Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Letter
  • Published:

Phosphodiesterase 9A controls nitric-oxide-independent cGMP and hypertrophic heart disease

This article has been updated

Abstract

Cyclic guanosine monophosphate (cGMP) is a second messenger molecule that transduces nitric-oxide- and natriuretic-peptide-coupled signalling, stimulating phosphorylation changes by protein kinase G. Enhancing cGMP synthesis or blocking its degradation by phosphodiesterase type 5A (PDE5A) protects against cardiovascular disease1,2. However, cGMP stimulation alone is limited by counter-adaptions including PDE upregulation3. Furthermore, although PDE5A regulates nitric-oxide-generated cGMP4,5, nitric oxide signalling is often depressed by heart disease6. PDEs controlling natriuretic-peptide-coupled cGMP remain uncertain. Here we show that cGMP-selective PDE9A (refs 7, 8) is expressed in the mammalian heart, including humans, and is upregulated by hypertrophy and cardiac failure. PDE9A regulates natriuretic-peptide- rather than nitric-oxide-stimulated cGMP in heart myocytes and muscle, and its genetic or selective pharmacological inhibition protects against pathological responses to neurohormones, and sustained pressure-overload stress. PDE9A inhibition reverses pre-established heart disease independent of nitric oxide synthase (NOS) activity, whereas PDE5A inhibition requires active NOS. Transcription factor activation and phosphoproteome analyses of myocytes with each PDE selectively inhibited reveals substantial differential targeting, with phosphorylation changes from PDE5A inhibition being more sensitive to NOS activation. Thus, unlike PDE5A, PDE9A can regulate cGMP signalling independent of the nitric oxide pathway, and its role in stress-induced heart disease suggests potential as a therapeutic target.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: PDE9A expression in heart and myocytes increases with disease.
Figure 2: PDE9A inhibition suppresses cardiac hypertrophy via natriuretic-peptide–cGMP pathway.
Figure 3: Pressure-overload-induced cardiac pathobiology is suppressed in Pde9a−/− mice.
Figure 4: Chronic PDE9A inhibition reverses pre-established hypertrophy/dysfunction in a NOS-independent manner.

Similar content being viewed by others

Change history

  • 25 March 2015

    The received date year was corrected.

References

  1. Takimoto, E. et al. Chronic inhibition of cyclic GMP phosphodiesterase 5A prevents and reverses cardiac hypertrophy. Nature Med. 11, 214–222 (2005)

    Article  CAS  Google Scholar 

  2. Kukreja, R. C., Salloum, F. N. & Das, A. Cyclic guanosine monophosphate signaling and phosphodiesterase-5 inhibitors in cardioprotection. J. Am. Coll. Cardiol. 59, 1921–1927 (2012)

    Article  CAS  Google Scholar 

  3. Mullershausen, F., Russwurm, M., Koesling, D. & Friebe, A. In vivo reconstitution of the negative feedback in nitric oxide/cGMP signaling: role of phosphodiesterase type 5 phosphorylation. Mol. Biol. Cell 15, 4023–4030 (2004)

    Article  CAS  Google Scholar 

  4. Castro, L. R., Verde, I., Cooper, D. M. & Fischmeister, R. Cyclic guanosine monophosphate compartmentation in rat cardiac myocytes. Circulation 113, 2221–2228 (2006)

    Article  CAS  Google Scholar 

  5. Takimoto, E. et al. Compartmentalization of cardiac β-adrenergic inotropy modulation by phosphodiesterase type 5. Circulation 115, 2159–2167 (2007)

    Article  CAS  Google Scholar 

  6. Carnicer, R., Crabtree, M. J., Sivakumaran, V., Casadei, B. & Kass, D. A. Nitric oxide synthases in heart failure. Antioxid. Redox Signal. 18, 1078–1099 (2013)

    Article  CAS  Google Scholar 

  7. Soderling, S. H., Bayuga, S. J. & Beavo, J. A. Identification and characterization of a novel family of cyclic nucleotide phosphodiesterases. J. Biol. Chem. 273, 15553–15558 (1998)

