxide, or GC-A which resides in the intracellular domain of the natriuretic peptide receptor (NPR1). Cyclic GMP in turn activates cGK1α (also PKG-1α) by binding to N-terminus regulatory domains. This kinase phosphorylates many similar myocyte calcium regulatory and sarcomere proteins and specific residues as protein kinase A (e.g. phospholamban, TnI, titin, myosin binding protein C). However, contractility is not generally altered as calcium transients are minimally changed. cGK1α also opposes neurohormonal stimulation by phosphorylating and/or binding to regulator of G-protein signaling 2 and 4 (RGS2, RGS4) to counter Gq- and Gi-protein receptor coupled agonism 12, 13. cGK1α also phosphorylates transient potential receptor canonical type 6 (TRPC6) to block calcineurin/NFAT signaling14-16, RhoA to suppress Rho-kinase signaling 17, and tuberin (TSC2) to block pathological growth and enhance autophagy in myocytes with stress-activation of the mechanistic target of rapamycin complex-1 (mTORC1) 18. The latter appears particularly important, as genetic mutation of a single phosphorylation site (S1364 human) on TSC2 to prevent its phosphorylation (serine to alanine substitution) was sufficient to eliminate the antihypertrophic efficacy of cGK1α activation against pressure-overload stress in vitro and in vivo. Lastly, activated cGK1α also enhances protein quality control by increasing proteasome activity to enhance clearance of damaged ubiquitinated proteins19.

PDE1: From cGMP to cAMP modulation in the heart

PDE1 is Ca2+/calmodulin (CaM)-activated20 and a dual cAMP/cGMP esterase expressed as one of three different isoforms. Of these, PDE1A and 1C are expressed in the heart. PDE1A has a 25-fold lower Km for cGMP than cAMP 21 so is more cGMP-selective, whereas PDE1C has an equal affinity for both cyclic nucleotides 22. PDE1A represents the dominant cardiac isoform in mice and rats, whereas PDE1C predominates in larger mammals such as humans, dogs, and rabbits23. The N-terminus contains two Ca2+/CaM binding sites and a phosphorylation site at Ser120, the latter modified by PKA in PDE1A and CaMKII in PDE1B to suppress sensitivity to Ca2+/CaM and thus PDE activation. PDE1A and PDE1C expression are upregulated in hypertrophied rodent hearts and myocardium from human heart failure21, 23-25. In mice, β-AR stimulated hypertrophy is suppressed by PDE1 inhibition by a mechanism most compatible with cGK1 activation26. PDE1A is also upregulated in rodent myofibroblasts after myocardial infarction, and PDE1 inhibition blocks expression of pro-fibrotic genes27. The anti-fibrotic response involves both cGMP and cAMP signaling. This is summarized to the center-right in Figure 1.

Figure 1:

Figure 1:

Cardiac myocyte regulation by PDE1A and PDE1C. PDE1A hydrolyzes both cAMP and cGMP, with greater affinity for cGMP derived from either GC-A (natriuretic peptide coupled) GC-1 (nitric oxide coupled) cyclases. Cyclic GMP activates cGK1α which phosphorylates to inhibit TRPC3/6, and suppresses Gq-coupled protein receptors by stimulating RGS2/4 proteins. Both results in attenuated hypertrophic signaling. PDE1A inhibition also enhances a cAMP that activates Epac to suppress profibrotic transcriptional regulation. PDE1C has equal affinitity for cAMP and cGMP. Its calcium-dependent activation is coupled to calcium influx through TRPC3, and colocalizes with adenosine Type 2 receptors (A2R). PDE1C hydrolyzes the cAMP coupled to A2R stimulation. The consequent PKA stimulation suppresses cell death pathways, as well as stimulates a positive inotropic response. The latter is independent of cAMP coupled to β-adrenergic stimulation. Abbreviations: NOS- nitric oxide synthase, NFAT – nuclear factor of activated T-cells, AngII – angiotensin II, Phe – phenylephrine, ET-1 endothelin-1, Epac: exchange protein directly activated by cAMP; CamKII – calcium calmodulin-dependent kinase II, GPCR – G protein coupled receptor, NP – natriuretic peptide, NPR – NP receptor, Cm – calmodulin, Cn – calcineurin.

The selective role of PDE1C, the isoform most prominent in larger mammalian hearts, was first suggested in genetic PDE1C-knockout mice. Despite having very low levels of basal expression of the 1C isoform, myocytes lacking the gene were protected against cytotoxic stress-induced apoptosis, and in vivo hearts subjected to pressure-overload displayed less pathological hypertrophy, fibrosis, apoptosis, and dysfunction 24. Interestingly, this was not coupled to cGMP signaling enhancement, but rather to cAMP-PKA activation.

Two recent studies have further identified a critical role of PDE1C in controlling a cAMP-modulated signaling cascade28. Continuing their work, the Yan laboratory uncovered a key mechanism underlying PDE1C-cAMP dependent protection against myocyte stress induced apoptosis. A key feature was identifying the source of the regulated cAMP to be coupled to type A2 adenosine receptors (A2R), and for PDE1C activation to occur by Ca2+ delivered via transient receptor potential canonical cation channel type 3 (Trpc3)29. The authors identified a novel protein complex linking PDE1C, TRPC3, and A2R, providing a regulatory pathway to modulate adenosine-mediated anti-apoptotic cytoprotection via PDE1C inhibition.

In a separate study reported in the same journal issue, the Kass laboratory explored the cardiovascular impact of inhibiting PDE1 (all isoforms) in larger mammals23. In both dogs and rabbits, they showed PDE1C>>PDE1A expression as observed in humans but opposite the PDE1A>>PDE1C expression in mice and rats. Using the selective PDE1 inhibitor ITI-214, they found positive inotropic, lusitropic, chronotropic, and vasodilator effects from PDE1 blockade in normal conscious dogs, with all but heart rate change being preserved in the same animals when their hearts were induced into failure following 3-4 weeks of tachycardia pacing. Similar profiles were observed in intact rabbits. PDE1 inhibition did not augment dobutamine stimulation, nor were its effects prevented by marked beta-receptor blockade (with the exception of heart rate that was suppressed by this blockade). Rather, cardiac and systemic arterial dilator effects from ITI-214 were blocked by A2B receptor inhibition. In myocytes, PDE3 but not PDE1 inhibition augmented resting and isoproterenol stimulated cell contraction and calcium.

These data have shifted the focus of PDE1 regulation in the intact heart from one emphasizing cGMP to cAMP signaling, with the latter being coupled to adenosine and not β-AR receptors (Left side, Figure 1). The protective effects of A2 receptor stimulation against ischemic injury are well known30, and the new data suggest such protection maybe leveraged by PDE1 inhibition. Furthermore, the positive inotropy and lusitropy may differ from that coupled to β-AR-cAMP signaling, with the potential for a different safety/toxicity profile than observed with PDE3 inhibitors. A Phase Ia-IIb clinical trial testing safety and hemodynamic responses to single dose of ITI-214 (placebo+ dose escalation, Clinicaltrials.gov: NCT03387215) in patients with dilated heart failure initiated in mid 2018 and continues.

PDE2: Signaling ambiguities and alternative roles

PDE2 is a dual substrate phosphodiesterase that hydrolyzes cAMP and cGMP at similar maximal rates. A single gene Pde2a gives rise to three isoforms (PDE2A1, 2A2, and 2A3) which are differentially localized in cytosol, membrane, and mitochondrial sub-domains. A primary characteristic of PDE2A is its allosteric activation by binding of cGMP to GAF domains residing in the N-terminus that augments catalytic activity of cAMP by 10-30 fold. The reverse, cAMP mediated activation for cGMP hydrolysis, does not occur given the low affinity of the GAF domain for cAMP. This positions the PDE for particular relevance to cGMP/cAMP crosstalk.

In keeping with its regulatory capacity, studies have found the consequences of PDE2 regulation to vary depending on the precise conditions (Figure 2). In quiescent adult cardiomyocytes, PDE2 inhibition at the plasma membrane augments cGMP generated by guanylyl cyclase GC-A coupled to natriuretic peptide receptors (NPR1, NPR2), or by GC-1 which is activated by nitric-oxide. This was first revealed in myocytes expressing a cGMP-sensitive cation channel (Cnga2)31. The role of PDE2 in compartmentalizing cGMP generated by ANP stimulation of NPR1 was also revealed using nanodomain ligand/receptor activation of the receptor in adult cardiomyocytes. The NPR1 receptor (activated by A-type and B-type NP) was found in T-tubule membranes, and cGMP detected by a FRET sensor shows it to be normally constrained to the membrane near where ANP stimulation had been introduced. However, following PDE2 inhibition, ANP-stimulated cGMP diffused more generally into the cytosol32. This local regulation was further confirmed using a cGMP-FRET-sensor targeted to the caveolin-enriched nanodomains where NPR1 resided. One consequence of such compartmentation was that while ANP stimulation did not normally alter phosphorylation of the calcium cycling regulating protein phospholamban, it did if PDE2 was concomitantly inhibited32. Coupling of PDE2 with NP but not NO-dependent signaling has also been demonstrated in pulmonary vascular smooth muscle33.

