Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Aug 20.
Published in final edited form as: Circ Res. 2011 Sep 8;109(9):1024–1030. doi: 10.1161/CIRCRESAHA.111.250464

Phosphodiesterase 4D (PDE4D) regulates baseline sarcoplasmic reticulum Ca2+ release and cardiac contractility, independently of L-type Ca2+current

Sanja Beca 1,4,#, Peter B Helli 1,4,#, Jeremy A Simpson 1,4, Dongling Zhao 1,4, Gerrie P Farman 1,4, Peter Jones 5, Xixi Tian 5, Lindsay S Wilson 6, Faiyaz Ahmad 8, SR Wayne Chen 5, Matthew A Movsesian 9, Vincent Manganiello 8, Donald H Maurice 6,7, Marco Conti 10, Peter H Backx 1,2,3,4
PMCID: PMC4138878  NIHMSID: NIHMS503188  PMID: 21903937

Abstract

Rationale

Baseline contractility of mouse hearts is modulated in a PI3Kγ-dependent manner by type 4 phosphodiesterases (PDE4), which regulate cAMP levels within microdomains containing the sarcoplasmic reticular (SR) calcium-ATPase (SERCA2a).

Objective

To determine whether PDE4D regulates basal cAMP levels, phospholamban (PLN) phosphorylation and SERCA2a activity in SR microdomains.

Methods & Results

We assessed myocardial function in PDE4D-deficient (PDE4D−/−) and littermate wild-type (WT) mice at 10-12 weeks of age. Baseline cardiac contractility in PDE4D−/− mice was elevated in vivo and in Langendorff perfused hearts, while isolated PDE4D−/− cardiomyocytes showed increased Ca2+ transient amplitudes and SR Ca2+content, but unchanged ICa(L), compared to WT. The PKA inhibitor, Rp-cAMPS, lowered Ca2+ transient amplitudes and SR Ca2+ content in PDE4D−/− cardiomyocytes to WT levels. The PDE4 inhibitor rolipram (ROL) had no effect on cardiac contractility, Ca2+ transients or SR Ca2+ content in PDE4D−/− preparations but increased these parameters in WT hearts to levels indistinguishable from those in PDE4D−/−. The functional changes in PDE4D−/− myocardium were associated with increased PLN phosphorylation (pPLN) but not RyR2 receptor phosphorylation. ROL increased pPLN in WT cardiomyocytes to levels indistinguishable from those in PDE4D−/− cardiomyocytes. In murine and failing human hearts, PDE4D co-immunoprecipitated with SERCA2a but not with RyR2.

Conclusions

PDE4D regulates basal cAMP levels in SR microdomains through its interactions with SERCA2a-PLN. Since Ca2+ transient amplitudes are reduced in failing human myocardium, these observations may have therapeutic implications for patients with heart failure.

Keywords: PDE4D, cAMP, cardiac function, excitation-contraction coupling

Introduction

Cyclic nucleotide phosphodiesterases (PDEs) hydrolyze and inactivate cAMP. The PDE super-family is comprised of eleven gene families. Since individual families can contain as many as four genes that can be processed to yield multiple transcripts, most cells express numerous PDEs. Recent studies have established that certain PDEs can selectively interact with other cellular proteins, assemble into specialized macromolecular complexes within discrete functional compartments, and allow precise spatio-temporal control of cellular cAMP-mediated, PKA-dependent signalling1;2. The heart can express PDEs from each of the PDE1 through PDE5 families, as well as the PDE8 family3. Since reductions in cAMP/PKA-signalling contribute to impaired cardiac function in heart disease patients4, PDE inhibitors were initially promoted for treating heart failure. However, despite their benefit in treating contractile failure, prolonged treatment of heart failure patients with PDE inhibitors increases mortality, principally by increasing sudden cardiac death5. Selective targeting of PDE isoenzymes may provide novel opportunities to correct the impaired cAMP-dependent signalling seen in heart disease without these adverse consequences.

Consistent with current paradigms of cAMP compartmentalization, PDE3 and PDE4 enzymes suppress basal cAMP/PKA-signalling and contractility in cellular microdomains containing sarcoplasmic reticulum (SR) Ca2+pumps, but not ryanodine receptors (RyR2s) or L-type Ca2+channels6;7. Since PDE4D isoforms associate with SERCA2a7 and RyR2 receptors8, we investigated PDE4D's role in regulating cardiac contractility. Mice lacking PDE4D have enhanced baseline cardiac contractility associated with increased PLN phosphorylation, SR Ca2+ content and Ca2+ transients but not elevated ICa,L or RyR2 phosphorylation. PDE4D also co-assembles with SERCA2a but not with RyR2 in both murine and human hearts.

Methods

PDE4D deficient (PDE4D−/−)9 and littermate wild-type (WT) mice (129vj/c57 background) were studied at 10-12 weeks of age. Experiments were conducted in accordance with the Canadian Council of Animal Care. Detailed methods are in the Online Data Supplement at http://circres.ahajournals.org.

Results

Consistent with previous studies8, PDE4D ablation did not affect heart-weight to bodyweight ratios or heart morphometry assessed with echocardiography (Online Table I) but did cause mild reductions (P<0.005) in mean arterial blood pressure (MAP) and elevations (P<0.05) in ventricular contractility compared to WT (Online Figure I & Table I). Since assessment of contractility is complicated by MAP differences, we studied Langendorff hearts, which showed elevated (P<0.01) left ventricular developed pressures (LVDP) as well as the peak time-derivatives of pressure (dP/dtmax and dP/dtmin) in PDE4D−/− hearts (Online Figure II, Online Table II). As expected7, the PDE4 inhibitor rolipram (ROL) increased contractility and elevated heart rates (HR) in WT hearts; to eliminate the effect of the latter on contractility, hearts were paced at 9Hz, faster than the beating rate of ROL-treated hearts. Contractility (LVDP, dP/dtmax and dP/dtmin) remained higher (P< 0.01) in paced PDE4D−/− hearts compared to WT (Figure 1, Online Table II). ROL had no effect on PDE4D−/− hearts, but increased (P<0.01) contractility in WT to levels indistinguishable (P= 0.14) from PDE4D−/− (Online Table II). Shortening of isolated PDE4D−/− cardiomyocytes was also higher (P<0.05) and ROL augmented shortening (P<0.05) in WT cardiomyocytes to levels indistinguishable (P=0.76) from PDE4D−/− (Online Figure III). Thus, PDE4D ablation increases baseline ventricular myocardial contractility and eliminates inotropic responses to PDE4 inhibition.

Figure 1. Assessment of Langendorff heart function.

Figure 1

(A) Records of left ventricular pressure (top), dP/dtmax and dP/dtmin (bottom) measured in paced (9Hz) Langendorff-perfused hearts before and after ROL infusion. (B) Mean data for LVP (left), dP/dtmax and dP/dtmin (middle) and % change from baseline following ROL treatment (right), with baseline recorded after a 20 min equilibration period. *P<0.05 vs.WT, **P<0.01 vs. WT

To explore the cellular mechanisms underlying these PDE4D effects, Ca2+ transients and L-type Ca2+ currents (ICa,L) were simultaneously recorded in voltage-clamped ventricular cardiomyocytes. PDE4D−/− myocytes had increased (P<0.01) Ca2+ transient amplitudes and decay rates, without alterations in ICa,L compared to WT (Figure 2, Online Table III). Intracellular dialysis with the PKA inhibitor Rp -cAMPS10 had no effect on WT myocytes but reduced Ca2+ transients in PDE4D−/− to WT levels (Figure 2, Online Table III). On the other hand, the SR Ca2+ content (i.e. integrated Na+/Ca2+ exchanger (NCX) currents following caffeine exposure) was higher (P<0.01) in PDE4D−/− myocytes than in WT myocytes, and Rp-cAMPS abolished these differences. ROL had no effect (P=0.91) on SR Ca2+ content of PDE4D−/− myocytes, but caused increases (P<0.01) in WT myocytes to levels indistinguishable (P=0.86) from those of PDE4D−/− myocytes (Figure 3).

Figure 2. Ca2+ transients and ICaL Measurements.

Figure 2

(A) Ca2+ transients (upper) and ICaL (lower) recorded for WT and PDE4D−/− cardiomyocytes in response to voltage steps (indicated) from –85mV holding potential and a 500 msec ramp to -45mV. (B) Typical Ca2+ transients and ICa,L at +10mV before and after ROL application. Mean Ca2+ transient and ICa,L peaks as a function of voltage in the presence and absence of RpcAMPS (C) and ROL (D). *P<0.01 versus control within same group; †P<0.01 versus WT control

Figure 3. Measurements of SR Ca2+ content (A) as well as PLN and RyR2 phosphorylation (B&C).

Figure 3

(A) INCX evoked by a 10s application of 20mM caffeine in the presence or absence of RpcAMPS or ROL (left). Mean (time) integrated INCX to assess SR Ca2+ content bottom (right). *P< 0.05 versus WT. (B) Representative Western blot of protein extracts from left ventricular cardiomyocytes to measure phosphorylated PLN (left). Average intensity ratios of pPLN/PLNtotal(right). *P< 0.05 versus WT; †P<0.05 vs. PDE4D−/− (n=5 hearts). (C) Representative WB of protein extracts from left ventricular cardiomyocytes illustrating the effect of PDE4D ablation on phosphorylated RyR2 (pRyR2) levels (left). Mean data showing changes in pRyR2/ RyR2total ratios in PDE4D−/− hearts (right). *P<0.05 versus PDE4D WT (n= 4 hearts).

Compared to WT, PDE4D−/− myocardium had elevated (P<0.05) PLN phosphorylation (pPLN) levels (Figure 3) which were indistinguishable from ROL-treated WT myocardium as well as either ROL-treated or ROL-untreated PDE4D−/− myocardium (not shown). In contrast, RyR2 phosphorylation (Figure 3) was not different (P=0.11) at Serine 2030, the PKA-dependent site regulating function11, or S2814 (P=0.79), but was unexpectedly8 decreased (8%, P=0.01) at S2808. Co-immunoprecipitation in preparations from mouse and human myocardium revealed that PDE4D associates with SERCA2a but not RyR2 (Figure 4). Although PI3Kγ is required for PDE4 activity in micro-domains containing SERCA2a8, it was not detected in SERCA2a immunoprecipitates, suggesting that PI3Kγ's enzymatic activity6;7;12 is required. This is consistent with elevated contractility seen in Langendorff hearts treated with the PI3Kinase inhibitor, wortmannin (Online Figure VI).

Figure 4. PDE4D interactions with SERCA2a in murine and human myocardium.

Figure 4

A shows representative Western Blots (repeated in 3 separate hearts) probing with PDE4D in heart lysates from a PDE4D−/− mouse as well as for WT mouse heart homogenates that had been immunoprecipitated using control IgG, or using RyR2- or SERCA2a-specific antibodies. Results show that PDE4D antibodies recognized strong bands at MWs of 97kDaltons (corresponding to PDED-3,-7 and -9 splice variants of PDE4D) in immunoprecipitation reactions with anti-SERCA2a antibodies, but not with anti-RYR2 antibodies or with IgG controls. A very weak nonspecific band having MW ~ 110kDaltons was detected in all immunoprecipitation groups as well as in homogenates from the PDE4D-null mice, confirming this is a nonspecific band of unknown origin. B shows representative inputs for immunoprecipitation reactions shown in A. C shows results of SERCA2a immunoprecipitation in human hearts. (D) Diagram summarizing implications of our results in WT (left) and PDE4D−/− (right) myocardium.