    Article  CAS  Google Scholar 

  8. Fisher, D. A., Smith, J. F., Pillar, J. S., St Denis, S. H. & Cheng, J. B. Isolation and characterization of PDE9A, a novel human cGMP-specific phosphodiesterase. J. Biol. Chem. 273, 15559–15564 (1998)

    Article  CAS  Google Scholar 

  9. Conti, M. & Beavo, J. Biochemistry and physiology of cyclic nucleotide phosphodiesterases: essential components in cyclic nucleotide signaling. Annu. Rev. Biochem. 76, 481–511 (2007)

    Article  CAS  Google Scholar 

  10. Kleiman, R. J. et al. Phosphodiesterase 9A regulates central cGMP and modulates responses to cholinergic and monoaminergic perturbation in vivo. J. Pharmacol. Exp. Ther. 341, 396–409 (2012)

    Article  CAS  Google Scholar 

  11. Heckman, P. R., Wouters, C. & Prickaerts, J. Phosphodiesterase inhibitors as a target for cognition enhancement in aging and Alzheimer’s disease: a translational overview. Curr. Pharm. Des. 21, 317–331 (2015)

    Article  CAS  Google Scholar 

  12. Sharma, K. & Kass, D. A. Heart failure with preserved ejection fraction: mechanisms, clinical features, and therapies. Circ. Res. 115, 79–96 (2014)

    Article  CAS  Google Scholar 

  13. Nausch, L. W., Ledoux, J., Bonev, A. D., Nelson, M. T. & Dostmann, W. R. Differential patterning of cGMP in vascular smooth muscle cells revealed by single GFP-linked biosensors. Proc. Natl Acad. Sci. USA 105, 365–370 (2008)

    Article  CAS  ADS  Google Scholar 

  14. Singh, G., Kuc, R. E., Maguire, J. J., Fidock, M. & Davenport, A. P. Novel snake venom ligand dendroaspis natriuretic peptide is selective for natriuretic peptide receptor-A in human heart: downregulation of natriuretic peptide receptor-A in heart failure. Circ. Res. 99, 183–190 (2006)

    Article  CAS  Google Scholar 

  15. Davis, J., Burr, A. R., Davis, G. F., Birnbaumer, L. & Molkentin, J. D. A. TRPC6-dependent pathway for myofibroblast transdifferentiation and wound healing in vivo. Dev. Cell 23, 705–715 (2012)

    Article  CAS  Google Scholar 

  16. Koitabashi, N. et al. 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. 48, 713–724 (2010)

    Article  CAS  Google Scholar 

  17. Kinoshita, H. et al. Inhibition of TRPC6 channel activity contributes to the antihypertrophic effects of natriuretic peptides-guanylyl cyclase-A signaling in the heart. Circ. Res. 106, 1849–1860 (2010)

    Article  CAS  Google Scholar 

  18. Stangherlin, A. et al. cGMP signals modulate cAMP levels in a compartment-specific manner to regulate catecholamine-dependent signaling in cardiac myocytes. Circ. Res. 108, 929–939 (2011)

    Article  CAS  Google Scholar 

  19. Pilz, R. B. & Broderick, K. E. Role of cyclic GMP in gene regulation. Front. Biosci. 10, 1239–1268 (2005)

    Article  CAS  Google Scholar 

  20. Oka, T. et al. Cardiac-specific deletion of Gata4 reveals its requirement for hypertrophy, compensation, and myocyte viability. Circ. Res. 98, 837–845 (2006)

    Article  CAS  Google Scholar 

  21. Maurice, D. H. et al. Advances in targeting cyclic nucleotide phosphodiesterases. Nature Rev. Drug Discov. 13, 290–314 (2014)

    Article  CAS  Google Scholar 

  22. Greene, S. J. et al. The cGMP signaling pathway as a therapeutic target in heart failure with preserved ejection fraction. J. Am Heart Assoc. 2, e000536 (2013)