Figure 2:

Figure 2:

Regulation of cardiac myocyte function by PDE2 and PDE3. PDE2 hydrolyzes cGMP and cAMP, and its activity is further stimulated by cGMP binding to regulatory domains. PDE2 inhibition can activate PKA-type II resulting in NFAT phosphorylation to prevent its nuclear translocation and hypertrophic signaling. It also stimulates PKA-type I that stimulates cardiac contraction and confers mitochondrial protection. PDE2 cGMP hydrolysis can provide antihypertrophic signaling effects. PDE3A can also hydrolyze cGMP and cAMP, the latter coupled to beta-adrenergic receptors. PKA- type I is activated phosphorylating L-type calcium channel (LTCC), phospholamban (PLN), and contractile proteins including troponin I, Tn; myosin binding protein C, MyBPC, and titin. This promotes contraction and relaxation. A deletion (del) in the PDE3A1 promoter prevents ATF3 (activating transcription factor 3) binding, disinhibiting effects of cAMP/CREB binding to enhance synthesis of PDE3A1. PDE3B hydrolyzes cAMP that has an impact on protection against ischemia/reperfusion injury linked PKA-I mitochondrial modifications. AC – adenylate cyclase, CREB – cyclic AMP responsive element binding protein.

The role of PDE2 in hydrolyzing cGMP generated by nitric-oxide stimulation of GC-1 has eluded detection by similar fluorescent sensor methods, most likely primarily due to insufficient sensitivity to the low amplitude cGMP signal. However, functional data support such interaction. For example, NO-stimulated cGMP, coupled to activation of the β3-adrenergic receptor, was shown to activate PDE2 (cGMP binding to regulatory GAF domains) in neonatal cardiomyocytes. This enhanced its hydrolysis of cAMP and thereby countered catecholamine stimulated contraction34. PDE2 inhibition also enhances cAMP-induced dilation induced by prostaglandin I2, supporting a role for cAMP hydrolysis in vascular tissue 33. Yet another study reported antihypertrophic effects from PDE2 inhibition in isolated neonatal cardiomyocytes exposed to angiotensin II, but this was observed when cGMP was co-stimulated using an NO-donor but not by a NPR1 agonist (ANP or BNP)35. Similarly, upregulation of PDE2 (genetic overexpression) in fibroblasts did not alter cGMP stimulated by NP agonism, but had a slight significant effect upon NO stimulation36. These latter studies are hard to reconcile with earlier data coupling PDE2 to NPR1 signaling, but they may reveal a critical role of experimental conditions and cAMP/cGMP balance.

PDE2 also hydrolyzes cAMP, as demonstrated by its control of β-adrenergic stimulation34 (Figure 2). In addition, PDE2 controls cAMP-dependent modulation of myocyte hypertrophy, fibrosis, and mitochondrial function. The primary effector of β1-adrenergic stimulation is cAMP-stimulated protein kinase A-I (PKA-I) that when sustained induces myocyte hypertrophy. By contrast, the PKA-II isoform confers anti-hypertrophic effects due to phosphorylation of nuclear factor of activated T-cells (NFAT) blocking its nuclear translocation and thereby myocyte hypertrophy37. PDE2 inhibition has been shown to enhance this cAMP-PKA-II pathway37. PDE2 overexpression in fibroblasts reduces cAMP levels with or without their co-stimulation, and results in an activated myofibroblast phenotype36. Interestingly, this transition is offset by co-stimulation of cGMP via NO or NP routes, indicating that PDE2 upregulation in this setting is preferentially via hydrolyzing cAMP. Lastly, PDE2A2 inhibition impacts mitochondrial elongation, increasing the transmembrane potential and thereby resistance to pro-apoptotic stimuli in MEFs and NRVMs. These changes are mediated by cAMP-PKA-dependent phosphorylation of dynamin-related protein 138.

The various features of PDE2 regulation identified in cellular systems have been examined in vivo in small rodents. PDE2 expression increases in diseased myocardium in various experimental animal models and in human heart failure35, 39. While there is general agreement that this plays a pathophysiological role in heart disease, there is controversy as to whether therapeutic benefit will derive from PDE2 inhibition or from enhancing its activity further40. The answer may ultimately depend on the nature of the cardiac disease. In models of pressure-load induced right or left ventricular hypertrophy, PDE2 inhibition reduces hypertrophy, fibrosis, and can improve cardiac function 33, 35, 37. Even here, the explanation varies, with some studies coupling this effect to enhancement of the cAMP-PKA-II pathway37, while others suggest this results from reduced cGMP hydrolysis and thus anti-hypertrophic signaling afforded by protein kinase G activation33, 35.

As in the cell studies, the source of cGMP regulated by PDE2 inhibition in vivo also varies among studies. In models of pulmonary hypertension and RV hypertrophy, gene deletion of the NPR1 eliminated efficacy of oral PDE2 inhibition to counter RV disease, whereas the latter remained effective despite NOS inhibition33. By contrast, in mice subjected to descending aortic constriction, LV hypertrophy was reduced by PDE2 inhibition independent of GC-A (GC coupled to the NPR1) expression, but was lost in mice lacking NO-stimulated GC-1α35. In contrast to pressure-stress hypertrophy models, in heart disease induced by catecholamine hyper-stimulation, or by myocardial infarction, PDE2 appears to play a protective role principally by suppressing adverse β-adrenergic stimulation41, 42. These studies propose upregulation of PDE2 as a protective component of β-AR desensitization limiting damage due to catecholamine hyper-stimulation, spawning efforts to further activate the PDE41. In mice with myocyte-targeted PDE2A overexpression, resting heart rate was reduced, catecholamine stimulated arrhythmia abetted, and cardiac function following myocardial infarction was improved over littermate controls42. The converse, e.g. reversal of downregulated β-AR signaling by inhibiting PDE2 has not been found in acute studies43, though it might occur with more chronic in vivo inhibition.

Viewed together, recent studies regarding the cardiac role of PDE2 and its modulation to treat heart disease reveal complexities regarding cyclic nucleotide substrate selectivity, the source of the substrate, and even whether blocking or activating the PDE is beneficial. This suggests the role of PDE2 in vivo depends very much on the ambient conditions, which may alter the balance of cGMP versus cAMP and the compartment being regulated. While these factors are more easily constrained in animal models, even these studies show considerable discrepancies. While intriguing, the basic work does raise concerns for translation into human diseases, where the underlying pathophysiology is generally complex, so predicting the response to PDE2 modulation may be difficult.

PDE3 – Isoform-specific signaling for therapeutic targeting

PDE3 is the other dual-substrate phosphodiesterase proposed to regulate cGMP in the heart. While its affinity for cAMP and cGMP is similar, it has a higher turnover rate for cAMP, providing its primary regulatory footprint in larger mammalian hearts including human. By competitive binding at the catalytic site, cAMP hydrolysis is inhibited by cGMP, yielding a mechanism for cGMP-dependent contractility augmentation at low levels of cGMP44. The PDE3 enzymes are transcribed from two genes, PDE3A and PDE3B. PDE3A exists as three isoforms that vary by alternative transcription and translation start sites, while PDE3B exists as only one isoform45, 46. All of the isoforms differ in their N-terminus where there are hydrophobic loops (PDE3A1 and PDE3B) to mediate lipid membrane insertion, and phosphorylation sites that promote protein-protein interactions. The latter is important for determining the intracellular localization of the isoforms. In particular, PDE3A has been found to complex with PI3Kγ and SERCA2A, and is mainly localized in the cardiomyocyte at the sarcoplasmic reticulum. By contrast, PDE3B localizes to myocyte T-tubule membranes (Figure 2).

Given its role in hydrolyzing cAMP, initial interest in PDE3 cardiac regulation focused on the potential for non-isoform selective inhibitors to enhance cardiac contractility and lusitropy while simultaneously producing venous and arterial vasodilation. These inodilator attributes were particularly attractive as therapy for heart failure with ventricular dilation and depressed systolic function. Acute hemodynamic studies in the 1980’s confirmed a favorable hemodynamic profile47, and the PDE3 inhibitor milrinone remains used in patients with acute decompensated heart failure. However, early optimism was countered by a major study of chronic PDE3 inhibition employing the identical compound that reported increased mortality, with concerns raised regarding lethal arrhythmia linked to excess calcium and energy demands48.