Discussion

PDE4D−/− mice were hypotensive and had enhanced myocardial contractility associated with increases in cardiomyocyte shortening, Ca2+ transient amplitudes, Ca2+ transient relaxation rates and SR Ca2+ loads, without changes in ICa,L, compared to WT. PKA-inhibition with cAMP antagonists eliminated these cellular differences. In addition, PLN phosphorylation (at PKA site S-16), but not RyR2 phosphorylation, was elevated in PDE4D−/− hearts. As shown previously7, PDE4D isoforms with molecular weights of ~97kDa (i.e. PDE4D3/8/9) co-immunoprecipitated with SERCA2a in both human and murine hearts. These findings support the conclusion that PDE4D regulates basal cAMP levels (and thus PKA activity) in macromolecular complexes containing the SR Ca2+ATPase without functionally influencing (spatially adjacent) RyR2 receptors or L-type Ca2+channels. Our inability to identify PI3Kγ in SERCA2 immunoprecipitates, suggests that PI3Kγ does not regulate PDE4D via protein-protein interactions, as with PDE3B13, but requires enzymatic activity, as in mice lacking PTEN-phosphatase12. Indeed, wortmannin increased ventricular contractility/ relaxation (Online Figure VI).

Selective inhibition of PDE414 elevated contractility, Ca2+ transients, Ca2+ SR loads and PLN phosphorylation in WT hearts, as reported7, to levels seen in PDE4D−/− myocardium without affecting PDE4D−/−. Thus, although PDE4A and PDE4B are expressed in mouse heart15, PDE4D underlies the cAMP-dependent baseline contractile responses to PDE4 inhibitors. Whilst PDE4D ablation does not induce expression changes of other PDE isoforms in different organs9, changes in activity/expression of other PDE isozymes might have occurred. In this regard, responses to PDE3 inhibitors (which also affect mouse baseline contractility7;16) were unaffected by PDE4D ablation (Online Figure V). Although inhibition of other PDE isozymes was not examined, the consequences of PDE4D ablation on cardiac function can be readily explained by the loss of PDE4D activity alone.

A previous study reported that PDE4D and RyR2 interact in murine cardiomyocytes and that PDE4D−/− mice develop both heart dysfunction and arrhythmias by 9 months of age resulting from RyR2 hyperphosphorylation at S-28088. We found neither elevated RyR2 phosphorylation in PDE4D−/− hearts nor evidence of direct interactions between PDE4D and RyR2 in murine hearts. These differences may be related to differences in reagents used in the two studies or to age-dependent changes in cAMP regulation and/or heart function17. In this regard, we did observe declines in cardiac function of PDE4D−/− mice at 9 months8, which helps explain why another study15, focusing on β-adrenergic responses, only identified acceleration of Ca2+ transient relaxation in PDE4D−/− myocytes at 5-6 months, accompanied by trends (non-significant) towards elevations of Ca2+ transients as well as myocyte shortening/relaxation (discussed further, Online Supplement).

Age-dependent deterioration of PDE4D−/− heart function is not inconsistent with human data showing that persistent cAMP-dependent stimulation with, for example, PDE3 inhibitors promotes disease progression, mortality and arrhythmias in heart disease patients, despite providing short-term benefit (by enhancing contractility) in transplant and heart failure patients18. On the other hand, clinical trials with PDE4 inhibitors did not identify cardiovascular side-effects (besides slight increases in atrial fibrillation)19. These differences between mouse and humans may arise from higher relative PDE4 activities in mouse (~35%) versus human (<10%) myocardium20. However, PDE activity is highly compartmentalized1, making local (not global) activity most relevant functionally. Thus, PDE4D tethering to SERCA2a combined with the high PDE4 activity (~50% of total activity) observed in PLN immunoprecipitates from human myocardium20 suggests that PDE4D fine-tunes the cAMP-dependent SR Ca2+-ATPase activity in human hearts20. This observation is particularly relevant since heart disease/failure is invariably associated with impairment of cAMP/PKA-dependent SR Ca2+-ATPase activity. Moreover, SERCA2a gene therapy21 or beta-blocker treatment (which enhance SERCA2a expression22) improves heart function and longevity. Thus, our findings support the possibility that selective PDE4D inhibition could prove beneficial for treating heart disease by specifically elevating cAMP in SR micro-domains containing SERCA2a/PLN. Remarkably, there is a paucity of definitive data available on direct actions of PDE4 inhibitors on cardiac function in humans or in animal models. Clearly, more studies are required to fully determine the role of PDE4D and its inhibition in heart disease patients.

Supplementary Material

Supplemental Material

Novelty and significance.

“What is known?”

  • Cyclic nucleotide phosphodiesterases (PDEs) are a complex family of enzymes encoded by 23 distinct genes that degrade cAMP/cGMP.

  • PDEs are typically found in macromolecular complexes allowing tight spatial and temporal control of cAMP-dependent signalling in cellular microdomains.

  • Family-specific PDE inhibitors are used clinically for inotropic support in heart failure patients; however, their prolonged use increases mortality.

“What new information does this article contribute?”

  • PDE4D is tethered to the sarcoplasmic reticular (SR) Ca2+ ATPase type 2a (SERCA2a) thereby suppressing baseline cAMP/PKA-dependent Ca2+ cycling

Although PDE inhibitors can improve cardiac function, their prolonged use is associated with heart-related morbidity and mortality. Given the diversity of PDE isozymes, precise targeting of selected PDE isoforms is predicted to enable pinpoint modulation cAMP-dependent signalling in cellular microdomains. Indeed, we show that genetic elimination of PDE4D (one of 4 genes in the PDE4 family) elevates cAMP levels in subcellular compartments containing the SR Ca2+ pump (i.e. SERCA2a/phospholamban), but not L-type Ca2+ channels or cardiac ryanodine receptors, leading to elevations in phospholamban phosphorylation, SR Ca2+ levels, Ca2+ transients and cardiac contractility. This data shows that, in principle, selective pharmacological targeting of PDE4D in SR microdomains allows precise pharmacological control of cardiac contractility, while potentially minimizing the side-effects associated with broader spectrum inhibition. These findings might have significant implications in developing treatment strategies for heart disease patients.

Acknowledgments

Sources of Funding

Supported by Canadian Institutes of Health Research (CIHR) grant to P.H.B, who is a Career Investigator with the Heart and Stroke Foundation (HSF) of Ontario. S.B. holds a postdoctoral fellowship from the Heart and Stroke Richard Lewar Centre of Excellence (HSRLCE), University of Toronto. P.B.H held postdoctoral fellowships from HSRLCE and HSF of Canada. Foundation Leducq grant (06CVD02 cycAMP) to M.C. and M.A.M. The NIH grant (HL0927088) to M.C. and grants from the United States Department of Veterans Affairs Medical Research Funds to M.A.M.

Non-standard Abbreviations and Acronyms

AKAP 18

A-kinase anchoring protein-18

cAMP

adenosine-3’, 5’ cyclic monophosphate

Ca2+ transients

whole cell Ca2+ transients

dP/dtmax

first positive time derivative of the maximum pressure development

dP/dtmin

first negative time derivative of the maximum pressure development

EDP

end diastolic pressure

ICa(L)

L-type calcium current

LVP

left ventricular pressure

LVDP

left ventricular developed pressure

MAP

mean arterial pressure

PDE4D

phosphodiesterase type 4D

PI3Kγ

phosphoinositide 3-kinase gamma

PLN

phospholamban

pPLN

phosphorylated phospholamban

PKA

protein kinase A

PP1

protein phosphatase 1

ROL

rolipram, PDE4 selective inhibitor

Rp-cAMPS

Rp-adenosine-3’, 5’ cyclic monophosphorothioate

RyR2

cardiac ryanodine receptor

SERCA2a

sarcoplasmic reticulum calcium ATPase type 2a

Footnotes

Disclosures

None.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Reference List

  • 1.Houslay MD, Adams DR. PDE4 cAMP phosphodiesterases: modular enzymes that orchestrate signalling cross-talk,desensitization and compartmentalization. Biochem J. 2003;370:1–18. doi: 10.1042/BJ20021698. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Zaccolo M, Movsesian MA. cAMP and cGMP signalling cross-talk:role of phosphodiesterases and implications for cardiac pathophysiology. Circ Res. 2007;100:1569–1578. doi: 10.1161/CIRCRESAHA.106.144501. [DOI] [PubMed] [Google Scholar]
  • 3.Patrucco E, Albergine MS, Santana LF, Beavo JA. Phosphodiesterase 8A(PDE8A) regulates excitation-contraction coupling in ventricular myocytes. J Mol Cell Cardiol. 2010;49:330–333. doi: 10.1016/j.yjmcc.2010.03.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Miller CL, Yan C. Targeting Cyclic Nucleotide Phosphodiesterase in the Heart:Therapeutic Implications. J Cardiovasc Transl Res. 2010 doi: 10.1007/s12265-010-9203-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Amsallem E, Kasparian C, Haddour G, Boissel JP, Nony P. Phosphodiesterase iii inhibitors for heart failure. Cochrane database of systematic reviews (Online) 2005:CD002230. doi: 10.1002/14651858.CD002230.pub2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Kerfant BG, Gidrewicz D, Sun H, Oudit GY, Penninger JM, Backx PH. Cardiac sarcoplasmic reticulum calcium release and load are enhanced by subcellular cAMP elevations in PI3Kgamma-deficient mice. Circ Res. 2005;96:1079–1086. doi: 10.1161/01.RES.0000168066.06333.df. [DOI] [PubMed] [Google Scholar]
  • 7.Kerfant BG, Zhao D, Lorenzen-Schmidt I, Wilson LS, Cai S, Chen SR, Maurice DH, Backx PH. PI3Kgamma is required for PDE4, not PDE3, activity in subcellular microdomains containing the sarcoplasmic reticular calcium ATPase in cardiomyocytes. Circ Res. 2007;101:400–408. doi: 10.1161/CIRCRESAHA.107.156422. [DOI] [PubMed] [Google Scholar]
  • 8.Lehnart SE, Wehrens XH, Reiken S, Warrier S, Belevych AE, Harvey RD, Richter W, Jin SL, Conti M, Marks AR. Phosphodiesterase 4D deficiency in the ryanodine-receptor complex promotes heart failure and arrhythmias. Cell. 2005;123:25–35. doi: 10.1016/j.cell.2005.07.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Jin SL, Richard FJ, Kuo WP, D'Ercole AJ, Conti M. Impaired growth and fertility of cAMP-specific phosphodiesterase PDE4D-deficient mice. Proc Natl Acad Sci U S A. 1999;96:11998–12003. doi: 10.1073/pnas.96.21.11998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Rothermel JD, Stec WJ, Baraniak J, Jastorff B, Botelho LH. Inhibition of glycogenolysis in isolated rat hepatocytes by the Rp diastereomer of adenosine cyclic 3′,5′-phosphorothioate. J Biol Chem. 1983;258:12125–12128. [PubMed] [Google Scholar]