    Article  Google Scholar 

  23. Redfield, M. M. et al. Effect of phosphodiesterase-5 inhibition on exercise capacity and clinical status in heart failure with preserved ejection fraction: a randomized clinical trial. J. Am. Med. Assoc. 309, 1268–1277 (2013)

    Article  CAS  Google Scholar 

  24. van Heerebeek, L. et al. Low myocardial protein kinase G activity in heart failure with preserved ejection fraction. Circulation 126, 830–839 (2012)

    Article  CAS  Google Scholar 

  25. McMurray, J. J. et al. Angiotensin-neprilysin inhibition versus enalapril in heart failure. N. Engl. J. Med. 993–1004 (2014)

  26. Zakeri, R. & Burnett, J. C. Designer natriuretic peptides: a vision for the future of heart failure therapeutics. Can. J. Physiol. Pharmacol. 89, 593–601 (2011)

    Article  CAS  Google Scholar 

  27. Seo, K. et al. Combined TRPC3 and TRPC6 blockade by selective small-molecule or genetic deletion inhibits pathological cardiac hypertrophy. Proc. Natl Acad. Sci. USA 111, 1551–1556 (2014)

    Article  CAS  ADS  Google Scholar 

  28. Verhoest, P. R. et al. Design and discovery of 6-[(3S,4S)-4-methyl-1-(pyrimidin-2-ylmethyl)pyrrolidin-3-yl]-1-(tetrahydro-2H-pyr an-4-yl)-1,5-dihydro-4H-pyrazolo[3,4-d]pyrimidin-4-one (PF-04447943), a selective brain penetrant PDE9A inhibitor for the treatment of cognitive disorders. J. Med. Chem. 55, 9045–9054 (2012)

    Article  CAS  Google Scholar 

  29. Zhou, B. et al. Epicardial progenitors contribute to the cardiomyocyte lineage in the developing heart. Nature 454, 109–113 (2008)

    Article  CAS  ADS  Google Scholar 

  30. Kirk, J. A. et al. Cardiac resynchronization sensitizes the sarcomere to calcium by reactivating GSK-3beta. J. Clin. Invest. 124, 129–139 (2014)

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank students R. D. Wardlow and X. Hu for their assistance with some of the assays and studies. This research was supported by: the National Institutes of Health (NIH) (HL-119012, HL-089297, HL-07227), Fondation Leducq TransAtlantic Network of Excellence, The Peter Belfer Foundation, Abraham and Virginia Weiss Professorship (D.A.K.); HL-093432 (E.T.), American Heart Association (D.I.L.) and Max Kade Fellowship of the Austrian Academy of Sciences (P.P.R). Procurement of human heart tissue was enabled by grants from the National Institutes of Health (HL089847 and HL105993) to K.B.M. N.H. and W.J.P. were supported by the European Commission FP7 project 2010 Health (MEDIA; 261409). R.H. and J.E.V.E. were supported by The Johns Hopkins Innovation Proteomics Center in Heart Failure (NHLBI-HV-10-05 (2) and HHSN268201000032C). W.R.D. was supported by NIH grant HL68891 and the Totman Trust for Biomedical Research. We thank Pfizer and in particular C. Schmidt and R. Kleiman for providing the Pde9a−/− mouse and PF-04449613, and L. Jaffe at the University of Connecticut Health Center for providing the PDE9A antibody.

Author information

Authors and Affiliations

Authors

Contributions

D.I.L. and D.A.K. conceived and directed the project, designed experiments and prepared the manuscript. D.I.L. conducted most of the experiments and analysed the data. G.Z. helped with in vivo experiments. T.S., S.-H.J., T.D., M.Z. and P.P.R. conducted molecular biology experiments. G.-S.C. and C.K. carried out immunostaining and in situ hybridization analyses. N.H. and W.J.P. performed experiments for HFPEF and aortic stenosis human samples. M.J.R. and R.H. performed the experiments for phosphoproteomics and J.A.K. contributed to the data analysis. D.B. performed echocardiography and drug treatment. K.B.M. coordinated the human sample and data collection at the University of Pennsylvania, and W.R.D., J.E.V.E. and E.T. helped with data interpretation and presentation.