The concept was resurrected over a decade later in the context of a transformation in HF treatment that now routinely included use of a β-AR receptor antagonist such as metroprolol and carvedilol. The expectation was that in such patients, the potentially toxic effects of cAMP enhancement via PDE3 inhibition would be mitigated by concomitant β-blockade. A subsequent ~2000 patient multicenter international trial of enoximone, another PDE3 inhibitor, reported no adverse impact on survival or other clinical outcomes, however, there was also no significant benefit49. This lack of benefit may be due in part to a 29-nt INS/DEL polymorphism in the Pde3a promoter region50. The transcription factor ATF3 normally binds to the promoter insertion site to repress cAMP response element activity, such that PDE3A1 transcription is not upregulated in a feedback manner by cAMP. In patients with the polymorphism that deletes this binding site, the cAMP response element maintains activation enhancing PDE3A1 mRNA. Since inhibiting PDE3 increases cAMP, those with the DEL polymorphism would offset this by enhancing PDE3A expression. This may provide a genetic basis for why some HF patients develop tolerance to PDE3 inhibitors over time, though direct proof of this remains lacking (lower left, Figure 2).

While short-term use of oral PDE3 inhibition in severe HF patients is used as a bridge to transplantation 51, the therapy is now also being explored in patients with HF and a preserved ejection fraction. This is related to the capacity of PDE3 inhibition to increase cardiac output reserve at lower ventricular filling pressures during exercise52. A new extended release form of milrinone (CRD-102) has been developed for advanced heart failure with some early favorable clinical results53 similar to those reported with enoximone51. Whether this formulation will ultimately alter the safety-efficacy profile of milrinone leading to a renaissance as a HF therapeutic remains to be determined.

An alternative approach to small-molecule PDE3 inhibition is to impede cAMP hydrolysis by competing at the catalytic site with cGMP. This happens when low levels of cGMP compete with cAMP catalytic binding and thereby enhance contractility54, 55. In the presence of adrenergic stimulation or co-existent PDE3 inhibition, PDE3 primarily impacts cAMP, and similar cGMP changes now antagonize cAMP-stimulated contraction56 primarily by activating PDE2 to reduce cAMP (as discussed)57. This exemplifies crosstalk regulation between PDE2 and PDE3 to control the fate of cGMP/cAMP modulation of contraction. Recent vascular smooth muscle findings suggest NO-GC-1 signaling also enhances PDE3A expression, as genetic deletion of GC-1 in these cells reduces expression and activity of the PDE in aortas58. This may help maintain cAMP in the absence of homeostatic regulation of the phosphodiesterase by GC-1 generated cGMP. Whether this applies to the myocardium remains unknown. Such crosstalk regulation requires co-signaling in the appropriate nanodomain. This likely explains the lack of impact from NPR1 stimulation by ANP or BNP to inhibit or enhance β-AR coupled signaling59, whereas NPR2 stimulation by CNP enhances cAMP signaling via PDE3 inhibition (much like that via NO), and improve contractility in normal and failing hearts60.

Another third approach to improve the safety and efficacy from PDE3 inhibition is to develop isoform selective inhibitors. Polidovitch et al61 studied mice with either global gene deletion of PDE3A or PDE3B that were then subjected to pressure overload induced by trans-aortic constriction (TAC) and also given the PDE3 inhibitor, milrinone. WT-TAC mice treated with milrinone had less maladaptive ventricular remodeling, and PDE3A−/− mice demonstrated similar protection with no added benefit from milrinone. By contrast, PDE3B−/− mice responded similarly to TAC as WT, highlighting the selective role of PDE3A in this disease model. By contrast, PDE3B gene deletion is protective against myocardial ischemia/reperfusion injury62, with a mechanism linked to cAMP/PKA-induced mitochondrial protection44.

As the catalytic site is identical between PDE3 isoforms, existing small molecule inhibitors are equipotent against them63. However, differences in the intracellular localization and protein partnering provides the potential for an alternative approach based on disrupting local cAMP controlled by individual isoforms44. PDE3A is a component of a multiprotein complex in the sarcoplasmic reticulum along with AKAP18, phospholamban, and SERCA264 (Figure 2). When PKA is activated by cAMP, PKA phosphorylates phospholamban and promotes its dissociation from the complex, leading to an increase in SERCA2 activity. PDE3 inhibition is capable of potentiating that phosphorylation and stimulation. PDE3A1 itself is phosphorylated by PKA, and that phosphorylation promotes its interaction with the SERCA2 complex64. If this interaction is disrupted, then selective cAMP elevation near phospholamban and SERCA2 could potentiate inotropic effects perhaps while avoiding toxic effects from general PDE3 inhibition. This remains to be tested. Disruption of PDE3B from its complex with EPAC1 and p84-regulated PI3Kγ has been studied in vascular endothelial cells65. Here, introduction of a small peptide to displace EPAC1 from PDE3B augmented PI3Kγ signaling by increasing cAMP binding to EPAC1. Whether such small peptide disrupters can evolve into a viable cardiac therapeutic remains to be seen, but early proof-of-concept studies hold promise.

PDE5 – Myocardial regulation and compartmentation

PDE5A is a cGMP-specific PDE expressed as one of three isoforms PDE5A1, A2, and A3, all three present in human and mouse and varying in their N-terminus66. In vitro biochemical function is similar among the isoforms, though there are organ-specific differences in expression, and evidence that this may also influence subcellular localization66. PDE5A cGMP-catalytic activity is stimulated by cGMP binding to GAF regulatory domains and by phosphorylation by PKG at serine S102 in human (S92 in mouse)67. In cardiac myocytes, localization of PDE5A appears in a striated banding pattern that co-localizes with the z-disk protein α-actinin. This localization normally favors hydrolysis of cGMP generated by the nitric oxide-GC-1α pathway as compared to natriuretic peptide (GC-A or GC-B) pathway. However, studies in both adult canine and murine cardiomyocytes found this localization changes to a more diffuse cytosolic one in models of heart failure and hypertrophy, or if NOS3 is pharmacologically or genetically suppressed68, 69. Interestingly, its normal z-disk localization is restored even in hearts with chronic NOS inhibition by directly co-activating GC1α to generate cGMP70. The precise mechanism for altered intracellular PDE5A localization with heart disease remains unknown.

Inhibition of PDE5A is widely used to induce vascular smooth muscle relaxation, most notably in the corpus cavernosum and pulmonary vasculature. However, studies over the past two decades have documented substantial impact for countering cardiac structural and functional pathology in response to a broad range of myocardial disease (Figure 3). Indeed, studies mostly performed with the PDE5 inhibitor sildenafil have revealed its importance to myocardial cGMP-PKG activation and consequent modulation of pathological stress-mediating proteins12, 14, 69, 71-74. Among these proteins are regulators of G-protein signaling (RGS2 and RGS4) that counter Gq-receptor coupled stimulation by enhancing GTP removal from the activated Gα-subunit12. Another is transient receptor potential canonical channel type 6 (TRPC6), a non-selective non-voltage gated cation channel which principally conducts calcium and activates pro-fibrotic and pro-hypertrophic signaling coupled to calcineurin/NFAT. PKG phosphorylation inhibits the channel14. TRPC6 is also linked to abnormal mechano-stress responses in models of Duchenne muscular dystrophy, where it transduces PKG amelioration of this pathophysiology75. Sildenafil also blocks the accumulation of misfolded proteins, such as a mutated ab crystalin by enhancing their clearance by the ubiquitin-proteasome system19. PDE5A inhibitors reduce infarct size and cardiomyocyte apoptosis in ischemia/reperfusion models, effects that require targeting of mitochondrial function and structure76-78, ameliorate toxicity associated with doxorubicin79, 80, and reduce cardiometabolic disease in the Zucker diabetic fatty rat81. The precise mechanisms, e.g. which exact proteins and residues are targeted, have not been identified for these effects, but PKG regulation of metabolism and mitochondrial function is now being actively pursued.

Figure 3:

Figure 3:

Cardiac myocyte regulation by PDE5A and PDE9A. Both enzymes are highly selective for cGMP; PDE5A hydrolyzes principally GC-1α (NO-coupled) and PDE9A GC-A (NP coupled) derived cGMP. PDE5A inhibition leads to cGK1α that in turn phosphorylates sarcomeric proteins to enhance myocardial compliance and counter adrenergic stimulated contractility, proteasome activity, reduce hypertrophic-stimulated microRNA (miRNA) changes, RGS2/4 and TRPC6 to suppress Gq-protein coupled receptors and calcineurin (Cn) signaling, and tuberous sclerosis complex 2 (TSC2) to negatively control mechanistic target of rapamycin complex 1 (mTORC1) and its downstream effects on autophagy and hypertrophic growth stimulation. PDE9A shares many of these targets, notably those converging on hypertrophic stimuli, though there are differences (e.g. no impact on miRNAs, less contractility modulation). cGK1α also confers mitochondrial protection via KATP channels. Oxidation of cGK1α by reactive oxygen species (ROS) results in a loss of function of the pathway, by oxidative changes in NOS, GC-1α, and cGK1α. This results in depressed anti- hypertrophic signaling. Other abbreviations are as in other panels.

In 2019, PDE5A inhibition was shown for the first time to suppress the mechanistic target of rapamycin complex 1 by cGK1α-dependent phosphorylation of tuberous sclerosis complex protein 2 (TSC2)18. This occurred at one (or both) of two adjacent serines (S1364, S1365 in human) to activate TSC2 and in turn suppress mTORC1 by a Rheb-dependent pathway. This study is the first to date in which a specific amino acid residue target of cGK1α was mutated in vivo using a gene knock-in approach, and the impact on PDE5 inhibition tested. The result is striking in that homozygote KI mice with a TSC2 serine-alanine substitution (S1365 mouse, corresponds to S1364 in human) exposed to pressure-overload displayed marked hypertrophy and dysfunction that was unresponsive to PDE5 inhibition. Heterozygotes expressing the same mutation exhibited similarly worsened responses to pressure-overload, however, the availability of one normal allele for phosphorylation was sufficient to rescue the therapeutic efficacy of PDE5A inhibition by sildenafil. Similar results were obtained in isolated cardiac myocytes. In addition to expanding the regulatory control of pathological muscle growth and associated dysfunction by PDE5-regulated cGK1α signaling, this study also revealed its impact on macro-autophagy coupled to mTORC1 regulation. Sildenafil stimulated autophagy in a cGK1α-dependent manner and this was coupled to TSC2 phosphorylation at S1364.

While the majority of evidence supporting ameliorative effects from chronic PDE5A inhibition have been derived from small rodents – mostly mice, large animal model data also exist. For example, in tachypacing induced HF in sheep, tadalafil was found to improve contractile function, beta-adrenergic reserve, myocyte peak calcium transients, and restructure normal T-tubular architecture82.

Despite the multitude of studies revealing molecular, cellular, and intact organ impact from PDE5A regulation (both overexpression models and inhibition) in the myocardium, controversy remains regarding whether this PDE is in fact expressed in myocytes, and if so at levels sufficient to explain the observed changes. The major line of evidence used to claim a minimal role is based on lack of gel electrophoresis protein detection in mouse, canine, and human myocytes and myocardium83, 84. However, this may reflect the assay specifics, as others have reported expression in each of these species, and in particular in humans with cardiac hypertrophy85, and dilated failure including in single ventricle86, 87. The second line of evidence fueling controversy is that PDE5A inhibition is impactful in some models of heart disease but not others83, 88. It is possible that some studies utilized inadequate dosing, as mice have nearly 100x greater catabolism than humans, and much higher doses than those used clinically are required to achieve therapeutic yet selective free plasma concentrations72. However, model specifics are also likely key. PDE5A regulation of cGMP by the NO-stimulation pathway depends upon the extent the pathway is activated, and loss of this cGMP pool negates amelioration from blocking PDE5A69. This was shown in female mice exposed to TAC that lost the anti-hypertrophic effects of sildenafil if their ovaries were removed, but had it restored if the mice also received exogenous estrogen replacement89. PDE5A regulation of heart disease also requires that sufficient stress is generated so relevant PKG-targeted proteins are engaged. In the case of mild pressure-load induced myocardial disease, PDE5A inhibition has minimal impact, yet it yields ameliorative effects with more severe disease progression as PKG-modifiable pathological signaling is activated90. In the often cited neutral clinical human trial of sildenafil in patients with heart failure and a preserved ejection fraction (HFpEF), only a minority of subjects had ventricular hypertrophy, and overall blood pressures were only slightly above normal limits91. Unlike HF with a reduced EF, there is no clear data supporting PDE5A upregulation in this form of disease. Thus, lack of efficacy from PDE5A inhibition in HFpEF may be due to a lack of adequate upregulation in this syndrome. Lastly, oxidative stress common to various forms of heart disease may influence the therapeutic efficacy from PDE5A inhibition. This works in both directions. Oxidative changes in NO synthase and cGC1 both result in reduced cGMP generation92, and oxidation of cGK1α at C42 residues to form a disulfide in the N terminus between its homodimers depresses its amelioration of cardiac disease79, 93. This appears due to changes in intracellular localization of oxidized cGK1α versus a C42S form that cannot be oxidized and favors a plasma membrane distribution in adult cardiomyocytes. Together, these redox changes compromise the impact of PDE5A inhibition in the diseased heart. On an opposing side, mouse hearts and myocytes expressing C42S mutant cGK1α fail to show enhanced cGK1α activation and corresponding beneficial effects from PDE5A inhibition, whereas stimulating cGK1α directly is beneficial independent of its oxidation status94. So in this situation, oxidative stress plays a positive role to sildenafil efficacy. The mechanism again appears related to altered intracellular localization of PKG1a as oxidation is needed for optimal engagement with cGMP regulated by PDE5A.

Clinical translation of PDE5A inhibitors for cardiac disease remains a work in progress. A meta-analysis of reported controlled clinical trials (555 patients in 13 studies) found significant improvement on clinical outcome, exercise capacity, and pulmonary hemodynamics in HF patients with a reduced ejection fraction (HFrEF)95. A caveat is that these were mostly single center trials. HFpEF patients did not benefit but this was mostly based on the sole multicenter trial91.

PDE9A - Differential Effector of NO versus NP-generated cGMP

PDE9A is a cGMP-specific PDE encoded by a single gene that is then alternatively spliced to produce multiple isoforms96. It is expressed throughout the body, including lung, kidney, heart, and skeletal muscle, but most prominently in cerebellar Purkinje neurons and at lower levels in cortex, hippocampus, and striatum. Brain expression is thought to play a role in synaptic plasticity96, and studies to date have focused on this role and potential therapeutic use for disorders with cognitive disease such as Alzheimer’s and schizophrenia. There is isoform specific expression in different tissues, with brain expressing PDE9A6/13 and three higher molecular weight isoforms – PDE9x-100, −120, and −175. These are not found in the heart, which instead expresses PDE9A2, and 9A996. Isoform-specific functional differences have yet to be identified, and inhibitors of the enzyme do not differentiate between them.

The role of PDE9A in the heart was first reported in 201569. Expression at both mRNA and protein level was demonstrated in human tissue, where it was also found to be increased in failing and hypertrophied hearts. It is distributed differently from PDE5A in the cardiac myocyte, exhibiting a longitudinal staining pattern that colocalizes with SERCA2A but not with α-actinin (the opposite holding for PDE5A). Rather than modifying cGMP synthesized by NO-stimulated GC-1, PDE9A regulates cGMP coupled to natriuretic peptide signaling in myocytes (Figure 3). Mice lacking Pde9a are protected against sustained pressure-overload, exhibiting reduced hypertrophy, fibrosis, and enhanced cardiac function, and similar protection was observed in WT mice treated with a selective oral PDE9A inhibitor (PF-00047943). The latter occurs whether NOS is concomitantly inhibited by L-NAME or not, revealing independence from NO-cGMP dependent signaling. This contrasts to effects from sildenafil, a PDE5A inhibitor, that is only effective in countering pressure-overload if NOS signaling remains intact.

Comparisons between cGMP and consequent cGK-1 regulation by inhibition of either PDE5A or PDE9A in the heart have yielded intriguing similarities and differences. Both have a strong impact on suppressing hypertrophic and fibrosis-related gene expression, notably signaling coupled to TRPC6 and downstream calcineurin activation69. However, the phosphokinome modified by either inhibition strategy also displays many unique targets, consistent with differences in cellular localization and substrate preference between the two PDEs. Among the most striking disparities is how PDE5A or PDE9A inhibitor treatment impacts microRNA expression altered by pressure-overload97. The loading stress results in the upregulation of many pro-hypertrophic/pro-fibrotic miRNAs (e.g. 34c, 21a, 199a, 208b) and anti-hypertrophic miRNAs (e.g. MiR 1a, 30b, 133a). Of 111 miRs significantly impacted by pressure-overload, PDE5A inhibition reduced most of them. By contrast, PDE9A inhibition had negligible impact on the miRNA profile. This was true despite both treatments conferring near identical phenotypic improvement in cardiac function, histology, and morphology. RNAseq analysis revealed ~70-90% of the genes with altered expression from each respective PDE inhibitor were different, though they converged on similar signaling pathways. Disparities in miR expression occurred in the cytosol, in that neither Pri-miRNA or Pre-miRNA showed differences between PDE-inhibitor treatments.

In addition to a myocardial role, recent studies suggest PDE9A may contribute to vascular smooth muscle (VSM) cGMP regulation98. Similar to brain, VSM cells were found to express PDE9A6/A13 but at a much lower level, as well as what may be PDE9x-100. PDE9A inhibition augmented cGMP coupled to NO stimulation but not ANP or CNP stimulation in rat arterial smooth muscle cells98, opposite to the substrate selectivity found in cardiomyocytes and the intact heart69. The reasons for this different are unknown, but might relate to expression of different splice variants and thus intracellular localization which has not been examined in VSM to date. The vascular study did not confirm changes in cells lacking the PDE9A gene. It is worth noting that PDE9A inhibition in vivo (both mice and humans) has not been found to impact systemic blood pressure. Ongoing studies with a PDE9A flox’d mouse will be helpful to sort out tissue specific roles.

Cardiovascular and renal effects of acute PDE9A inhibition were recently reported in sheep with normal or failing hearts99. A different PDE9A inhibitor compound (PF- 04749982) was administered intravenously, and this dose dependently increased plasma cGMP/natriuretic peptide ratio, urinary cGMP, and in heart failure conditions, urine volume, sodium excretion, and creatinine clearance. An observed increase in cardiac output and arterial vasodilation in animals with failing hearts administered the highest dose may reflect PDE9A-mediated changes, though off target effects on PDE1 are also possible. No prior studies in humans have reported blood pressure changes. These data in a large mammal support interactions between PDE9A and NP-derived cGMP, and suggest HF benefits may derive from cardiac and renal improvement.

Clinical exploration of the cardiovascular role of PDE9A is in early stages. Several small molecule inhibitors have been generated, and at least three have been studied in humans for treatment of Alzheimer’s disease100, schizophrenia, or prostatic hypertrophy. None of these indications proved sufficiently encouraging to continue development. However, Cardurion Pharmaceuticals initiated safety and dose-finding human studies in heart failure patients in late 2018, using the PDE9A inhibitor CRD-733, and these trials are ongoing. Another potential avenue is to synergize the regulation of NP-dependent cGMP levels resulting from neutral endopeptidase (neprilysin) inhibition with suppression of hydrolysis of the synthesized cGMP by a PDE9A inhibitor. Preclinical studies are underway to assess this interaction.

Summary

In just the past few years, studies have revealed striking new roles of various PDEs on cardiovascular physiology and disease. With PDE1, the disparity in isoform expression between rodent and larger mammal may explain the cAMP-related changes observed in the latter but not former species. After near abandonment for several decades, new therapeutic opportunities for PDE1 inhibition to treat myocardial disease and heart failure have led to clinical studies. PDE3A inhibition by non-conventional means, including allosteric protein disruptors or altered pharmacokinetics may reignite interest. The role of PDE5A and PDE9A inhibition continues to be elucidated, but their impact on molecular signaling in the heart, and differential control over cGMP synthesized by NO versus NP pathways, support ongoing efforts to test their therapeutic utility for heart disease.

Supplementary Material

Supplemental Materials - Tables

REFERENCES:

  • 1.Maurice DH, Ke H, Ahmad F, Wang Y, Chung J and Manganiello VC. Advances in targeting cyclic nucleotide phosphodiesterases. Nat Rev Drug Discov. 2014;13:290–314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Bender AT and Beavo JA. Cyclic nucleotide phosphodiesterases: molecular regulation to clinical use. Pharmacol Rev. 2006;58:488–520. [DOI] [PubMed] [Google Scholar]
  • 3.Francis SH, Blount MA and Corbin JD. Mammalian cyclic nucleotide phosphodiesterases: Molecular mechanisms and physiological functions. Physiol Rev. 2011;91:651–690. [DOI] [PubMed] [Google Scholar]
  • 4.Kentish JC, McCloskey DT, Layland J, Palmer S, Leiden JM, Martin AF and Solaro RJ. Phosphorylation of troponin I by protein kinase A accelerates relaxation and crossbridge cycle kinetics in mouse ventricular muscle. Circ Res. 2001;88:1059–1065. [DOI] [PubMed] [Google Scholar]
  • 5.Yamasaki R, Wu Y, McNabb M, Greaser M, Labeit S and Granzier H. Protein kinase A phosphorylates titin's cardiac-specific N2B domain and reduces passive tension in rat cardiac myocytes. Circ Res. 2002;90:1181–1188. [DOI] [PubMed] [Google Scholar]
  • 6.Stelzer JE, Patel JR and Moss RL. Protein kinase A-mediated acceleration of the stretch activation response in murine skinned myocardium is eliminated by ablation of cMyBP-C. Circ Res. 2006;99:884–890. [DOI] [PubMed] [Google Scholar]
  • 7.Nagayama T, Takimoto E, Sadayappan S, Mudd JO, Seidman JG, Robbins J and Kass DA. Control of in vivo left ventricular [correction] contraction/relaxation kinetics by myosin binding protein C: protein kinase A phosphorylation dependent and independent regulation. Circulation. 2007;116:2399–2408. [DOI] [PubMed] [Google Scholar]
  • 8.MacLennan DH and Kranias EG. Phospholamban: a crucial regulator of cardiac contractility. Nat Rev Mol Cell Biol. 2003;4:566–577. [DOI] [PubMed] [Google Scholar]
  • 9.Reiken SR, Gaburjakova M, Guatimosim S, Gomez AM, D'Armiento J, Burkhoff D, Wang J, Vassort G, Lederer J and Marks AR. PKA phosphorylation of the cardiac calcium release channel (ryanodine receptor) in normal and failing hearts: role of phosphatases and response to isoproterenol. J Biol Chem. 2002. [DOI] [PubMed] [Google Scholar]
  • 10.Verde I, Vandecasteele G, Lezoualc'h F and Fischmeister R. Characterization of the cyclic nucleotide phosphodiesterase subtypes involved in the regulation of the L-type Ca2+ current in rat ventricular myocytes. Br J Pharmacol. 1999;127:65–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Gloerich M and Bos JL. Epac: defining a new mechanism for cAMP action. Annu Rev Pharmacol Toxicol. 2010;50:355–75. [DOI] [PubMed] [Google Scholar]
  • 12.Takimoto E, Koitabashi N, Hsu S, Ketner EA, Zhang M, Nagayama T, Bedja D, Gabrielson KL, Blanton R, Siderovski DP, Mendelsohn ME and 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;119:408–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Tokudome T, Kishimoto I, Horio T, Arai Y, Schwenke DO, Hino J, Okano I, Kawano Y, Kohno M, Miyazato M, Nakao K and Kangawa K. Regulator of G-protein signaling subtype 4 mediates antihypertrophic effect of locally secreted natriuretic peptides in the heart. Circulation. 2008;117:2329–39. [DOI] [PubMed] [Google Scholar]
  • 14.Koitabashi N, Aiba T, Hesketh GG, Rowell J, Zhang M, Takimoto E, Tomaselli GF and 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;48:713–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Nishida M, Watanabe K, Sato Y, Nakaya M, Kitajima N, Ide T, Inoue R and Kurose H. Phosphorylation of TRPC6 channels at Thr69 is required for anti-hypertrophic effects of phosphodiesterase 5 inhibition. J Biol Chem. 2010;285:13244–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 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 and 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;106:1849–60. [DOI] [PubMed] [Google Scholar]
  • 17.Sawada N, Itoh H, Yamashita J, Doi K, Inoue M, Masatsugu K, Fukunaga Y, Sakaguchi S, Sone M, Yamahara K, Yurugi T and Nakao K. cGMP-dependent protein kinase phosphorylates and inactivates RhoA. Biochem Biophys Res Commun. 2001;280:798–805. [DOI] [PubMed] [Google Scholar]
  • 18.Ranek MJ, Kokkonen-Simon KM, Chen A, Dunkerly-Eyring BL, Vera MP, Oeing CU, Patel CH, Nakamura T, Zhu G, Bedja D, Sasaki M, Holewinski RJ, Van Eyk JE, Powell JD, Lee DI and Kass DA. PKG1-modified TSC2 regulates mTORC1 activity to counter adverse cardiac stress. Nature. 2019;566:264–269. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Ranek MJ, Terpstra EJ, Li J, Kass DA and Wang X. Protein kinase g positively regulates proteasome-mediated degradation of misfolded proteins. Circulation. 2013;128:365–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Sonnenburg WK, Seger D, Kwak KS, Huang J, Charbonneau H and Beavo JA. Identification of inhibitory and calmodulin-binding domains of the PDE1A1 and PDE1A2 calmodulin-stimulated cyclic nucleotide phosphodiesterases. J Biol Chem. 1995;270:30989–31000. [DOI] [PubMed] [Google Scholar]
  • 21.Nagel DJ, Aizawa T, Jeon KI, Liu W, Mohan A, Wei H, Miano JM, Florio VA, Gao P, Korshunov VA, Berk BC and Yan C. Role of nuclear Ca2+/calmodulin-stimulated phosphodiesterase 1A in vascular smooth muscle cell growth and survival. Circ Res. 2006;98:777–784. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Yan C, Zhao AZ, Bentley JK and Beavo JA. The calmodulin-dependent phosphodiesterase gene PDE1C encodes several functionally different splice variants in a tissue-specific manner. J Biol Chem. 1996;271:25699–706. [DOI] [PubMed] [Google Scholar]
  • 23.Hashimoto T, Kim GE, Tunin RS, Adesiyun T, Hsu S, Nakagawa R, Zhu G, O'Brien JJ, Hendrick JP, Davis RE, Yao W, Beard D, Hoxie HR, Wennogle LP, Lee DI and Kass DA. Acute Enhancement of Cardiac Function by Phosphodiesterase Type 1 Inhibition. Circulation. 2018;138:1974–1987. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Knight WE, Chen S, Zhang Y, Oikawa M, Wu M, Zhou Q, Miller CL, Cai Y, Mickelsen DM, Moravec C, Small EM, Abe J and Yan C. PDE1C deficiency antagonizes pathological cardiac remodeling and dysfunction. Proc Natl Acad Sci U S A. 2016;113:E7116–E7125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Wu MP, Zhang YS, Xu X, Zhou Q, Li JD and Yan C. Vinpocetine Attenuates Pathological Cardiac Remodeling by Inhibiting Cardiac Hypertrophy and Fibrosis. Cardiovasc Drugs Ther. 2017;31:157–166. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.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 and Yan C. Role of Ca2+/calmodulin-stimulated cyclic nucleotide phosphodiesterase 1 in mediating cardiomyocyte hypertrophy. Circ Res. 2009;105:956–964. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Miller CL, Cai Y, Oikawa M, Thomas T, Dostmann WR, Zaccolo M, Fujiwara K and Yan C. Cyclic nucleotide phosphodiesterase 1A: a key regulator of cardiac fibroblast activation and extracellular matrix remodeling in the heart. Basic Res Cardiol. 2011;106:1023–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Leroy J and Fischmeister R. Inhibit a Phosphodiesterase to Treat Heart Failure? Circulation. 2018;138:2003–2006. [DOI] [PubMed] [Google Scholar]
  • 29.Zhang Y, Knight W, Chen S, Mohan A and Yan C. Multiprotein Complex With TRPC (Transient Receptor Potential-Canonical) Channel, PDE1C (Phosphodiesterase 1C), and A2R (Adenosine A2 Receptor) Plays a Critical Role in Regulating Cardiomyocyte cAMP and Survival. Circulation. 2018;138:1988–2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Xi J, McIntosh R, Shen X, Lee S, Chanoit G, Criswell H, Zvara DA and Xu Z. Adenosine A2A and A2B receptors work in concert to induce a strong protection against reperfusion injury in rat hearts. J Mol Cell Cardiol. 2009;47:684–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Castro LR, Verde I, Cooper DM and Fischmeister R. Cyclic guanosine monophosphate compartmentation in rat cardiac myocytes. Circulation. 2006;113:2221–2228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Subramanian H, Froese A, Jönsson P, Schmidt H, Gorelik J and Nikolaev VO. Distinct submembrane localisation compartmentalises cardiac NPR1 and NPR2 signalling to cGMP. Nat Commun. 2018;9:2446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Bubb KJ, Trinder SL, Baliga RS, Patel J, Clapp LH, MacAllister RJ and Hobbs AJ. Inhibition of phosphodiesterase 2 augments cGMP and cAMP signaling to ameliorate pulmonary hypertension. Circulation. 2014;130:496–507. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Mongillo M, Tocchetti CG, Terrin A, Lissandron V, Cheung YF, Dostmann WR, Pozzan T, Kass DA, Paolocci N, Houslay MD and Zaccolo M. Compartmentalized phosphodiesterase-2 activity blunts beta-adrenergic cardiac inotropy via an NO/cGMP-dependent pathway. Circ Res. 2006;98:226–234. [DOI] [PubMed] [Google Scholar]
  • 35.Baliga RS, Preedy MEJ, Dukinfield MS, Chu SM, Aubdool AA, Bubb KJ, Moyes AJ, Tones MA and Hobbs AJ. Phosphodiesterase 2 inhibition preferentially promotes NO/guanylyl cyclase/cGMP signaling to reverse the development of heart failure. Proc Natl Acad Sci U S A. 2018;115:E7428–e7437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Vettel C, Lammle S, Ewens S, Cervirgen C, Emons J, Ongherth A, Dewenter M, Lindner D, Westermann D, Nikolaev VO, Lutz S, Zimmermann WH and El-Armouche A. PDE2-mediated cAMP hydrolysis accelerates cardiac fibroblast to myofibroblast conversion and is antagonized by exogenous activation of cGMP signaling pathways. Am J Physiol Heart Circ Physiol. 2014;306:H1246–52. [DOI] [PubMed] [Google Scholar]
  • 37.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 and Zaccolo M. Cardiac Hypertrophy Is Inhibited by a Local Pool of cAMP Regulated by Phosphodiesterase 2. Circ Res. 2015;117:707–19. [DOI] [PubMed] [Google Scholar]
  • 38.Monterisi S, Lobo MJ, Livie C, Castle JC, Weinberger M, Baillie G, Surdo NC, Musheshe N, Stangherlin A, Gottlieb E, Maizels R, Bortolozzi M, Micaroni M and Zaccolo M. PDE2A2 regulates mitochondria morphology and apoptotic cell death via local modulation of cAMP/PKA signalling. Elife. 2017;6:e21374 DOI: 10.7554/eLife.21374. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.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 and El-Armouche A. Phosphodiesterase-2 is up-regulated in human failing hearts and blunts beta-adrenergic responses in cardiomyocytes. J Am Coll Cardiol. 2013;62:1596–606. [DOI] [PubMed] [Google Scholar]
  • 40.Wagner M, Mehel H, Fischmeister R and El-Armouche A. Phosphodiesterase 2: anti-adrenergic friend or hypertrophic foe in heart disease? Naunyn Schmiedebergs Arch Pharmacol. 2016;389:1139–1141. [DOI] [PubMed] [Google Scholar]
  • 41.Mehel H, Emons J, Vettel C, Wittköpper K, Seppelt D, Dewenter M, Lutz S, Sossalla S, Maier LS, Lechêne P, Leroy J, Lefebvre F, Varin A, Eschenhagen T, Nattel S, Dobrev D, Zimmermann WH, Nikolaev VO, Vandecasteele G, Fischmeister R and El-Armouche A. Phosphodiesterase-2 is up-regulated in human failing hearts and blunts β-adrenergic responses in cardiomyocytes. J Am Coll Cardiol. 2013;62:1596–606. [DOI] [PubMed] [Google Scholar]
  • 42.Vettel C, Lindner M, Dewenter M, Lorenz K, Schanbacher C, Riedel M, Lämmle S, Meinecke S, Mason FE, Sossalla S, Geerts A, Hoffmann M, Wunder F, Brunner FJ, Wieland T, Mehel H, Karam S, Lechêne P, Leroy J, Vandecasteele G, Wagner M, Fischmeister R and El-Armouche A. Phosphodiesterase 2 Protects Against Catecholamine-Induced Arrhythmia and Preserves Contractile Function After Myocardial Infarction. Circ Res. 2017;120:120–132. [DOI] [PubMed] [Google Scholar]
  • 43.Galindo-Tovar A, Vargas ML and Kaumann AJ. Phosphodiesterase PDE2 activity, increased by isoprenaline, does not reduce β-adrenoceptor-mediated chronotropic and inotropic effects in rat heart. Naunyn Schmiedebergs Arch Pharmacol. 2018;391:571–585. [DOI] [PubMed] [Google Scholar]
  • 44.Movsesian M, Ahmad F and Hirsch E. Functions of PDE3 Isoforms in Cardiac Muscle. J Cardiovasc Dev Dis. 2018;5 pii: E10. doi: 10.3390/jcdd5010010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Miki T, Taira M, Hockman S, Shimada F, Lieman J, Napolitano M, Ward D, Makino H and Manganiello VC. Characterization of the cDNA and gene encoding human PDE3B, the cGIP1 isoform of the human cyclic GMP-inhibited cyclic nucleotide phosphodiesterase family. Genomics. 1996;36:476–85. [DOI] [PubMed] [Google Scholar]
  • 46.Wechsler J, Choi YH, Krall J, Ahmad F, Manganiello VC and Movsesian MA. Isoforms of cyclic nucleotide phosphodiesterase PDE3A in cardiac myocytes. J Biol Chem. 2002;277:38072–8. [DOI] [PubMed] [Google Scholar]
  • 47.Jaski BE, Fifer MA, Wright RF, Braunwald E and Colucci WS. Positive inotropic and vasodilator actions of milrinone in patients with severe congestive heart failure. Dose-response relationships and comparison to nitroprusside. J Clin Invest. 1985;75:643–649. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Amsallem E, Kasparian C, Haddour G, Boissel JP and Nony P. Phosphodiesterase III inhibitors for heart failure. Cochrane Database Syst Rev. 2005:CD002230 DOI: 10.1002/14651858.CD002230/pub2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Metra M, Eichhorn E, Abraham WT, Linseman J, Bohm M, Corbalan R, DeMets D, De Marco T, Elkayam U, Gerber M, Komajda M, Liu P, Mareev V, Perrone SV, Poole-Wilson P, Roecker E, Stewart J, Swedberg K, Tendera M, Wiens B and Bristow MR. Effects of low-dose oral enoximone administration on mortality, morbidity, and exercise capacity in patients with advanced heart failure: the randomized, double-blind, placebo-controlled, parallel group ESSENTIAL trials. Eur Heart J. 2009;30:3015–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Sucharov CC, Nakano SJ, Slavov D, Schwisow JA, Rodriguez E, Nunley K, Medway A, Stafford N, Nelson P, McKinsey TA, Movsesian M, Minobe W, Carroll IA, Taylor MRG and Bristow MR. A PDE3A Promoter Polymorphism Regulates cAMP-Induced Transcriptional Activity in Failing Human Myocardium. J Am Coll Cardiol. 2019;73:1173–1184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Shakar SF, Abraham WT, Gilbert EM, Robertson AD, Lowes BD, Zisman LS, Ferguson DA and Bristow MR. Combined oral positive inotropic and beta-blocker therapy for treatment of refractory class IV heart failure. J Am Coll Cardiol. 1998;31:1336–40. [DOI] [PubMed] [Google Scholar]
  • 52.Kaye DM, Nanayakkara S, Vizi D, Byrne M and Mariani JA. Effects of Milrinone on Rest and Exercise Hemodynamics in Heart Failure With Preserved Ejection Fraction. J Am Coll Cardiol. 2016;67:2554–6. [DOI] [PubMed] [Google Scholar]
  • 53.Nanayakkara S, Mak V, Crannitch K, Byrne M and Kaye DM. Extended Release Oral Milrinone, CRD-102, for Advanced Heart Failure. Am J Cardiol. 2018;122:1017–1020. [DOI] [PubMed] [Google Scholar]
  • 54.Vila-Petroff MG, Younes A, Egan J, Lakatta EG and Sollott SJ. Activation of distinct cAMP-dependent and cGMP-dependent pathways by nitric oxide in cardiac myocytes. Circ Res. 1999;84:1020–1031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Rozmaritsa N, Christ T, Van Wagoner DR, Haase H, Stasch JP, Matschke K and Ravens U. Attenuated response of L-type calcium current to nitric oxide in atrial fibrillation. Cardiovasc Res. 2014;101:533–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Mongillo M, Tocchetti CG, Terrin A, Lissandron V, Cheung YF, Dostmann WR, Pozzan T, Kass DA, Paolocci N, Houslay MD and Zaccolo M. Compartmentalized phosphodiesterase-2 activity blunts beta-adrenergic cardiac inotropy via an NO/cGMP-dependent pathway. Circ Res. 2006;98:226–34. [DOI] [PubMed] [Google Scholar]
  • 57.Stangherlin A, Gesellchen F, Zoccarato A, Terrin A, Fields LA, Berrera M, Surdo NC, Craig MA, Smith G, Hamilton G and Zaccolo M. cGMP signals modulate cAMP levels in a compartment-specific manner to regulate catecholamine-dependent signaling in cardiac myocytes. Circ Res. 2011;108:929–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Dunnes S, Voussen B, Aue A, Groneberg K, Nikolaev V, Groneberg D and Friebe A. Phosphodiesterase 3A expression and activity in the murine vasculature is influenced by NO-sensitive guanylyl cyclase. Pflugers Arch. 2018;470:693–702. [DOI] [PubMed] [Google Scholar]
  • 59.Takimoto E, Belardi D, Tocchetti CG, Vahebi S, Cormaci G, Ketner EA, Moens AL, Champion HC and Kass DA. Compartmentalization of cardiac beta-adrenergic inotropy modulation by phosphodiesterase type 5. Circulation. 2007;115:2159–67. [DOI] [PubMed] [Google Scholar]
  • 60.Meier S, Andressen KW, Aronsen JM, Sjaastad I, Hougen K, Skomedal T, Osnes JB, Qvigstad E, Levy FO and Moltzau LR. PDE3 inhibition by C-type natriuretic peptide-induced cGMP enhances cAMP-mediated signaling in both non-failing and failing hearts. Eur J Pharmacol. 2017;812:174–183. [DOI] [PubMed] [Google Scholar]
  • 61.Polidovitch N, Yang S, Sun H, Lakin R, Ahmad F, Gao X, Turnbull PC, Chiarello C, Perry CGR, Manganiello V, Yang P and Backx PH. Phosphodiesterase type 3A (PDE3A), but not type 3B (PDE3B), contributes to the adverse cardiac remodeling induced by pressure overload. J Mol Cell Cardiol. 2019;132:60–70. [DOI] [PubMed] [Google Scholar]
  • 62.Chung YW, Lagranha C, Chen Y, Sun J, Tong G, Hockman SC, Ahmad F, Esfahani SG, Bae DH, Polidovitch N, Wu J, Rhee DK, Lee BS, Gucek M, Daniels MP, Brantner CA, Backx PH, Murphy E and Manganiello VC. Targeted disruption of PDE3B, but not PDE3A, protects murine heart from ischemia/reperfusion injury. Proc Natl Acad Sci U S A. 2015;112:E2253–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Hambleton R, Krall J, Tikishvili E, Honeggar M, Ahmad F, Manganiello VC and Movsesian MA. Isoforms of cyclic nucleotide phosphodiesterase PDE3 and their contribution to cAMP hydrolytic activity in subcellular fractions of human myocardium. J Biol Chem. 2005;280:39168–74. [DOI] [PubMed] [Google Scholar]
  • 64.Ahmad F, Shen W, Vandeput F, Szabo-Fresnais N, Krall J, Degerman E, Goetz F, Klussmann E, Movsesian M and Manganiello V. Regulation of sarcoplasmic reticulum Ca2+ ATPase 2 (SERCA2) activity by phosphodiesterase 3A (PDE3A) in human myocardium: phosphorylation-dependent interaction of PDE3A1 with SERCA2. J Biol Chem. 2015;290:6763–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Wilson LS, Baillie GS, Pritchard LM, Umana B, Terrin A, Zaccolo M, Houslay MD and Maurice DH. A phosphodiesterase 3B-based signaling complex integrates exchange protein activated by cAMP 1 and phosphatidylinositol 3-kinase signals in human arterial endothelial cells. J Biol Chem. 2011;286:16285–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Campolo F, Zevini A, Cardarelli S, Monaco L, Barbagallo F, Pellegrini M, Cornacchione M, Di Grazia A, De Arcangelis V, Gianfrilli D, Giorgi M, Lenzi A, Isidori AM and Naro F. Identification of murine phosphodiesterase 5A isoforms and their functional characterization in HL-1 cardiac cell line. J Cell Physiol. 2018;233:325–337. [DOI] [PubMed] [Google Scholar]
  • 67.Thomas MK, Francis SH and Corbin JD. Substrate- and kinase-directed regulation of phosphorylation of a cGMP-binding phosphodiesterase by cGMP. J Biol Chem. 1990;265:14971–14978. [PubMed] [Google Scholar]
  • 68.Takimoto E, Champion HC, Belardi D, Moslehi J, Mongillo M, Mergia E, Montrose DC, Isoda T, Aufiero K, Zaccolo M, Dostmann WR, Smith CJ and Kass DA. cGMP catabolism by phosphodiesterase 5A regulates cardiac adrenergic stimulation by NOS3-dependent mechanism. Circ Res. 2005;96:100–109. [DOI] [PubMed] [Google Scholar]
  • 69.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 and Kass DA. Phosphodiesterase 9A controls nitric-oxide-independent cGMP and hypertrophic heart disease. Nature. 2015;519:472–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Nagayama T, Zhang M, Hsu S, Takimoto E and Kass DA. Sustained soluble guanylate cyclase stimulation offsets nitric-oxide synthase inhibition to restore acute cardiac modulation by sildenafil. J Pharmacol Exp Ther. 2008;326:380–387. [DOI] [PubMed] [Google Scholar]
  • 71.Takimoto E, Koitabashi N, Hsu S, Ketner EA, Zhang M, Nagayama T, Bedja D, Gabrielson KL, Blanton R, Siderovski DP, Mendelsohn ME and Kass DA. RGS2 mediates cardiac compensation to pressure-overload and anti-hypertrophic effects of PDE5 inhibition. J Clin Invest. 2009;119:408–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Hutchings DC, Anderson SG, Caldwell JL and Trafford AW. Phosphodiesterase-5 inhibitors and the heart: compound cardioprotection? Heart. 2018;104:1244–1250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Korkmaz-Icoz S, Radovits T and Szabo G. Targeting phosphodiesterase 5 as a therapeutic option against myocardial ischaemia/reperfusion injury and for treating heart failure. Br J Pharmacol. 2018;175:223–231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Takimoto E, Champion HC, Li M, Belardi D, Ren S, Rodriguez ER, Bedja D, Gabrielson KL, Wang Y and Kass DA. Chronic inhibition of cyclic GMP phosphodiesterase 5A prevents and reverses cardiac hypertrophy. Nat Med. 2005;11:214–222. [DOI] [PubMed] [Google Scholar]
  • 75.Seo K, Rainer PP, Lee DI, Hao S, Bedja D, Birnbaumer L, Cingolani OH and 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;114:823–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Salloum FN, Abbate A, Das A, Houser JE, Mudrick CA, Qureshi I, Hoke NN, Roy SK, Brown WR, Prabhakar S and Kukreja RC. Sildenafil (Viagra) Attenuates IschemicCardiomyopathy and Improves Left VentricularFunction in Mice. Am J Physiol Heart Circ Physiol. 2008;294:H1398–H1406. [DOI] [PubMed] [Google Scholar]
  • 77.Li N, Yuan Y, Li S, Zeng C, Yu W, Shen M, Zhang R, Li C, Zhang Y and Wang H. PDE5 inhibitors protect against post-infarction heart failure. Front Biosci. 2016;21:1194–210. [DOI] [PubMed] [Google Scholar]
  • 78.Ockaili R, Salloum F, Hawkins J and 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. [DOI] [PubMed] [Google Scholar]
  • 79.Prysyazhna O, Burgoyne JR, Scotcher J, Grover S, Kass D and Eaton P. Phosphodiesterase 5 Inhibition Limits Doxorubicin-induced Heart Failure by Attenuating Protein Kinase G Ialpha Oxidation. J Biol Chem. 2016;291:17427–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Koka S, Das A, Zhu SG, Durrant D, Xi L and Kukreja RC. Long-acting phosphodiesterase-5 inhibitor tadalafil attenuates doxorubicin-induced cardiomyopathy without interfering with chemotherapeutic effect. J Pharmacol Exp Ther. 2010;334:1023–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Matyas C, Nemeth BT, Olah A, Torok M, Ruppert M, Kellermayer D, Barta BA, Szabo G, Kokeny G, Horvath EM, Bodi B, Papp Z, Merkely B and Radovits T. Prevention of the development of heart failure with preserved ejection fraction by the phosphodiesterase-5A inhibitor vardenafil in rats with type 2 diabetes. Eur J Heart Fail. 2017;19:326–336. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Lawless M, Caldwell JL, Radcliffe EJ, Smith CER, Madders GWP, Hutchings DC, Woods LS, Church SJ, Unwin RD, Kirkwood GJ, Becker LK, Pearman CM, Taylor RF, Eisner DA, Dibb KM and Trafford AW. Phosphodiesterase 5 inhibition improves contractile function and restores transverse tubule loss and catecholamine responsiveness in heart failure. Sci Rep. 2019;9:6801. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Patrucco E, Domes K, Sbroggio M, Blaich A, Schlossmann J, Desch M, Rybalkin SD, Beavo JA, Lukowski R and 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 U S A. 2014;111:12925–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Li EA, Xi W, Han YS and Brozovich FV. Phosphodiesterase expression in the normal and failing heart. Arch Biochem Biophys. 2019;662:160–168. [DOI] [PubMed] [Google Scholar]
  • 85.Nagendran J, Archer SL, Soliman D, Gurtu V, Moudgil R, Haromy A, St.Aubin C, Webster L, Rebeyka IM, Ross DB, Light PE and Michelakis ED. Phosphodiesteras type 5 (PDE5) is highly expressed in the hypertrophied human right ventricle and acute inhibition of PDE5 improves contractility. Circulation. 2007;116:238–248. [DOI] [PubMed] [Google Scholar]
  • 86.Shan X, Quaile MP, Monk JK, French B, Cappola TP and Margulies KB. Differential expression of PDE5 in failing and nonfailing human myocardium. Circ Heart Fail. 2012;5:79–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Garcia AM, Nakano SJ, Karimpour-Fard A, Nunley K, Blain-Nelson P, Stafford NM, Stauffer BL, Sucharov CC and Miyamoto SD. Phosphodiesterase-5 Is Elevated in Failing Single Ventricle Myocardium and Affects Cardiomyocyte Remodeling In Vitro. Circ Heart Fail. 2018;11:e004571. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Lukowski R, Rybalkin SD, Loga F, Leiss V, Beavo JA and Hofmann F. Cardiac hypertrophy is not amplified by deletion of cGMP-dependent protein kinase I in cardiomyocytes. Proc Natl Acad Sci U S A. 2010;107:5646–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.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 and Takimoto E. PDE5 inhibitor efficacy is estrogen dependent in female heart disease. J Clin Invest. 2014;124:2464–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Nagayama T, Hsu S, Zhang M, Koitabashi N, Bedja D, Gabrielson KL, Takimoto E and Kass DA. Pressure-overload magnitude-dependence of the anti-hypertrophic efficacy of PDE5A inhibition. J Mol Cell Cardiol. 2009;46:560–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.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 and 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;309:1268–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Carnicer R, Crabtree MJ, Sivakumaran V, Casadei B and Kass DA. Nitric oxide synthases in heart failure. Antioxid Redox Signal. 2013;18:1078–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Nakamura T, Ranek MJ, Lee DI, Shalkey Hahn V, Kim C, Eaton P and Kass DA. Prevention of PKG1alpha oxidation augments cardioprotection in the stressed heart. J Clin Invest. 2015;125:2468–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Nakamura T, Zhu G, Ranek MJ, Kokkonen-Simon K, Zhang M, Kim GE, Tsujita K and Kass DA. Prevention of PKG-1alpha Oxidation Suppresses Antihypertrophic/Antifibrotic Effects From PDE5 Inhibition but not sGC Stimulation. Circ Heart Fail. 2018;11:e004740. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.De Vecchis R, Cesaro A and Ariano C. Differential effects of the phosphodiesterase inhibition in chronic heart failure depending on the echocardiographic phenotype (HFREF or HFpEF): a meta-analysis. Minerva Cardioangiol. 2018;66:659–670. [DOI] [PubMed] [Google Scholar]
  • 96.Patel NS, Klett J, Pilarzyk K, Lee DI, Kass D, Menniti FS and Kelly MP. Identification of new PDE9A isoforms and how their expression and subcellular compartmentalization in the brain change across the life span. Neurobiol Aging. 2018;65:217–234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Kokkonen-Simon KM, Saberi A, Nakamura T, Ranek MJ, Zhu G, Bedja D, Kuhn M, Halushka MK, Lee DI and Kass DA. Marked disparity of microRNA modulation by cGMP-selective PDE5 versus PDE9 inhibitors in heart disease. JCI Insight. 2018;3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Zhang L, Bouadjel K, Manoury B, Vandecasteele G, Fischmeister R and Leblais V. Cyclic nucleotide signalling compartmentation by PDEs in cultured vascular smooth muscle cells. Br J Pharmacol. 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Scott NJA, Rademaker MT, Charles CJ, Espiner EA and Richards AM. Hemodynamic, Hormonal, and Renal Actions of Phosphodiesterase-9 Inhibition in Experimental Heart Failure. J Am Coll Cardiol. 2019;74:889–901. [DOI] [PubMed] [Google Scholar]
  • 100.Schwam EM, Nicholas T, Chew R, Billing CB, Davidson W, Ambrose D and Altstiel LD. A multicenter, double-blind, placebo-controlled trial of the PDE9A inhibitor, PF-04447943, in Alzheimer's disease. Curr Alzheimer Res. 2014;11:413–21. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplemental Materials - Tables

RESOURCES