  • 11.Xiao B, Zhong G, Obayashi M, Yang D, Chen K, Walsh MP, Shimoni Y, Cheng H, Ter Keurs H, Chen SR. Ser-2030, but not Ser-2808, is the major phosphorylation site in cardiac ryanodine receptors responding to protein kinase A activation upon beta-adrenergic stimulation in normal and failing hearts. Biochem J. 2006;396:7–16. doi: 10.1042/BJ20060116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Crackower MA, Oudit GY, Kozieradzki I, Sarao R, Sun H, Sasaki T, Hirsch E, Suzuki A, Shioi T, Irie-Sasaki J, Sah R, Cheng HY, Rybin VO, Lembo G, Fratta L, Oliveira-dos-Santos AJ, Benovic JL, Kahn CR, Izumo S, Steinberg SF, Wymann MP, Backx PH, Penninger JM. Regulation of myocardial contractility and cell size by distinct PI3K-PTEN signaling pathways. Cell. 2002;110:737–749. doi: 10.1016/s0092-8674(02)00969-8. [DOI] [PubMed] [Google Scholar]
  • 13.Patrucco E, Notte A, Barberis L, Selvetella G, Maffei A, Brancaccio M, Marengo S, Russo G, Azzolino O, Rybalkin SD, Silengo L, Altruda F, Wetzker R, Wymann MP, Lembo G, Hirsch E. PI3Kgamma modulates the cardiac response to chronic pressure overload by distinct kinase-dependent and -independent effects. Cell. 2004;118:375–387. doi: 10.1016/j.cell.2004.07.017. [DOI] [PubMed] [Google Scholar]
  • 14.Reeves ML, Leigh BK, England PJ. The identification of a new cyclic nucleotide phosphodiesterase activity in human and guinea-pig cardiac ventricle. Implications for the mechanism of action of selective phosphodiesterase inhibitors. Biochem J. 1987;241:535–541. doi: 10.1042/bj2410535. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Leroy J, Richter W, Mika D, Castro LR, Abi-Gerges A, Xie M, Scheitrum C, Lefebvre F, Schittl J, Mateo P, Westenbroek R, Catterall WA, Charpentier F, Conti M, Fischmeister R, Vandecasteele G. Phosphodiesterase 4B in the cardiac L-type Ca2+ channel complex regulates Ca2+ current and protects against ventricular arrhythmias in mice. J Clin Invest. 2011;121:2651–2661. doi: 10.1172/JCI44747. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Osadchii OE. Myocardial phosphodiesterases and regulation of cardiac contractility in health and cardiac disease. Cardiovasc Drugs Ther. 2007;21:171–194. doi: 10.1007/s10557-007-6014-6. [DOI] [PubMed] [Google Scholar]
  • 17.Lakatta EG. Arterial and cardiac aging: major shareholders in cardiovascular disease enterprises: Part III: cellular and molecular clues to heart and arterial aging. Circulation. 2003;107:490–497. doi: 10.1161/01.cir.0000048894.99865.02. [DOI] [PubMed] [Google Scholar]
  • 18.Packer M, Carver JR, Rodeheffer RJ, Ivanhoe RJ, DiBianco R, Zeldis SM, Hendrix GH, Bommer WJ, Elkayam U, Kukin ML. Effect of oral milrinone on mortality in severe chronic heart failure.The PROMISE Study Research Group. N Engl J Med. 1991;325:1468–1475. doi: 10.1056/NEJM199111213252103. [DOI] [PubMed] [Google Scholar]
  • 19.Calverley PM, Rabe KF, Goehring UM, Kristiansen S, Fabbri LM, Martinez FJ. Roflumilast in symptomatic chronic obstructive pulmonary disease:two randomised clinical trials. Lancet. 2009;374:685–694. doi: 10.1016/S0140-6736(09)61255-1. [DOI] [PubMed] [Google Scholar]
  • 20.Richter W, Xie M, Scheitrum C, Krall J, Movsesian MA, Conti M. Conserved expression and functions of PDE4 in rodent and human heart. Basic Res Cardiol. 2011;106:249–262. doi: 10.1007/s00395-010-0138-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.del Monte F, Williams E, Lebeche D, Schmidt U, Rosenzweig A, Gwathmey JK, Lewandowski ED, Hajjar RJ. Improvement in survival and cardiac metabolism after gene transfer of sarcoplasmic reticulum Ca(2+)-ATPase in a rat model of heart failure. Circulation. 2001;104:1424–1429. doi: 10.1161/hc3601.095574. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Lowes BD, Gilbert EM, Abraham WT, Minobe WA, Larrabee P, Ferguson D, Wolfel EE, Lindenfeld J, Tsvetkova T, Robertson AD, Quaife RA, Bristow MR. Myocardial gene expression in dilated cardiomyopathy treated with beta-blocking agents. New England Journal of Medicine. 2002;346:1357–1365. doi: 10.1056/NEJMoa012630. [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 Material

RESOURCES