Corresponding author

Correspondence to David A. Kass.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Extended data figures and tables

Extended Data Figure 1 Development of Pde9a knockout (Pde9a−/−) and specificity of Pde5a or Pde9a siRNA.

a, Pde9a knockout (Pde9a−/−) mice were developed by replacing the exon 12 region with lacZ-neomycin cassette in the catalytic domain of the carboxy terminal in the Pde9a gene. The genotyping was performed using specific primers designed between exons 11 and 13 including neomycin as following: Gs1 (5′-CACAGATGATGTACAGTATGGTCTGG-3′), Gs2 (5′-TGCAGTCATCAGGACCAAGATGTCC-3′) and Neo (5′-GACGAGTTCTTCTGAGGGGATCGATC-3′). b, The typical genotyping pattern of Pde9a−/− mice was shown on 2% agarose gel (250 bp for wild type and 500 bp for Pde9a−/− mice). c, Selective gene silencing using siRNAs targeting PDE5A or PDE9A. PCR confirms specificity and substantial gene knockdown achieved in cell culture (n = 6 per group).

Extended Data Figure 2 Expression of PDE9A protein in RNCM, mouse brain and human heart.

a, Immunoblot for PDE9A in neonatal cardiomyocytes transfected with either scrambled control siRNA (Sc) or Pde9a siRNA (siR), confirming the suppression of protein expression by the siRNA. A control gel for comparison was derived from brain tissue using Pde9a−/− mice and littermate controls. The band identified at 60 to 65 kDa was similar in both tissues. PDE9A bands are usually identified between 55–70 kDa depending on the splice variants expressed in a given tissue and species. b, Control immunohistochemistry showing that PDE9A detected by antibody can be largely quenched (inactivated) selectively by preincubation with recombinant ligand; scale bar, 50 μm. c, Immunostaining of PDE9A from all 8 control and DCM patients; scale bar, 200 μm. There was consistent enhanced staining in DCM patients versus controls. All images were obtained at an identical level of laser illumination and have not been altered.

Extended Data Figure 3 Selectivity of PDE9A inhibitors.

a, The dose responses of recombinant PDE9A or PDE5A to a selective PDE9A inhibitor (PF-9613) and sildenafil. Data performed in triplicate at each point. A dose of 5 μM PF-9613 inhibited PDE9A effectively, but had negligible impact on PDE5A. By contrast, the PDE5A inhibitor sildenafil inhibited PDE5A by 80% at a dose of 1 μM, commonly used for cells and tissue, but had no impact on PDE9A at this dose. These doses were therefore used in our cell-based studies. b, Confirmation that an alternative PDE9A inhibitor (PF-04447943), currently being tested in humans, shows similar anti-hypertrophic effects as PF-9613 in cardiac myocytes (n = 4 per group); P < 0.001 vs. baseline; #P < 0.01 vs. phenylephrine. c, Inhibition of PKG activity with DT3 reverses the suppression of phenylephrine-stimulated Nppb gene expression by PF-9613. This is a companion panel to Fig. 2b, bottom. n = 6 for basal (no drugs), n = 8 for other groups; P < 0.001 vs. baseline; #P < 0.01 vs. phenylephrine. Data are mean ±s.e.m.

Extended Data Figure 4 PKG activity or cGMP measurement of ANP with inhibitors in RNCM.

a, PF-9613 significantly increases PKG activity assessed by in vitro assay upon stimulation with ANP; n = 4 per group. P < 0.01 vs. other groups. b, PF-9613 augmentation of ANP-stimulated cGMP is not altered due to gene silencing of Pde5a; n = 5. This differs from the complete suppression of cGMP modulation by PF-9613 in myocytes with genetically silenced Pde9a (Fig. 1g). c, SIL does not enhance cGMP stimulated by ANP. This contrasts to its augmentation of nitric-oxide-donor-derived cGMP (Fig. 1h); n = 4. P < 0.01 vs. basal state, #P < 0.01 vs. ANP. Data are mean ±s.e.m.

Extended Data Figure 5 Confocal immunostaining of cardiomyocyte PDE9A and PDE5A.

a, PDE9A does not co-localize with α-actinin at the Z-band, whereas PDE5A does. b, PDE9A does not co-localize with α-actinin in rat neonatal myocytes. c, PDE9A co-localizes with T-tubular membranes as defined by antibody staining against the sarcoplasmic reticular ATPase-2 (SERCA2a). This differed from the localization of PDE5A. Scale bars, 20 μm.

Extended Data Figure 6 Myocardial cAMP levels in controls (sham wild type) and Pde9a−/− mice before and after TAC.

The cAMP levels were increased in TAC wild type, but they were not affected by modulation of PDE9A expression. Sham wild type, n = 4; TAC wild type, n = 5; sham Pde9a−/− n = 6; TAC Pde9a−/− n = 10. Data are mean ±s.e.m.

Extended Data Figure 7 Effect of PF-9613 on blood pressure and cardiac function in mouse.

a, Acute administration of PF-9613 by gavage was studied to assess effects on cardiac pressures, and contractility (end-systolic elastance; Ees). Over a 1-h observation period (peak plasma concentrations found after 30 min) there was no change in any of these parameters; n = 3. b, Chronic treatment of sham control mice with PF-9613 for 3 weeks (n = 3) revealed no effect on cardiac function, mass or volumes. EF, ejection fraction; FS, fractional shortening; LV-ESD, left ventricular end-systolic cross-sectional dimension; LV-EDD, left ventricular end-diastolic cross-sectional dimension. Data are mean ±s.e.m.

Extended Data Figure 8 Effect of chronic PDE9A inhibition on left ventricular mass, lung weight and alteration of TAC-responsive genes.

a, Post-mortem analysis of heart mass and lung weight (both normalized to tibia length) from mice subjected to 5 weeks of pressure overload (TAC) and co-treated with either a vehicle control, PDE9A inhibitor or PDE5A inhibitor. A sham-operation control group is also shown; sham, n = 6; TAC, n = 9; TAC + PF9613, n = 9; TAC + SIL, n = 5. b, Molecular analysis of TAC-responsive (increased expression) genes, including showing similar reductions from either PDE inhibitor in some (for example, Trpc6), a disparity between inhibitors with significant or borderline greater efficacy from PDE9A inhibition in others (for example, Ctgf, Nppa, P < 0.02 and P < 0.1, respectively between PDE5A and PDE9A inhibitor response), and substantial disparities in others (for example, Fn1, P < 0.001 between PDE5A and PDE9A inhibition). Sham, n = 5; TAC, n = 5; TAC + PF9613, n = 6; TAC + SIL, n = 5. P < 0.01 vs. sham; †P < 0.001 vs. sham; #P ≤ 0.05; ‡P < 0.01; §P < 0.001 vs. TAC. Data are mean ±s.e.m.

Extended Data Figure 9 Gene expression of cGMP-hydrolyzing PDEs in Pde9a−/− and littermate controls.

n = 10 per group. The mouse model deleted Pde9a gene expression (normalized to Gapdh), but did not impact the expression of the two other cGMP-regulating PDEs in mouse: Pde1a or Pde5a.

Extended Data Table 1 Clinical characteristics of dilated heart failure and donor control patients

Supplementary information

Supplementary Information

This file contains Supplementary Tables 1-3. (PDF 245 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lee, D., Zhu, G., Sasaki, T. et al. Phosphodiesterase 9A controls nitric-oxide-independent cGMP and hypertrophic heart disease. Nature 519, 472–476 (2015). https://doi.org/10.1038/nature14332

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature14332

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing