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.

  • Review Article
  • Published:

Therapeutic targeting of 3′,5′-cyclic nucleotide phosphodiesterases: inhibition and beyond

This article has been updated

Abstract

Phosphodiesterases (PDEs), enzymes that degrade 3′,5′-cyclic nucleotides, are being pursued as therapeutic targets for several diseases, including those affecting the nervous system, the cardiovascular system, fertility, immunity, cancer and metabolism. Clinical development programmes have focused exclusively on catalytic inhibition, which continues to be a strong focus of ongoing drug discovery efforts. However, emerging evidence supports novel strategies to therapeutically target PDE function, including enhancing catalytic activity, normalizing altered compartmentalization and modulating post-translational modifications, as well as the potential use of PDEs as disease biomarkers. Importantly, a more refined appreciation of the intramolecular mechanisms regulating PDE function and trafficking is emerging, making these pioneering drug discovery efforts tractable.

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

Access options

Buy this article

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

Fig. 1: Cyclic nucleotide signalling pathways.
Fig. 2: The 21 PDE genes, grouped into families.
Fig. 3: Mechanisms that activate PDE catalytic activity.
Fig. 4: Methods for targeting PDE signalling with increasing specificity.
Fig. 5: PDE regulation by post-translational modification.

Similar content being viewed by others

Change history

  • 15 August 2019

    The acknowledgements were amended to include the grants supporting the work of G.S.B. and to indicate that G.S.T is a fellow of the Astrazeneca postdoctoral programme.

References

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

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Schneider, E. H. & Seifert, R. Inactivation of non-canonical cyclic nucleotides: hydrolysis and transport. Handb. Exp. Pharmacol. 238, 169–205 (2017). This work is a helpful review of the role PDEs play in regulating non-canonical cyclic nucleotides.

    CAS  PubMed  Google Scholar 

  3. Herbst, S. et al. Transmembrane redox control and proteolysis of PdeC, a novel type of c-di-GMP phosphodiesterase. EMBO J. 37, e97825 (2018).

    PubMed  PubMed Central  Google Scholar 

  4. Seifert, R. cCMP and cUMP across the tree of life: from cCMP and cUMP generators to cCMP- and cUMP-regulated cell functions. Handb. Exp. Pharmacol. 238, 3–23 (2017).

    CAS  PubMed  Google Scholar 

  5. Patel, N. S. et al. Identification of new PDE9A isoforms and how their expression and subcellular compartmentalization in the brain change across the life span. Neurobiol. Aging 65, 217–234 (2018). This study is a first report showing that the subcellular compartmentalization of a PDE family differs depending on the specific isoform, age and tissue investigated.

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Huston, E., Gall, I., Houslay, T. M. & Houslay, M. D. Helix-1 of the cAMP-specific phosphodiesterase PDE4A1 regulates its phospholipase-D-dependent redistribution in response to release of Ca2+. J. Cell Sci. 119, 3799–3810 (2006). This study provides the first evidence that PDEs can be relocated by Ca 2+ , providing a potential mechanism by which cAMP, phospholipase D and Ca 2+ -signalling pathways interact.

    CAS  PubMed  Google Scholar 

  7. Nagel, D. J. et al. Role of nuclear Ca2+/calmodulin-stimulated phosphodiesterase 1A in vascular smooth muscle cell growth and survival. Circ. Res. 98, 777–784 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Houslay, M. D. & Baillie, G. S. Beta-arrestin-recruited phosphodiesterase-4 desensitizes the AKAP79/PKA-mediated switching of beta2-adrenoceptor signalling to activation of ERK. Biochem. Soc. Trans. 33, 1333–1336 (2005).

    CAS  PubMed  Google Scholar 

  9. Perera, R. K. et al. Microdomain switch of cGMP-regulated phosphodiesterases leads to ANP-induced augmentation of beta-adrenoceptor-stimulated contractility in early cardiac hypertrophy. Circ. Res. 116, 1304–1311 (2015). This study is an important work showing that the subcellular compartmentalization of a PDE may change with disease.

    CAS  PubMed  Google Scholar 

  10. Penmatsa, H. et al. Compartmentalized cyclic adenosine 3΄,5΄-monophosphate at the plasma membrane clusters PDE3A and cystic fibrosis transmembrane conductance regulator into microdomains. Mol. Biol. Cell 21, 1097–1110 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Ahmad, F. et al. Differential regulation of adipocyte PDE3B in distinct membrane compartments by insulin and the beta3-adrenergic receptor agonist CL316243: effects of caveolin-1 knockdown on formation/maintenance of macromolecular signalling complexes. Biochem. J. 424, 399–410 (2009). The results of this study suggest elevations in cAMP can trigger a subcellular redistribution of PDEs.

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Al-Tawashi, A. & Gehring, C. Phosphodiesterase activity is regulated by CC2D1A that is implicated in non-syndromic intellectual disability. Cell Commun. Signal. 11, 47 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Bonkale, W. L., Winblad, B., Ravid, R. & Cowburn, R. F. Reduced nitric oxide responsive soluble guanylyl cyclase activity in the superior temporal cortex of patients with Alzheimer’s disease. Neurosci. Lett. 187, 5–8 (1995). This work is an example of a human study showing that cyclic nucleotide signalling deficits associated with disease can be specific to a particular subcellular compartment.

    CAS  PubMed  Google Scholar 

  14. Baltrons, M. A., Pifarre, P., Ferrer, I., Carot, J. M. & Garcia, A. Reduced expression of NO-sensitive guanylyl cyclase in reactive astrocytes of Alzheimer disease, Creutzfeldt-Jakob disease, and multiple sclerosis brains. Neurobiol. Dis. 17, 462–472 (2004).

    CAS  PubMed  Google Scholar 

  15. Baltrons, M. A., Pedraza, C. E., Heneka, M. T. & Garcia, A. Beta-amyloid peptides decrease soluble guanylyl cyclase expression in astroglial cells. Neurobiol. Dis. 10, 139–149 (2002).

    CAS  PubMed  Google Scholar 

  16. Yarla, N. S. et al. Targeting the paracrine hormone-dependent guanylate cyclase/cGMP/phosphodiesterases signaling pathway for colorectal cancer prevention. Semin. Cancer Biol. 56, 168–174 (2018).

    PubMed  Google Scholar 

  17. Li, N. et al. Suppression of beta-catenin/TCF transcriptional activity and colon tumor cell growth by dual inhibition of PDE5 and 10. Oncotarget 6, 27403–27415 (2015).

    PubMed  PubMed Central  Google Scholar 

  18. Lee, D. I. et al. Phosphodiesterase 9A controls nitric-oxide-independent cGMP and hypertrophic heart disease. Nature 519, 472–476 (2015). This study clarifies that, in the heart, PDE9 regulates pools of cGMP downstream of particulate guanylyl cyclases whereas PDE5 regulates pools of cGMP downstream of soluble guanylyl cyclases.

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Hartell, N. A., Furuya, S., Jacoby, S. & Okada, D. Intercellular action of nitric oxide increases cGMP in cerebellar Purkinje cells. Neuroreport 12, 25–28 (2001).

    CAS  PubMed  Google Scholar 

  20. Rahman, S. et al. Reduced [3H]cyclic AMP binding in postmortem brain from subjects with bipolar affective disorder. J. Neurochem. 68, 297–304 (1997). This work is another example of a human study showing cyclic nucleotide signalling deficits associated with disease can be specific to a particular subcellular compartment.

    CAS  PubMed  Google Scholar 

  21. Mori, S. et al. Effects of lithium on cAMP-dependent protein kinase in rat brain. Neuropsychopharmacology 19, 233–240 (1998).

    CAS  PubMed  Google Scholar 

  22. Francis, S. H. & Corbin, J. D. Phosphodiesterase-5 inhibition: the molecular biology of erectile function and dysfunction. Urol. Clin. North Am. 32, 419–429 (2005).

    PubMed  Google Scholar 

  23. Schwartz, B. G. & Kloner, R. A. Drug interactions with phosphodiesterase-5 inhibitors used for the treatment of erectile dysfunction or pulmonary hypertension. Circulation 122, 88–95 (2010).

    PubMed  Google Scholar 

  24. Zoccarato, A. et al. Cardiac hypertrophy is inhibited by a local pool of cAMP regulated by phosphodiesterase 2. Circ. Res. 117, 707–719 (2015).

    CAS  PubMed  Google Scholar 

  25. Kaname, T. et al. Heterozygous mutations in cyclic AMP phosphodiesterase-4D (PDE4D) and protein kinase A (PKA) provide new insights into the molecular pathology of acrodysostosis. Cell. Signal. 26, 2446–2459 (2014).

    CAS  PubMed  Google Scholar 

  26. Motte, E., Le Stunff, C., Briet, C., Dumaz, N. & Silve, C. Modulation of signaling through GPCR–cAMP–PKA pathways by PDE4 depends on stimulus intensity: possible implications for the pathogenesis of acrodysostosis without hormone resistance. Mol. Cell. Endocrinol. 442, 1–11 (2017).

    CAS  PubMed  Google Scholar 

  27. Boscutti, G., Rabiner, E. A. & Plisson, C. PET radioligands for imaging of the PDE10A in human: current status. Neurosci. Lett. 691, 11–17 (2019).

    CAS  PubMed  Google Scholar 

  28. Russell, D. S. et al. Change in PDE10 across early Huntington disease assessed by [18F]MNI-659 and PET imaging. Neurology 86, 748–754 (2016).

    CAS  PubMed  Google Scholar 

  29. Heckman, P. R. A., Blokland, A., Bollen, E. P. P. & Prickaerts, J. Phosphodiesterase inhibition and modulation of corticostriatal and hippocampal circuits: clinical overview and translational considerations. Neurosci. Biobehav. Rev. 87, 233–254 (2018).

    CAS  PubMed  Google Scholar 

  30. [No authors listed.] Vinpocetine. Monograph. Altern. Med. Rev. 7, 240–243 (2002).

  31. Asal, N. J. & Wojciak, K. A. Effect of cilostazol in treating diabetes-associated microvascular complications. Endocrine 56, 240–244 (2017).

    CAS  PubMed  Google Scholar 

  32. Chapman, T. M. & Goa, K. L. Cilostazol: a review of its use in intermittent claudication. Am. J. Cardiovasc. Drugs 3, 117–138 (2003).

    CAS  PubMed  Google Scholar 

  33. Chong, L. Y. Z., Satya, K., Kim, B. & Berkowitz, R. Milrinone dosing and a culture of caution in clinical practice. Cardiol. Rev. 26, 35–42 (2018).

    PubMed  Google Scholar 

  34. Celli, B. R. Pharmacological therapy of COPD: reasons for optimism. Chest 154, 1404–1415 (2018).

    PubMed  Google Scholar 

  35. Cada, D. J., Ingram, K. & Baker, D. E. Apremilast. Hosp. Pharm. 49, 752–762 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Papp, K. et al. Apremilast, an oral phosphodiesterase 4 (PDE4) inhibitor, in patients with moderate to severe plaque psoriasis: results of a phase III, randomized, controlled trial (efficacy and safety trial evaluating the effects of apremilast in psoriasis [ESTEEM] 1). J. Am. Acad. Dermatol. 73, 37–49 (2015).

    CAS  PubMed  Google Scholar 

  37. Edwards, C. J. et al. Apremilast, an oral phosphodiesterase 4 inhibitor, in patients with psoriatic arthritis and current skin involvement: a phase III, randomised, controlled trial (PALACE 3). Ann. Rheum. Dis. 75, 1065–1073 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Terburg, D. et al. Acute effects of Sceletium tortuosum (Zembrin), a dual 5-HT reuptake and PDE4 inhibitor, in the human amygdala and its connection to the hypothalamus. Neuropsychopharmacology 38, 2708–2716 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Chiu, S. et al. Proof-of-concept randomized controlled study of cognition effects of the proprietary extract Sceletium tortuosum (Zembrin) targeting phosphodiesterase-4 in cognitively healthy subjects: implications for Alzheimer’s dementia. Evid. Based Complement. Alternat. Med. 2014, 682014 (2014).

    PubMed  PubMed Central  Google Scholar 

  40. Campbell, H. E. Clinical monograph for drug formulary review: erectile dysfunction agents. J. Manag. Care Pharm. 11, 151–171 (2005).

    PubMed  Google Scholar 

  41. Cho, M. C. & Paick, J. S. Udenafil for the treatment of erectile dysfunction. Ther. Clin. Risk Manag. 10, 341–354 (2014).

    PubMed  PubMed Central  Google Scholar 

  42. Kang, S. G. & Kim, J. J. Udenafil: efficacy and tolerability in the management of erectile dysfunction. Ther. Adv. Urol. 5, 101–110 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Shim, Y. S. et al. Effects of daily low-dose treatment with phosphodiesterase type 5 inhibitor on cognition, depression, somatization and erectile function in patients with erectile dysfunction: a double-blind, placebo-controlled study. Int. J. Impot. Res. 26, 76–80 (2014).

    CAS  PubMed  Google Scholar 

  44. Shim, Y. S. et al. Effects of repeated dosing with udenafil (Zydena) on cognition, somatization and erection in patients with erectile dysfunction: a pilot study. Int. J. Impot. Res. 23, 109–114 (2011).

    CAS  PubMed  Google Scholar 

  45. Helal, C. J. et al. Identification of a potent, highly selective, and brain penetrant phosphodiesterase 2A Inhibitor clinical candidate. J. Med. Chem. 61, 1001–1018 (2018).

    CAS  PubMed  Google Scholar 

  46. [No authors listed.] Exisulind: aptosyn, FGN 1, prevatac, sulindac sulfone. Drugs R. D. 5, 220–226 (2004).

  47. Griffiths, G. J. Exisulind cell pathways. Curr. Opin. Investig. Drugs 1, 386–391 (2000).

    CAS  PubMed  Google Scholar 

  48. Rosales, R. L., Santos, M. M. & Mercado-Asis, L. B. Cilostazol: a pilot study on safety and clinical efficacy in neuropathies of diabetes mellitus type 2 (ASCEND). Angiology 62, 625–635 (2011).

    CAS  PubMed  Google Scholar 

  49. Giembycz, M. A. An update and appraisal of the cilomilast phase III clinical development programme for chronic obstructive pulmonary disease. Br. J. Clin. Pharmacol. 62, 138–152 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Ratziu, V. et al. Lack of efficacy of an inhibitor of PDE4 in phase 1 and 2 trials of patients with nonalcoholic steatohepatitis. Clin. Gastroenterol. Hepatol. 12, 1724–1730 (2014).

    CAS  PubMed  Google Scholar 

  51. Schultheiss, D. et al. Central effects of sildenafil (Viagra) on auditory selective attention and verbal recognition memory in humans: a study with event-related brain potentials. World J. Urol. 19, 46–50 (2001).

    CAS  PubMed  Google Scholar 

  52. Grass, H. et al. Sildenafil (Viagra): is there an influence on psychological performance? Int. Urol. Nephrol. 32, 409–412 (2001).

    CAS  PubMed  Google Scholar 

  53. Reneerkens, O. A. et al. The effects of the phosphodiesterase type 5 inhibitor vardenafil on cognitive performance in healthy adults: a behavioral-electroencephalography study. J. Psychopharmacol. 27, 600–608 (2013).

    CAS  PubMed  Google Scholar 

  54. Reneerkens, O. A. et al. The PDE5 inhibitor vardenafil does not affect auditory sensory gating in rats and humans. Psychopharmacology 225, 303–312 (2013).

    CAS  PubMed  Google Scholar 

  55. Goff, D. C. et al. A placebo-controlled study of sildenafil effects on cognition in schizophrenia. Psychopharmacology 202, 411–417 (2009).

    CAS  PubMed  Google Scholar 

  56. Akhondzadeh, S. et al. Sildenafil adjunctive therapy to risperidone in the treatment of the negative symptoms of schizophrenia: a double-blind randomized placebo-controlled trial. Psychopharmacology 213, 809–815 (2011).

    CAS  PubMed  Google Scholar 

  57. Nelson, M. D. et al. PDE5 inhibition alleviates functional muscle ischemia in boys with Duchenne muscular dystrophy. Neurology 82, 2085–2091 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Martin, E. A. et al. Tadalafil alleviates muscle ischemia in patients with Becker muscular dystrophy. Sci. Transl Med. 4, 162ra155 (2012).

    PubMed  PubMed Central  Google Scholar 

  59. Schwam, E. M. et al. A multicenter, double-blind, placebo-controlled trial of the PDE9A inhibitor, PF-04447943, in Alzheimer’s disease. Curr. Alzheimer Res. 11, 413–421 (2014). Despite proving target engagement by measuring a change in cerebrospinal fluid cGMP levels, this clinical trial failed to detect any significant behavioural effect of a PDE9 inhibitor in patients with AD.

    CAS  PubMed  Google Scholar 

  60. Boehringer Ingelheim International GmbH. Boehringer Ingelheim refocuses PDE9 inhibition brain research on schizophrenia following results from phase II Alzheimer’s trials. Boehringer Ingelheim https://www.boehringer-ingelheim.com/PDE9-Inhibition-in-AD (2018).

  61. Brown, D. et al. Evaluation of the efficacy, safety, and tolerability of BI 409306, a novel phosphodiesterase 9 inhibitor, in cognitive impairment in schizophrenia: a randomized, double-blind, placebo-controlled, phase II trial. Schizophr. Bull. 45, 350–359 (2018).

    PubMed Central  Google Scholar 

  62. Moschetti, V. et al. The safety, tolerability and pharmacokinetics of BI 409306, a novel and potent PDE9 inhibitor: overview of three phase I randomised trials in healthy volunteers. Eur. Neuropsychopharmacol. 28, 643–655 (2018).

    CAS  PubMed  Google Scholar 

  63. Brown, D., Daniels, K., Pichereau, S., & Sand, M. A. Phase IC study evaluating the safety, tolerability, pharmacokinetics, and cognitive outcomes of BI 409306 in patients with mild-to-moderate schizophrenia. Neurol. Ther. 7, 129–139 (2018).

    PubMed  Google Scholar 

  64. Wunderlich, G. et al. Study design and characteristics of two phase II proof-of-concept clinical trials of the pde9 inhibitor BI 409306 in early Alzheimer’s disease. Alzheimers Dement. 12, P820–P821 (2016).

    Google Scholar 

  65. Grauer, S. M. et al. Phosphodiesterase 10A inhibitor activity in preclinical models of the positive, cognitive, and negative symptoms of schizophrenia. J. Pharmacol. Exp. Ther. 331, 574–590 (2009).

    CAS  PubMed  Google Scholar 

  66. Schmidt, C. J. et al. Preclinical characterization of selective phosphodiesterase 10A inhibitors: a new therapeutic approach to the treatment of schizophrenia. J. Pharmacol. Exp. Ther. 325, 681–690 (2008).

    CAS  PubMed  Google Scholar 

  67. DeMartinis, N. et al. Results of a phase 2A proof-of-concept trial with a PDE10A inhibitor in the treatment of acute exacerbation of schizophrenia. Schizophr. Res. 136, S262 (2012).

    Google Scholar 

  68. Russell, D. S. et al. The phosphodiesterase 10 positron emission tomography tracer, [18F]MNI-659, as a novel biomarker for early Huntington disease. JAMA Neurol. 71, 1520–1528 (2014).

    PubMed  Google Scholar 

  69. Beaumont, V. et al. Phosphodiesterase 10A inhibition improves cortico-basal ganglia function in Huntington’s disease models. Neuron 92, 1220–1237 (2016).

    CAS  PubMed  Google Scholar 

  70. Kelly, M. P. in Encyclopedia of Signaling Molecules (ed. Choi, S.) 3804–3826 (Springer International Publishing, 2018).

  71. Kelly, M. P. et al. Select 3΄,5΄-cyclic nucleotide phosphodiesterases exhibit altered expression in the aged rodent brain. Cell. Signal. 26, 383–397 (2014). This study directly compares expression patterns of all brain-expressed PDEs using both in situ hybridization and PCR in mouse and rat brains.

    CAS  PubMed  Google Scholar 

  72. Raheem, I. T. et al. Discovery of pyrazolopyrimidine phosphodiesterase 10A inhibitors for the treatment of schizophrenia. Bioorg. Med. Chem. Lett. 26, 126–132 (2016).

    CAS  PubMed  Google Scholar 

  73. Burgin, A. B. et al. Design of phosphodiesterase 4D (PDE4D) allosteric modulators for enhancing cognition with improved safety. Nat. Biotechnol. 28, 63–70 (2010). This study presents a rare example of a PDE inhibitor that does not simply compete for the catalytic site.

    CAS  PubMed  Google Scholar 

  74. Hagen, T. J. et al. Discovery of triazines as selective PDE4B versus PDE4D inhibitors. Bioorg. Med. Chem. Lett. 24, 4031–4034 (2014). This study is an important example of how knowledge of biology can drive more sophisticated medicinal chemistry efforts that end up achieving more selective pharmacological targeting of a given PDE.

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Pushpakom, S. et al. Drug repurposing: progress, challenges and recommendations. Nat. Rev. Drug Discov. 18, 41–58 (2018).

    PubMed  Google Scholar 

  76. Ledeboer, A., Hutchinson, M. R., Watkins, L. R. & Johnson, K. W. Ibudilast (AV-411). A new class therapeutic candidate for neuropathic pain and opioid withdrawal syndromes. Expert Opin. Investig. Drugs 16, 935–950 (2007).

    CAS  PubMed  Google Scholar 

  77. Schwenkgrub, J., Zaremba, M., Mirowska-Guzel, D. & Kurkowska-Jastrzebska, I. Ibudilast: a nonselective phosphodiesterase inhibitor in brain disorders. Postepy Hig. Med. Dosw. 71, 137–148 (2017).

    Google Scholar 

  78. DeYoung, D. Z. et al. Safety of intravenous methamphetamine administration during ibudilast treatment. J. Clin. Psychopharmacol. 36, 347–354 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Worley, M. J., Swanson, A. N., Heinzerling, K. G., Roche, D. J. & Shoptaw, S. Ibudilast attenuates subjective effects of methamphetamine in a placebo-controlled inpatient study. Drug Alcohol Depend. 162, 245–250 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  80. Cooper, Z. D. et al. Effects of ibudilast on oxycodone-induced analgesia and subjective effects in opioid-dependent volunteers. Drug Alcohol Depend. 178, 340–347 (2017).

    CAS  PubMed  Google Scholar 

  81. Ray, L. A. et al. Development of the neuroimmune modulator ibudilast for the treatment of alcoholism: a randomized, placebo-controlled, human laboratory trial. Neuropsychopharmacology 42, 1776–1788 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Snyder, G. L. et al. Preclinical profile of ITI-214, an inhibitor of phosphodiesterase 1, for enhancement of memory performance in rats. Psychopharmacology 233, 3113–3124 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Li, P. et al. Discovery of potent and selective inhibitors of phosphodiesterase 1 for the treatment of cognitive impairment associated with neurodegenerative and neuropsychiatric diseases. J. Med. Chem. 59, 1149–1164 (2016).

    CAS  PubMed  Google Scholar 

  84. Pekcec, A. et al. Targeting the dopamine D1 receptor or its downstream signalling by inhibiting phosphodiesterase-1 improves cognitive performance. Br. J. Pharmacol. 175, 3021–3033 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Dyck, B. et al. Discovery of selective phosphodiesterase 1 inhibitors with memory enhancing properties. J. Med. Chem. 60, 3472–3483 (2017).

    CAS  PubMed  Google Scholar 

  86. Hashimoto, T. et al. Acute enhancement of cardiac function by phosphodiesterase type 1 inhibition — translational study in the dog and rabbit. Circulation 138, 1974–1987 (2018).

    CAS  PubMed  Google Scholar 

  87. Zhang, C., Lueptow, L. M., Zhang, H. T., O’Donnell, J. M. & Xu, Y. The role of phosphodiesterase-2 in psychiatric and neurodegenerative disorders. Adv. Neurobiol. 17, 307–347 (2017).

    PubMed  Google Scholar 

  88. Mikami, S. et al. Discovery of a novel series of pyrazolo[1,5-a]pyrimidine-based phosphodiesterase 2A inhibitors structurally different from N-((1S)-1-(3-fluoro-4-(trifluoromethoxy)phenyl)-2-methoxyethyl)-7-methoxy-2-oxo-2,3-dihydropyrido[2,3-b]pyrazine-4(1H)-carboxamide (TAK-915), for the treatment of cognitive disorders. Chem. Pharm. Bull. 65, 1058–1077 (2017).

    CAS  PubMed  Google Scholar 

  89. Mikami, S. et al. Discovery of clinical candidate N-((1S)-1-(3-fluoro-4-(trifluoromethoxy)phenyl)-2-methoxyethyl)-7-methoxy-2-oxo-2,3-dihydropyrido[2,3-b]pyrazine-4(1H)-carboxamide (TAK-915): a highly potent, selective, and brain-penetrating phosphodiesterase 2A inhibitor for the treatment of cognitive disorders. J. Med. Chem. 60, 7677–7702 (2017).

    CAS  PubMed  Google Scholar 

  90. Mikami, S. et al. Discovery of an orally bioavailable, brain-penetrating, in vivo active phosphodiesterase 2A inhibitor lead series for the treatment of cognitive disorders. J. Med. Chem. 60, 7658–7676 (2017).

    CAS  PubMed  Google Scholar 

  91. Weber, S. et al. PDE2 at the crossway between cAMP and cGMP signalling in the heart. Cell. Signal. 38, 76–84 (2017).

    CAS  PubMed  Google Scholar 

  92. Zhang, C. et al. The roles of phosphodiesterase 2 in the central nervous and peripheral systems. Curr. Pharm. Des. 21, 274–290 (2015).

    CAS  PubMed  Google Scholar 

  93. Bundhun, P. K., Qin, T. & Chen, M. H. Comparing the effectiveness and safety between triple antiplatelet therapy and dual antiplatelet therapy in type 2 diabetes mellitus patients after coronary stents implantation: a systematic review and meta-analysis of randomized controlled trials. BMC Cardiovasc. Disord. 15, 118 (2015).

    PubMed  PubMed Central  Google Scholar 

  94. Neel, J. D., Kruse, R. L., Dombrovskiy, V. Y. & Vogel, T. R. Cilostazol and freedom from amputation after lower extremity revascularization. J. Vasc. Surg. 61, 960–964 (2015).

    PubMed  Google Scholar 

  95. Lim, H. W. et al. Effect of a 4-week treatment with cilostazol in patients with chronic tinnitus: a randomized, prospective, placebo-controlled, double-blind, pilot study. J. Int. Adv. Otol. 12, 170–176 (2016).

    PubMed  Google Scholar 

  96. Lakics, V., Karran, E. H. & Boess, F. G. Quantitative comparison of phosphodiesterase mRNA distribution in human brain and peripheral tissues. Neuropharmacology 59, 367–374 (2010). This work presents a head-to-head comparison of PDE expression patterns in various human tissues.

    CAS  PubMed  Google Scholar 

  97. Mochizuki, Y., Oishi, M. & Mizutani, T. Effects of cilostazol on cerebral blood flow, P300, and serum lipid levels in the chronic stage of cerebral infarction. J. Stroke Cerebrovasc. Dis. 10, 63–69 (2001).

    CAS  PubMed  Google Scholar 

  98. Jung, K. I., Kim, J. H., Park, H. Y. & Park, C. K. Neuroprotective effects of cilostazol on retinal ganglion cell damage in diabetic rats. J. Pharmacol. Exp. Ther. 345, 457–463 (2013).

    CAS  PubMed  Google Scholar 

  99. Masciarelli, S. et al. Cyclic nucleotide phosphodiesterase 3A-deficient mice as a model of female infertility. J. Clin. Invest. 114, 196–205 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  100. Taiyeb, A. M. et al. Cilostazol blocks pregnancy in naturally cycling swine: an animal model. Life Sci. 142, 92–96 (2015).

    CAS  PubMed  Google Scholar 

  101. Taiyeb, A. M., Muhsen-Alanssari, S. A., Dees, W. L., Ridha-Albarzanchi, M. T. & Kraemer, D. C. Improvement in in vitro fertilization outcome following in vivo synchronization of oocyte maturation in mice. Exp. Biol. Med. 240, 519–526 (2015).

    CAS  Google Scholar 

  102. Duan, L. M., Yu, H. Y., Li, Y. L. & Jia, C. J. Design and discovery of 2-(4-(1H-tetrazol-5-yl)-1H-pyrazol-1-yl)-4-(4-phenyl)thiazole derivatives as cardiotonic agents via inhibition of PDE3. Bioorg. Med. Chem. 23, 6111–6117 (2015).

    CAS  PubMed  Google Scholar 

  103. Kim, K. Y., Lee, H., Yoo, S. E., Kim, S. H. & Kang, N. S. Discovery of new inhibitor for PDE3 by virtual screening. Bioorg. Med. Chem. Lett. 21, 1617–1620 (2011).

    CAS  PubMed  Google Scholar 

  104. Nikpour, M. et al. Design, synthesis and biological evaluation of 6-(benzyloxy)-4-methylquinolin-2(1H)-one derivatives as PDE3 inhibitors. Bioorg. Med. Chem. 18, 855–862 (2010).

    CAS  PubMed  Google Scholar 

  105. Huff, S. B. & Gottwald, L. D. Repigmentation of tenacious vitiligo on apremilast. Case Rep. Dermatol. Med. 2017, 2386234 (2017).

    PubMed  PubMed Central  Google Scholar 

  106. AbuHilal, M., Walsh, S. & Shear, N. Treatment of recalcitrant erosive oral lichen planus and desquamative gingivitis with oral apremilast. J. Dermatol. Case Rep. 10, 56–57 (2016).

    PubMed  PubMed Central  Google Scholar 

  107. Hafner, J. et al. Apremilast is effective in lichen planus mucosae-associated stenotic esophagitis. Case Rep. Dermatol. 8, 224–226 (2016).

    PubMed  PubMed Central  Google Scholar 

  108. Bettencourt, M. Oral lichen planus treated with apremilast. J. Drugs Dermatol. 15, 1026–1028 (2016).

    PubMed  Google Scholar 

  109. Jensterle, M., Kocjan, T. & Janez, A. Phosphodiesterase 4 inhibition as a potential new therapeutic target in obese women with polycystic ovary syndrome. J. Clin. Endocrinol. Metab. 99, E1476–E1481 (2014).

    CAS  PubMed  Google Scholar 

  110. Jensterle, M., Salamun, V., Kocjan, T., Vrtacnik Bokal, E. & Janez, A. Short term monotherapy with GLP-1 receptor agonist liraglutide or PDE 4 inhibitor roflumilast is superior to metformin in weight loss in obese PCOS women: a pilot randomized study. J. Ovarian Res. 8, 32 (2015).

    PubMed  PubMed Central  Google Scholar 

  111. Plock, N., Vollert, S., Mayer, M., Hanauer, G. & Lahu, G. Pharmacokinetic/pharmacodynamic modeling of the PDE4 inhibitor TAK-648 in type 2 diabetes: early translational approaches for human dose prediction. Clin. Transl Sci. 10, 185–193 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  112. Van Duinen, M. A. et al. Acute administration of roflumilast enhances immediate recall of verbal word memory in healthy young adults. Neuropharmacology 131, 31–38 (2018). This work is one of the first studies to demonstrate a pro-cognitive effect in humans of a commercially available PDE4 inhibitor.

    PubMed  Google Scholar 

  113. Gilleen, J. et al. An experimental medicine study of the phosphodiesterase-4 inhibitor, roflumilast, on working memory-related brain activity and episodic memory in schizophrenia patients. Psychopharmacology https://doi.org/10.1007/s00213-018-5134-y (2018).

    Article  PubMed  Google Scholar 

  114. Blokland, A. et al. Acute treatment with the PDE4 inhibitor roflumilast improves verbal word memory in healthy old individuals: a double-blind placebo-controlled study. Neurobiol. Aging 77, 37–43 (2019).

    CAS  PubMed  Google Scholar 

  115. Kanes, S. J. et al. Rolipram: a specific phosphodiesterase 4 inhibitor with potential antipsychotic activity. Neuroscience 144, 239–246 (2007).

    CAS  PubMed  Google Scholar 

  116. Kelly, M. P. et al. Constitutive activation of Gαs within forebrain neurons causes deficits in sensorimotor gating because of PKA-dependent decreases in cAMP. Neuropsychopharmacology 32, 577–588 (2007). This work is an important study showing that chronic signalling through Gα s will lead to PKA-dependent compensatory increases in PDE activity in some brain regions but not others.

    CAS  PubMed  Google Scholar 

  117. Kelly, M. P. et al. Developmental etiology for neuroanatomical and cognitive deficits in mice overexpressing Gαs, a G-protein subunit genetically linked to schizophrenia. Mol. Psychiatry 14, 398–415 (2009).

    CAS  PubMed  Google Scholar 

  118. Rodefer, J. S., Saland, S. K. & Eckrich, S. J. Selective phosphodiesterase inhibitors improve performance on the ED/ID cognitive task in rats. Neuropharmacology 62, 1182–1190 (2012).

    CAS  PubMed  Google Scholar 

  119. Helicon Therapeutics, Inc. Helicon study of HT-0712 on primate memory formation reveals significant enhancement. Dart Neuroscience http://dartneuroscience.com/press_releases/july_22_2008.pdf (2008).

  120. MacDonald, E. et al. A novel phosphodiesterase type 4 inhibitor, HT-0712, enhances rehabilitation-dependent motor recovery and cortical reorganization after focal cortical ischemia. Neurorehabil. Neural Repair 21, 486–496 (2007).

    PubMed  Google Scholar 

  121. Peters, M. et al. The PDE4 inhibitor HT-0712 improves hippocampus-dependent memory in aged mice. Neuropsychopharmacology 39, 2938–2948 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  122. Tetra Discovery Partners. Tetra Discovery Partners announces positive results from phase 1 studies of cognition drug candidate, BPN14770. Tetra Discovery http://tetradiscovery.com/wp-content/uploads/2016/11/FINAL-Tetra-Phase-1-121616-FINAL.pdf (2016).

  123. Gurney, M. E., Cogram, P., Deacon, R. M., Rex, C. & Tranfaglia, M. Multiple behavior phenotypes of the fragile-X syndrome mouse model respond to chronic inhibition of phosphodiesterase-4D (PDE4D). Sci. Rep. 7, 14653 (2017).

    PubMed  PubMed Central  Google Scholar 

  124. Zhang, C. et al. Memory enhancing effects of BPN14770, an allosteric inhibitor of phosphodiesterase-4D, in wild-type and humanized mice. Neuropsychopharmacology 43, 2299–2309 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  125. Rutter, A. R. et al. GSK356278, a potent, selective, brain-penetrant phosphodiesterase 4 inhibitor that demonstrates anxiolytic and cognition-enhancing effects without inducing side effects in preclinical species. J. Pharmacol. Exp. Ther. 350, 153–163 (2014).

    PubMed  Google Scholar 

  126. Munshi, A. & Das, S. Genetic understanding of stroke treatment: potential role for phosphodiesterase inhibitors. Adv. Neurobiol. 17, 445–461 (2017).

    PubMed  Google Scholar 

  127. Chen, J. et al. The phosphodiesterase-4 inhibitor, FCPR16, attenuates ischemia–reperfusion injury in rats subjected to middle cerebral artery occlusion and reperfusion. Brain Res. Bull. 137, 98–106 (2018).

    CAS  PubMed  Google Scholar 

  128. Soares, L. M. et al. Rolipram improves cognition, reduces anxiety- and despair-like behaviors and impacts hippocampal neuroplasticity after transient global cerebral ischemia. Neuroscience 326, 69–83 (2016).

    CAS  PubMed  Google Scholar 

  129. Li, L. X. et al. Prevention of cerebral ischemia-induced memory deficits by inhibition of phosphodiesterase-4 in rats. Metab. Brain Dis. 26, 37–47 (2011).

    PubMed  Google Scholar 

  130. Titus, D. J. et al. A negative allosteric modulator of PDE4D enhances learning after traumatic brain injury. Neurobiol. Learn. Mem. 148, 38–49 (2018).

    CAS  PubMed  Google Scholar 

  131. Knott, E. P., Assi, M., Rao, S. N., Ghosh, M. & Pearse, D. D. Phosphodiesterase inhibitors as a therapeutic approach to neuroprotection and repair. Int. J. Mol. Sci. 18, E696 (2017).

    PubMed  Google Scholar 

  132. Olsen, C. M. & Liu, Q. S. Phosphodiesterase 4 inhibitors and drugs of abuse: current knowledge and therapeutic opportunities. Front. Biol. 11, 376–386 (2016).

    CAS  Google Scholar 

  133. Logrip, M. L. Phosphodiesterase regulation of alcohol drinking in rodents. Alcohol 49, 795–802 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  134. Avila, D. V. et al. Dysregulation of hepatic cAMP levels via altered Pde4b expression plays a critical role in alcohol-induced steatosis. J. Pathol. 240, 96–107 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  135. Avila, D. V. et al. Phosphodiesterase 4b expression plays a major role in alcohol-induced neuro-inflammation. Neuropharmacology 125, 376–385 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  136. Favilla, C., Abel, T. & Kelly, M. P. Chronic Gαs signaling in the striatum increases anxiety-related behaviors independent of developmental effects. J. Neurosci. 28, 13952–13956 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  137. Kelly, M. P. et al. Constitutive activation of the G-protein subunit Gαs within forebrain neurons causes PKA-dependent alterations in fear conditioning and cortical Arc mRNA expression. Learn. Mem. 15, 75–83 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  138. Cooney, J. D. & Aguiar, R. C. Phosphodiesterase 4 inhibitors have wide-ranging activity in B cell malignancies. Blood 128, 2886–2890 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  139. Wang, W. et al. Triple negative breast cancer development can be selectively suppressed by sustaining an elevated level of cellular cyclic AMP through simultaneously blocking its efflux and decomposition. Oncotarget 7, 87232–87245 (2016).

    PubMed  PubMed Central  Google Scholar 

  140. Nishi, K. et al. Apremilast induces apoptosis of human colorectal cancer cells with mutant KRAS. Anticancer Res. 37, 3833–3839 (2017).

    CAS  PubMed  Google Scholar 

  141. Mishra, R. R. et al. Reactivation of cAMP pathway by PDE4D inhibition represents a novel druggable axis for overcoming tamoxifen resistance in ER-positive breast cancer. Clin. Cancer Res. 24, 1987–2001 (2018).

    CAS  PubMed  Google Scholar 

  142. Wortsman, X., Del Barrio-Diaz, P., Meza-Romero, R., Poehls-Risco, C. & Vera-Kellet, C. Nifedipine cream versus sildenafil cream for patients with secondary Raynaud phenomenon: a randomized, double-blind, controlled pilot study. J. Am. Acad. Dermatol. 78, 189–190 (2018).

    PubMed  Google Scholar 

  143. Maged, M., Wageh, A., Shams, M. & Elmetwally, A. Use of sildenafil citrate in cases of intrauterine growth restriction (IUGR); a prospective trial. Taiwan J. Obstet. Gynecol. 57, 483–486 (2018).

    PubMed  Google Scholar 

  144. Pels, A. et al. STRIDER (Sildenafil TheRapy in dismal prognosis early onset fetal growth restriction): an international consortium of randomised placebo-controlled trials. BMC Pregnancy Childbirth 17, 440 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  145. El-Sayed, M. A., Saleh, S. A., Maher, M. A. & Khidre, A. M. Utero-placental perfusion Doppler indices in growth restricted fetuses: effect of sildenafil citrate. J. Matern. Fetal Neonatal Med. 31, 1045–1050 (2018).

    PubMed  Google Scholar 

  146. Paauw, N. D. et al. Sildenafil during pregnancy: a preclinical meta-analysis on fetal growth and maternal blood pressure. Hypertension 70, 998–1006 (2017).

    CAS  PubMed  Google Scholar 

  147. Aversa, A. et al. Tadalafil improves lean mass and endothelial function in nonobese men with mild ED/LUTS: in vivo and in vitro characterization. Endocrine 56, 639–648 (2017).

    CAS  PubMed  Google Scholar 

  148. Giannetta, E. et al. Chronic inhibition of cGMP phosphodiesterase 5A improves diabetic cardiomyopathy: a randomized, controlled clinical trial using magnetic resonance imaging with myocardial tagging. Circulation 125, 2323–2333 (2012).

    CAS  PubMed  Google Scholar 

  149. Di Luigi, L. et al. Phosphodiesterase type 5 inhibitor sildenafil decreases the proinflammatory chemokine CXCL10 in human cardiomyocytes and in subjects with diabetic cardiomyopathy. Inflammation 39, 1238–1252 (2016).

    PubMed  PubMed Central  Google Scholar 

  150. Fiore, D. et al. PDE5 inhibition ameliorates visceral adiposity targeting the miR-22/SIRT1 pathway: evidence from the CECSID trial. J. Clin. Endocrinol. Metab. 101, 1525–1534 (2016).

    CAS  PubMed  Google Scholar 

  151. Mandosi, E. et al. Endothelial dysfunction markers as a therapeutic target for sildenafil treatment and effects on metabolic control in type 2 diabetes. Expert Opin. Ther. Targets 19, 1617–1622 (2015).

    CAS  PubMed  Google Scholar 

  152. Santi, D. et al. Could chronic vardenafil administration influence the cardiovascular risk in men with type 2 diabetes mellitus? PLOS ONE 13, e0199299 (2018).

    PubMed  PubMed Central  Google Scholar 

  153. Montes Cardona, C. E. & Garcia-Perdomo, H. A. Efficacy of phosphodiesterase type 5 inhibitors for the treatment of distal ureteral calculi: a systematic review and meta-analysis. Investig. Clin. Urol. 58, 82–89 (2017).

    PubMed  PubMed Central  Google Scholar 

  154. Booth, L. et al. Neratinib augments the lethality of [regorafenib + sildenafil]. J. Cell. Physiol. 234, 4874–4887 (2018).

    PubMed  Google Scholar 

  155. Islam, B. N. et al. Sildenafil suppresses inflammation-driven colorectal cancer in mice. Cancer Prev. Res. 10, 377–388 (2017).

    CAS  Google Scholar 

  156. Califano, J. A. et al. Tadalafil augments tumor specific immunity in patients with head and neck squamous cell carcinoma. Clin. Cancer Res. 21, 30–38 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  157. Weed, D. T. et al. Tadalafil reduces myeloid-derived suppressor cells and regulatory T cells and promotes tumor immunity in patients with head and neck squamous cell carcinoma. Clin. Cancer Res. 21, 39–48 (2015).

    CAS  PubMed  Google Scholar 

  158. Charych, E. I., Jiang, L. X., Lo, F., Sullivan, K. & Brandon, N. J. Interplay of palmitoylation and phosphorylation in the trafficking and localization of phosphodiesterase 10A: implications for the treatment of schizophrenia. J. Neurosci. 30, 9027–9037 (2010). This study presents a key finding showing that two PTMs can compete to regulate the subcellular compartmentalization of a PDE.

    CAS  PubMed  PubMed Central  Google Scholar 

  159. Lee, K. et al. β-catenin nuclear translocation in colorectal cancer cells is suppressed by PDE10A inhibition, cGMP elevation, and activation of PKG. Oncotarget 7, 5353–5365 (2016).

    PubMed  Google Scholar 

  160. Li, N. et al. Phosphodiesterase 10A: a novel target for selective inhibition of colon tumor cell growth and beta-catenin-dependent TCF transcriptional activity. Oncogene 34, 1499–1509 (2015).

    CAS  PubMed  Google Scholar 

  161. Barone, I., Giordano, C., Bonofiglio, D., Ando, S. & Catalano, S. Phosphodiesterase type 5 and cancers: progress and challenges. Oncotarget 8, 99179–99202 (2017).

    PubMed  PubMed Central  Google Scholar 

  162. Aoun, F. et al. Association between phosphodiesterase type 5 inhibitors and prostate cancer: a systematic review. Prog. Urol. 28, 560–566 (2018).

    PubMed  Google Scholar 

  163. Arozarena, I. et al. Oncogenic BRAF induces melanoma cell invasion by downregulating the cGMP-specific phosphodiesterase PDE5A. Cancer Cell 19, 45–57 (2011).

    CAS  PubMed  Google Scholar 

  164. Feng, S. et al. Are phosphodiesterase type 5 inhibitors associated with increased risk of melanoma? A systematic review and meta-analysis. Medicine 97, e9601 (2018). This study presents an important reminder that attention must be paid not only to acute side-effect profiles, but also long-term risks of PDE-inhibitor treatment.

    CAS  PubMed  PubMed Central  Google Scholar 

  165. Almeida, C. B. et al. Hydroxyurea and a cGMP-amplifying agent have immediate benefits on acute vaso-occlusive events in sickle cell disease mice. Blood 120, 2879–2888 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  166. Goldsmith, P. et al. A randomized multiple dose pharmacokinetic study of a novel PDE10A inhibitor TAK-063 in subjects with stable schizophrenia and Japanese subjects and modeling of exposure relationships to adverse events. Drugs R. D. 17, 631–643 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  167. Macek, T. A. et al. A phase 2, randomized, placebo-controlled study of the efficacy and safety of TAK-063 in subjects with an acute exacerbation of schizophrenia. Schizophr. Res. 204, 289–294 (2019).

    PubMed  Google Scholar 

  168. Suzuki, K., Harada, A., Suzuki, H., Miyamoto, M. & Kimura, H. TAK-063, a PDE10A inhibitor with balanced activation of direct and indirect pathways, provides potent antipsychotic-like effects in multiple paradigms. Neuropsychopharmacology 41, 2252–2262 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  169. Wilson, J. M. et al. Phosphodiesterase 10A inhibitor, MP-10 (PF-2545920), produces greater induction of c-Fos in dopamine D2 neurons than in D1 neurons in the neostriatum. Neuropharmacology 99, 379–386 (2015).

    CAS  PubMed  Google Scholar 

  170. Padovan-Neto, F. E. & West, A. R. Regulation of striatal neuron activity by cyclic nucleotide signaling and phosphodiesterase inhibition: implications for the treatment of Parkinson’s disease. Adv. Neurobiol. 17, 257–283 (2017).

    PubMed  Google Scholar 

  171. Jankowska, A., Swierczek, A., Chlon-Rzepa, G., Pawlowski, M. & Wyska, E. PDE7-selective and dual inhibitors: advances in chemical and biological research. Curr. Med. Chem. 24, 673–700 (2017).

    CAS  PubMed  Google Scholar 

  172. Redondo, M. et al. Effect of phosphodiesterase 7 (PDE7) inhibitors in experimental autoimmune encephalomyelitis mice. Discovery of a new chemically diverse family of compounds. J. Med. Chem. 55, 3274–3284 (2012).

    CAS  PubMed  Google Scholar 

  173. Morales-Garcia, J. A. et al. Silencing phosphodiesterase 7B gene by lentiviral-shRNA interference attenuates neurodegeneration and motor deficits in hemiparkinsonian mice. Neurobiol. Aging 36, 1160–1173 (2015).

    CAS  PubMed  Google Scholar 

  174. Morales-Garcia, J. A. et al. Phosphodiesterase7 inhibition activates adult neurogenesis in hippocampus and subventricular zone in vitro and in vivo. Stem Cells 35, 458–472 (2017).

    CAS  PubMed  Google Scholar 

  175. Tsai, L. C. et al. Inactivation of Pde8b enhances memory, motor performance, and protects against age-induced motor coordination decay. Genes Brain Behav. 11, 837–847 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  176. Pilarzyk, K. et al. Loss of function of phosphodiesterase 11A4 shows that recent and remote long term memories can be uncoupled. Curr. Biol. https://doi.org/10.1016/j.cub.2019.06.018 (2019).

    PubMed  Google Scholar 

  177. Pathak, G. et al. PDE11A negatively regulates lithium responsivity. Mol. Psychiatry 22, 1714–1724 (2017). This study not only identifies PDE homodimerization as a molecular mechanism regulating PDE protein stability and subcellular compartmentalization but also provides proof of principle for how homodimerization could be therapeutically targeted using a biologic.

    CAS  PubMed  Google Scholar 

  178. Mertens, J. et al. Differential responses to lithium in hyperexcitable neurons from patients with bipolar disorder. Nature 527, 95–99 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  179. Dong, H., Zitt, C., Auriga, C., Hatzelmann, A. & Epstein, P. M. Inhibition of PDE3, PDE4 and PDE7 potentiates glucocorticoid-induced apoptosis and overcomes glucocorticoid resistance in CEM T leukemic cells. Biochem. Pharmacol. 79, 321–329 (2010).

    CAS  PubMed  Google Scholar 

  180. de Medeiros, A. S. et al. Identification and characterization of a potent and biologically-active PDE4/7 inhibitor via fission yeast-based assays. Cell. Signal. 40, 73–80 (2017). This study reports a novel yeast-based assay for screening PDE modulators, which not only reads out changes in PDE activity but also proves compounds are cell permeable, are chemically stable and do not exhibit widespread protein binding.

    PubMed  PubMed Central  Google Scholar 

  181. Tsai, L. C., Shimizu-Albergine, M. & Beavo, J. A. The high-affinity cAMP-specific phosphodiesterase 8B controls steroidogenesis in the mouse adrenal gland. Mol. Pharmacol. 79, 639–648 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  182. Shimizu-Albergine, M., Tsai, L. C., Patrucco, E. & Beavo, J. A. cAMP-specific phosphodiesterases 8A and 8B, essential regulators of Leydig cell steroidogenesis. Mol. Pharmacol. 81, 556–566 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  183. Jang, I. S. et al. Lysophosphatidic acid-induced changes in cAMP profiles in young and senescent human fibroblasts as a clue to the ageing process. Mech. Ageing Dev. 127, 481–489 (2006).

    CAS  PubMed  Google Scholar 

  184. Ramos, B. P. et al. Dysregulation of protein kinase a signaling in the aged prefrontal cortex: new strategy for treating age-related cognitive decline. Neuron 40, 835–845 (2003).

    CAS  PubMed  Google Scholar 

  185. Giralt, A. et al. Increased PKA signaling disrupts recognition memory and spatial memory: role in Huntington’s disease. Hum. Mol. Genet. 20, 4232–4247 (2011).

    CAS  PubMed  Google Scholar 

  186. Hegde, S. et al. PDE11A regulates social behaviors and is a key mechanism by which social experience sculpts the brain. Neuroscience 335, 151–169 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  187. Guo, S., Olesen, J. & Ashina, M. Phosphodiesterase 3 inhibitor cilostazol induces migraine-like attacks via cyclic AMP increase. Brain 137, 2951–2959 (2014).

    PubMed  Google Scholar 

  188. Holland, P. R. & Strother, L. Cilostazol as a chemically induced preclinical model of migraine. Cephalalgia 38, 415–416 (2018).

    PubMed  Google Scholar 

  189. Khan, S., Deen, M., Hougaard, A., Amin, F. M. & Ashina, M. Reproducibility of migraine-like attacks induced by phosphodiesterase-3-inhibitor cilostazol. Cephalalgia 38, 892–903 (2018).

    PubMed  Google Scholar 

  190. Hansen, E. K., Guo, S., Ashina, M. & Olesen, J. Toward a pragmatic migraine model for drug testing: I. Cilostazol in healthy volunteers. Cephalalgia 36, 172–178 (2016).

    PubMed  Google Scholar 

  191. Birk, S., Kruuse, C., Petersen, K. A., Tfelt-Hansen, P. & Olesen, J. The headache-inducing effect of cilostazol in human volunteers. Cephalalgia 26, 1304–1309 (2006).

    CAS  PubMed  Google Scholar 

  192. Mowat, F. M. et al. Gene therapy in a large animal model of PDE6A-retinitis pigmentosa. Front. Neurosci. 11, 342 (2017).

    PubMed  PubMed Central  Google Scholar 

  193. Henderson, D. J. et al. The cAMP phosphodiesterase-4D7 (PDE4D7) is downregulated in androgen-independent prostate cancer cells and mediates proliferation by compartmentalising cAMP at the plasma membrane of VCaP prostate cancer cells. Br. J. Cancer 110, 1278–1287 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  194. Abi-Gerges, A. et al. Decreased expression and activity of cAMP phosphodiesterases in cardiac hypertrophy and its impact on beta-adrenergic cAMP signals. Circ. Res. 105, 784–792 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  195. Wang, X., Ward, C. J., Harris, P. C. & Torres, V. E. Cyclic nucleotide signaling in polycystic kidney disease. Kidney Int. 77, 129–140 (2010).

    CAS  PubMed  Google Scholar 

  196. Malekshahabi, T., Khoshdel Rad, N., Serra, A. L. & Moghadasali, R. Autosomal dominant polycystic kidney disease: disrupted pathways and potential therapeutic interventions. J. Cell. Physiol. 234, 12451–12470 (2019).

    CAS  PubMed  Google Scholar 

  197. Zoraghi, R., Corbin, J. D. & Francis, S. H. Properties and functions of GAF domains in cyclic nucleotide phosphodiesterases and other proteins. Mol. Pharmacol. 65, 267–278 (2004).

    CAS  PubMed  Google Scholar 

  198. Schultz, J. E. Structural and biochemical aspects of tandem GAF domains. Handb. Exp. Pharmacol. 191, 93–109 (2009).

    Google Scholar 

  199. Francis, S. H., Blount, M. A. & Corbin, J. D. Mammalian cyclic nucleotide phosphodiesterases: molecular mechanisms and physiological functions. Physiol. Rev. 91, 651–690 (2011).

    CAS  PubMed  Google Scholar 

  200. Jager, R., Schwede, F., Genieser, H. G., Koesling, D. & Russwurm, M. Activation of PDE2 and PDE5 by specific GAF ligands: delayed activation of PDE5. Br. J. Pharmacol. 161, 1645–1660 (2010).

    PubMed  PubMed Central  Google Scholar 

  201. Martinez, S. E. et al. The two GAF domains in phosphodiesterase 2A have distinct roles in dimerization and in cGMP binding. Proc. Natl Acad. Sci. USA 99, 13260–13265 (2002).

    CAS  PubMed  Google Scholar 

  202. Beavo, J. A., Hardman, J. G. & Sutherland, E. W. Stimulation of adenosine 3΄,5΄-monophosphate hydrolysis by guanosine 3΄,5΄-monophosphate. J. Biol. Chem. 246, 3841–3846 (1971).

    CAS  PubMed  Google Scholar 

  203. Jager, R. et al. Activation of PDE10 and PDE11 phosphodiesterases. J. Biol. Chem. 287, 1210–1219 (2012). This study is the first demonstration that PDEs can be pharmacologically activated by a molecule other than cAMP or cGMP itself (that is, PDE11A can be activated by a cGMP analogue binding the GAF-A domain).

    PubMed  Google Scholar 

  204. D’Amours, M. R. & Cote, R. H. Regulation of photoreceptor phosphodiesterase catalysis by its non-catalytic cGMP-binding sites. Biochem. J. 340, 863–869 (1999).

    PubMed  PubMed Central  Google Scholar 

  205. Schultz, J. E., Dunkern, T., Gawlitta-Gorka, E. & Sorg, G. The GAF-tandem domain of phosphodiesterase 5 as a potential drug target. Handb. Exp. Pharmacol. 204, 151–166 (2011).

    CAS  Google Scholar 

  206. Kelly, M. P. Does phosphodiesterase 11A (PDE11A) hold promise as a future therapeutic target? Curr. Pharm. Des. 21, 389–416 (2015). This review introduces the unique idea that it may be possible to therapeutically target dual-specificity PDEs in a functionally selective manner (that is, target only their cAMP or cGMP hydrolytic activity).

    CAS  PubMed  Google Scholar 

  207. Murthy, K. S., Zhou, H. & Makhlouf, G. M. PKA-dependent activation of PDE3A and PDE4 and inhibition of adenylyl cyclase V/VI in smooth muscle. Am. J. Physiol. Cell Physiol. 282, C508–C517 (2002).

    CAS  PubMed  Google Scholar 

  208. Manganiello, V. C., Taira, M., Degerman, E. & Belfrage, P. Type III cGMP-inhibited cyclic nucleotide phosphodiesterases (PDE3 gene family). Cell. Signal. 7, 445–455 (1995).

    CAS  PubMed  Google Scholar 

  209. MacKenzie, S. J. et al. Long PDE4 cAMP specific phosphodiesterases are activated by protein kinase A-mediated phosphorylation of a single serine residue in upstream conserved region 1 (UCR1). Br. J. Pharmacol. 136, 421–433 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  210. Corbin, J. D., Turko, I. V., Beasley, A. & Francis, S. H. Phosphorylation of phosphodiesterase-5 by cyclic nucleotide-dependent protein kinase alters its catalytic and allosteric cGMP-binding activities. Eur. J. Biochem. 267, 2760–2767 (2000). This work is one of the first studies to identify phosphorylation as a PTM capable of changing PDE catalytic activity.

    CAS  PubMed  Google Scholar 

  211. Brown, K. M., Lee, L. C., Findlay, J. E., Day, J. P. & Baillie, G. S. Cyclic AMP-specific phosphodiesterase, PDE8A1, is activated by protein kinase A-mediated phosphorylation. FEBS Lett. 586, 1631–1637 (2012).

    CAS  PubMed  Google Scholar 

  212. Jacobitz, S., McLaughlin, M. M., Livi, G. P., Burman, M. & Torphy, T. J. Mapping the functional domains of human recombinant phosphodiesterase 4A: structural requirements for catalytic activity and rolipram binding. Mol. Pharmacol. 50, 891–899 (1996).

    CAS  PubMed  Google Scholar 

  213. Marcoz, P., Nemoz, G., Prigent, A. F. & Lagarde, M. Phosphatidic acid stimulates the rolipram-sensitive cyclic nucleotide phosphodiesterase from rat thymocytes. Biochim. Biophys. Acta 1176, 129–136 (1993).

    CAS  PubMed  Google Scholar 

  214. Norambuena, A. et al. Phosphatidic acid induces ligand-independent epidermal growth factor receptor endocytic traffic through PDE4 activation. Mol. Biol. Cell 21, 2916–2929 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  215. Grange, M. et al. The cAMP-specific phosphodiesterase PDE4D3 is regulated by phosphatidic acid binding. Consequences for cAMP signaling pathway and characterization of a phosphatidic acid binding site. J. Biol. Chem. 275, 33379–33387 (2000).

    CAS  PubMed  Google Scholar 

  216. Wang, L. et al. UCR1C is a novel activator of phosphodiesterase 4 (PDE4) long isoforms and attenuates cardiomyocyte hypertrophy. Cell. Signal. 27, 908–922 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  217. Guo, L. W. et al. The inhibitory gamma subunit of the rod cGMP phosphodiesterase binds the catalytic subunits in an extended linear structure. J. Biol. Chem. 281, 15412–15422 (2006).

    CAS  PubMed  Google Scholar 

  218. Zhang, X. J., Skiba, N. P. & Cote, R. H. Structural requirements of the photoreceptor phosphodiesterase gamma-subunit for inhibition of rod PDE6 holoenzyme and for its activation by transducin. J. Biol. Chem. 285, 4455–4463 (2010).

    CAS  PubMed  Google Scholar 

  219. Slep, K. C. et al. Structural determinants for regulation of phosphodiesterase by a G protein at 2.0 A. Nature 409, 1071–1077 (2001).

    CAS  PubMed  Google Scholar 

  220. Zhang, Z. et al. Domain organization and conformational plasticity of the G protein effector, PDE6. J. Biol. Chem. 290, 12833–12843 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  221. Lynch, M. J. et al. RNA silencing identifies PDE4D5 as the functionally relevant cAMP phosphodiesterase interacting with beta arrestin to control the protein kinase A/AKAP79-mediated switching of the beta2-adrenergic receptor to activation of ERK in HEK293B2 cells. J. Biol. Chem. 280, 33178–33189 (2005).

    CAS  PubMed  Google Scholar 

  222. Ren, L. et al. MiR-541-5p regulates lung fibrosis by targeting cyclic nucleotide phosphodiesterase 1A. Exp. Lung Res. 43, 249–258 (2017).

    CAS  PubMed  Google Scholar 

  223. Zhang, D. D., Li, Y., Xu, Y., Kim, J. & Huang, S. Phosphodiesterase 7B/microRNA-200c relationship regulates triple-negative breast cancer cell growth. Oncogene 38, 1106–1120 (2019).

    CAS  PubMed  Google Scholar 

  224. Yang, G. et al. Phosphodiesterase 7A-deficient mice have functional T cells. J. Immunol. 171, 6414–6420 (2003).

    CAS  PubMed  Google Scholar 

  225. Ding, B. et al. Functional role of phosphodiesterase 3 in cardiomyocyte apoptosis: implication in heart failure. Circulation 111, 2469–2476 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  226. Wu, W. H. et al. CRISPR repair reveals causative mutation in a preclinical model of retinitis pigmentosa. Mol. Ther. 24, 1388–1394 (2016). This work is a proof-of-concept study showing how gene editing might be used to therapeutically target a PDE.

    CAS  PubMed  PubMed Central  Google Scholar 

  227. Wensel, T. G. et al. Structural and molecular bases of rod photoreceptor morphogenesis and disease. Prog. Retin. Eye Res. 55, 32–51 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  228. Deng, W. T. et al. Cone phosphodiesterase-6α’ restores rod function and confers distinct physiological properties in the rod phosphodiesterase-6β-deficient rd10 mouse. J. Neurosci. 33, 11745–11753 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  229. Pang, J. J. et al. AAV-mediated gene therapy for retinal degeneration in the rd10 mouse containing a recessive PDEβ mutation. Invest. Ophthalmol. Vis. Sci. 49, 4278–4283 (2008). This work is one of the first studies showing that viral constructs can be used to therapeutically target a PDE.

    PubMed  PubMed Central  Google Scholar 

  230. Wert, K. J., Davis, R. J., Sancho-Pelluz, J., Nishina, P. M. & Tsang, S. H. Gene therapy provides long-term visual function in a pre-clinical model of retinitis pigmentosa. Hum. Mol. Genet. 22, 558–567 (2013).

    CAS  PubMed  Google Scholar 

  231. Deng, W. T. et al. Cone phosphodiesterase-6γ’ subunit augments cone PDE6 holoenzyme assembly and stability in a mouse model lacking both rod and cone PDE6 catalytic subunits. Front. Mol. Neurosci. 11, 233 (2018).

    PubMed  PubMed Central  Google Scholar 

  232. Carvalho, L. S. et al. Synthetic adeno-associated viral vector efficiently targets mouse and nonhuman primate retina in vivo. Hum. Gene Ther. 29, 771–784 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  233. Ye, H. & Fussenegger, M. Optogenetic medicine: synthetic therapeutic solutions precision-guided by light. Cold Spring Harb. Perspect. Med. https://doi.org/10.1101/cshperspect.a034371 (2018).

    Article  Google Scholar 

  234. Yoshida, K., Tsunoda, S. P., Brown, L. S. & Kandori, H. A unique choanoflagellate enzyme rhodopsin exhibits light-dependent cyclic nucleotide phosphodiesterase activity. J. Biol. Chem. 292, 7531–7541 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  235. Lamarche, L. B. et al. Purification and characterization of RhoPDE, a retinylidene/phosphodiesterase fusion protein and potential optogenetic tool from the choanoflagellate Salpingoeca rosetta. Biochemistry 56, 5812–5822 (2017). This work together with that of Yoshida et al. (2017) are the first studies to identify a light-dependent PDE with potential utility for optogenetic applications.

    CAS  PubMed  PubMed Central  Google Scholar 

  236. Tian, Y., Gao, S., Yang, S. & Nagel, G. A novel rhodopsin phosphodiesterase from Salpingoeca rosetta shows light-enhanced substrate affinity. Biochem. J. 475, 1121–1128 (2018).

    CAS  PubMed  Google Scholar 

  237. Gasser, C. et al. Engineering of a red-light-activated human cAMP/cGMP-specific phosphodiesterase. Proc. Natl Acad. Sci. USA 111, 8803–8808 (2014).

    CAS  PubMed  Google Scholar 

  238. Kelly, M. P. Cyclic nucleotide signaling changes associated with normal aging and age-related diseases of the brain. Cell. Signal. 42, 281–291 (2018). This review underscores the fact that any targeting of PDE activity in the context of age-related disease will likely need to be done in a tissue-specific manner, as some tissues see increased cyclic nucleotide signalling with age whereas others see decreased signalling.

    CAS  PubMed  Google Scholar 

  239. Fertig, B. A. & Baillie, G. S. PDE4-mediated cAMP signalling. J. Cardiovasc. Dev. Dis. 5, E8 (2018).

    PubMed  Google Scholar 

  240. McCahill, A. et al. In resting COS1 cells a dominant negative approach shows that specific, anchored PDE4 cAMP phosphodiesterase isoforms gate the activation, by basal cyclic AMP production, of AKAP-tethered protein kinase A type II located in the centrosomal region. Cell. Signal. 17, 1158–1173 (2005).

    CAS  PubMed  Google Scholar 

  241. Bolger, G. B. et al. Scanning peptide array analyses identify overlapping binding sites for the signalling scaffold proteins, beta-arrestin and RACK1, in cAMP-specific phosphodiesterase PDE4D5. Biochem. J. 398, 23–36 (2006). This work is a first report of a key technique in the field (that is, the scanning peptide array) that has been successfully used to identify many PDE protein–protein binding partners.

    CAS  PubMed  PubMed Central  Google Scholar 

  242. Baillie, G. S. et al. β-Arrestin-mediated PDE4 cAMP phosphodiesterase recruitment regulates β-adrenoceptor switching from Gs to Gi. Proc. Natl Acad. Sci. USA 100, 940–945 (2003). This work presents a seminal finding showing how PDEs can trigger negative feedback loops upstream (that is, by making a receptor switch from Gs to Gi signalling).

    CAS  PubMed  Google Scholar 

  243. Willoughby, D. et al. Dynamic regulation, desensitization, and cross-talk in discrete subcellular microdomains during beta2-adrenoceptor and prostanoid receptor cAMP signaling. J. Biol. Chem. 282, 34235–34249 (2007).

    CAS  PubMed  Google Scholar 

  244. Ong, W. K. et al. The role of the PDE4D cAMP phosphodiesterase in the regulation of glucagon-like peptide-1 release. Br. J. Pharmacol. 157, 633–644 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  245. Terrin, A. et al. PKA and PDE4D3 anchoring to AKAP9 provides distinct regulation of cAMP signals at the centrosome. J. Cell Biol. 198, 607–621 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  246. Vecsey, C. G. et al. Sleep deprivation impairs cAMP signalling in the hippocampus. Nature 461, 1122–1125 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  247. Havekes, R. et al. Sleep deprivation causes memory deficits by negatively impacting neuronal connectivity in hippocampal area CA1. eLife 5, e13424 (2016).

    PubMed  PubMed Central  Google Scholar 

  248. Havekes, R. et al. Compartmentalized PDE4A5 signaling impairs hippocampal synaptic plasticity and long-term memory. J. Neurosci. 36, 8936–8946 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  249. Campbell, S. L., van Groen, T., Kadish, I., Smoot, L. H. M. & Bolger, G. B. Altered phosphorylation, electrophysiology, and behavior on attenuation of PDE4B action in hippocampus. BMC Neurosci. 18, 77 (2017).

    PubMed  PubMed Central  Google Scholar 

  250. McGirr, A. et al. Specific inhibition of phosphodiesterase-4B results in anxiolysis and facilitates memory acquisition. Neuropsychopharmacology 41, 1080–1092 (2016).

    CAS  PubMed  Google Scholar 

  251. Johnston, L. A. et al. Expression, intracellular distribution and basis for lack of catalytic activity of the PDE4A7 isoform encoded by the human PDE4A cAMP-specific phosphodiesterase gene. Biochem. J. 380, 371–384 (2004). This report identifies a catalytically dead isoform of PDE4A, showing that nature has developed its own dominant-negative approach.

    CAS  PubMed  PubMed Central  Google Scholar 

  252. Lynch, M. J., Baillie, G. S. & Houslay, M. D. cAMP-specific phosphodiesterase-4D5 (PDE4D5) provides a paradigm for understanding the unique non-redundant roles that PDE4 isoforms play in shaping compartmentalized cAMP cell signalling. Biochem. Soc. Trans. 35, 938–941 (2007).

    CAS  PubMed  Google Scholar 

  253. Wills, L., Ehsan, M., Whiteley, E. L. & Baillie, G. S. Location, location, location: PDE4D5 function is directed by its unique N-terminal region. Cell. Signal. 28, (701–705 (2016).

    Google Scholar 

  254. Yun, S. et al. Interaction between integrin α5 and PDE4D regulates endothelial inflammatory signalling. Nat. Cell Biol. 18, 1043–1053 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  255. Ahmad, F. et al. 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. 290, 6763–6776 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  256. Lee, L. C., Maurice, D. H. & Baillie, G. S. Targeting protein–protein interactions within the cyclic AMP signaling system as a therapeutic strategy for cardiovascular disease. Future Med. Chem. 5, 451–464 (2013).

    CAS  PubMed  Google Scholar 

  257. Delyon, J. et al. PDE4D promotes FAK-mediated cell invasion in BRAF-mutated melanoma. Oncogene 36, 3252–3262 (2017).

    CAS  PubMed  Google Scholar 

  258. Serrels, B. et al. A complex between FAK, RACK1, and PDE4D5 controls spreading initiation and cancer cell polarity. Curr. Biol. 20, 1086–1092 (2010).

    CAS  PubMed  Google Scholar 

  259. Martin, T. P. et al. Targeted disruption of the heat shock protein 20-phosphodiesterase 4D (PDE4D) interaction protects against pathological cardiac remodelling in a mouse model of hypertrophy. FEBS Open Bio 4, 923–927 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  260. Keravis, T. & Lugnier, C. Cyclic nucleotide phosphodiesterases (PDE) and peptide motifs. Curr. Pharm. Des. 16, 1114–1125 (2010).

    CAS  PubMed  Google Scholar 

  261. MacMullen, C. M., Vick, K., Pacifico, R., Fallahi-Sichani, M. & Davis, R. L. Novel, primate-specific PDE10A isoform highlights gene expression complexity in human striatum with implications on the molecular pathology of bipolar disorder. Transl Psychiatry 6, e742 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  262. Movsesian, M. Novel approaches to targeting PDE3 in cardiovascular disease. Pharmacol. Ther. 163, 74–81 (2016).

    CAS  PubMed  Google Scholar 

  263. Kotera, J. et al. Subcellular localization of cyclic nucleotide phosphodiesterase type 10A variants, and alteration of the localization by cAMP-dependent protein kinase-dependent phosphorylation. J. Biol. Chem. 279, 4366–4375 (2004).

    CAS  PubMed  Google Scholar 

  264. Russwurm, C., Koesling, D. & Russwurm, M. Phosphodiesterase 10A is tethered to a synaptic signaling complex in striatum. J. Biol. Chem. 290, 11936–11947 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  265. Kelly, M. P. A. Role for phosphodiesterase 11A (PDE11A) in the formation of social memories and the stabilization of mood. Adv. Neurobiol. 17, 201–230 (2017).

    PubMed  PubMed Central  Google Scholar 

  266. Carlisle Michel, J. J. et al. PKA-phosphorylation of PDE4D3 facilitates recruitment of the mAKAP signalling complex. Biochem. J. 381, 587–592 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  267. Suryavanshi, S. V. et al. Human muscle-specific A-kinase anchoring protein polymorphisms modulate the susceptibility to cardiovascular diseases by altering cAMP/PKA signaling. Am. J. Physiol. Heart Circ. Physiol. 315, H109–H121 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  268. Zhu, H. et al. Evolutionarily conserved role of calcineurin in phosphodegron-dependent degradation of phosphodiesterase 4D. Mol. Cell. Biol. 30, 4379–4390 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  269. VerPlank, J. J. S. & Goldberg, A. L. Regulating protein breakdown through proteasome phosphorylation. Biochem. J. 474, 3355–3371 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  270. Cai, Y. et al. Smurf2, an E3 ubiquitin ligase, interacts with PDE4B and attenuates liver fibrosis through miR-132 mediated CTGF inhibition. Biochim. Biophys. Acta Mol. Cell Res. 1865, 297–308 (2018).

    CAS  PubMed  Google Scholar 

  271. Li, X., Baillie, G. S. & Houslay, M. D. Mdm2 directs the ubiquitination of beta-arrestin-sequestered cAMP phosphodiesterase-4D5. J. Biol. Chem. 284, 16170–16182 (2009). This study is the first to identify a PTM that regulates PDE signalling via degradation.

    CAS  PubMed  PubMed Central  Google Scholar 

  272. Li, X. et al. Selective SUMO modification of cAMP-specific phosphodiesterase-4D5 (PDE4D5) regulates the functional consequences of phosphorylation by PKA and ERK. Biochem. J. 428, 55–65 (2010).

    CAS  PubMed  Google Scholar 

  273. Hay, R. T. Decoding the SUMO signal. Biochem. Soc. Trans. 41, 463–473 (2013).

    CAS  PubMed  Google Scholar 

  274. Hoffmann, R., Baillie, G. S., MacKenzie, S. J., Yarwood, S. J. & Houslay, M. D. The MAP kinase ERK2 inhibits the cyclic AMP-specific phosphodiesterase HSPDE4D3 by phosphorylating it at Ser579. EMBO J. 18, 893–903 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  275. Wang, Y. et al. S-Nitrosylation of PDE5 increases its ubiquitin-proteasomal degradation. Free Radic. Biol. Med. 86, 343–351 (2015).

    CAS  PubMed  Google Scholar 

  276. Garcia, J. A. Aiming straight for the heart: prolyl hydroxylases set the BAR. Sci. Signal. 2, e70 (2009).

    Google Scholar 

  277. Huo, Z. et al. Prolyl hydroxylase domain protein 2 regulates the intracellular cyclic AMP level in cardiomyocytes through its interaction with phosphodiesterase 4D. Biochem. Biophys. Res. Commun. 427, 73–79 (2012).

    CAS  PubMed  Google Scholar 

  278. Lorenz, R., Bertinetti, D. & Herberg, F. W. cAMP-dependent protein kinase and cGMP-dependent protein kinase as cyclic nucleotide effectors. Handb. Exp. Pharmacol. 238, 105–122 (2017).

    CAS  PubMed  Google Scholar 

  279. VanSchouwen, B. & Melacini, G. Regulation of HCN ion channels by non-canonical cyclic nucleotides. Handb. Exp. Pharmacol. 238, 123–133 (2017).

    CAS  PubMed  Google Scholar 

  280. Rehmann, H. Interaction of Epac with non-canonical cyclic nucleotides. Handb. Exp. Pharmacol. 238, 135–147 (2017).

    CAS  PubMed  Google Scholar 

  281. Hauser, A. R. The type III secretion system of Pseudomonas aeruginosa: infection by injection. Nat. Rev. Microbiol. 7, 654–665 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  282. Wolter, S. et al. cCMP causes caspase-dependent apoptosis in mouse lymphoma cell lines. Biochem. Pharmacol. 98, 119–131 (2015).

    CAS  PubMed  Google Scholar 

  283. Wong, C. H., Siah, K. W. & Lo, A. W. Estimation of clinical trial success rates and related parameters. Biostatistics 20, 273–286 (2019).

    PubMed  Google Scholar 

  284. Liu, F. C. et al. Use of cilomilast-loaded phosphatiosomes to suppress neutrophilic inflammation for attenuating acute lung injury: the effect of nanovesicular surface charge. J. Nanobiotechnol. 16, 35 (2018).

    Google Scholar 

  285. Yu, S. et al. Targeted delivery of an anti-inflammatory PDE4 inhibitor to immune cells via an antibody-drug conjugate. Mol. Ther. 24, 2078–2089 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  286. Raymond, D. R., Wilson, L. S., Carter, R. L. & Maurice, D. H. Numerous distinct PKA-, or EPAC-based, signalling complexes allow selective phosphodiesterase 3 and phosphodiesterase 4 coordination of cell adhesion. Cell. Signal. 19, 2507–2518 (2007).

    CAS  PubMed  Google Scholar 

  287. Sapio, L. et al. Targeting protein kinase A in cancer therapy: an update. EXCLI J. 13, 843–855 (2014).

    PubMed  PubMed Central  Google Scholar 

  288. Raker, V. K., Becker, C. & Steinbrink, K. The cAMP pathway as therapeutic target in autoimmune and inflammatory diseases. Front. Immunol. 7, 123 (2016).

    PubMed  PubMed Central  Google Scholar 

  289. Kumar, N. et al. Insights into exchange factor directly activated by cAMP (EPAC) as potential target for cancer treatment. Mol. Cell Biochem. 447, 77–92 (2018).

    CAS  PubMed  Google Scholar 

  290. Parnell, E., Palmer, T. M. & Yarwood, S. J. The future of EPAC-targeted therapies: agonism versus antagonism. Trends Pharmacol. Sci. 36, 203–214 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  291. Holland, N. A. et al. Cyclic nucleotide-directed protein kinases in cardiovascular inflammation and growth. J. Cardiovasc. Dev. Dis. 5, E6 (2018).

    PubMed  Google Scholar 

  292. Kleppe, R., Krakstad, C., Selheim, F., Kopperud, R. & Doskeland, S. O. The cAMP-dependent protein kinase pathway as therapeutic target: possibilities and pitfalls. Curr. Top. Med. Chem. 11, 1393–1405 (2011).

    CAS  PubMed  Google Scholar 

  293. Bollen, E. et al. Improved long-term memory via enhancing cGMP-PKG signaling requires cAMP-PKA signaling. Neuropsychopharmacology 39, 2497–2505 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  294. Joshi, R. et al. Phosphodiesterase (PDE) inhibitor torbafylline (HWA 448) attenuates burn-induced rat skeletal muscle proteolysis through the PDE4/cAMP/EPAC/PI3K/Akt pathway. Mol. Cell. Endocrinol. 393, 152–163 (2014).

    CAS  PubMed  Google Scholar 

  295. Miller, C. L. et al. Cyclic nucleotide phosphodiesterase 1A: a key regulator of cardiac fibroblast activation and extracellular matrix remodeling in the heart. Bas. Res. Cardiol. 106, 1023–1039 (2011).

    CAS  Google Scholar 

  296. Zhang, H. T. et al. Antidepressant-like profile and reduced sensitivity to rolipram in mice deficient in the PDE4D phosphodiesterase enzyme. Neuropsychopharmacology 27, 587–595 (2002).

    CAS  PubMed  Google Scholar 

  297. Salpietro, V. et al. A homozygous loss-of-function mutation in PDE2A associated to early-onset hereditary chorea. Mov Disord. 33, 482–488 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  298. Iribarne, M. & Masai, I. Neurotoxicity of cGMP in the vertebrate retina: from the initial research on rd mutant mice to zebrafish genetic approaches. J. Neurogenet. 31, 88–101 (2017).

    CAS  PubMed  Google Scholar 

  299. Savai, R. et al. Targeting cancer with phosphodiesterase inhibitors. Expert Opin. Investig. Drugs 19, 117–131 (2010).

    CAS  PubMed  Google Scholar 

  300. Ahmad, F. et al. Cyclic nucleotide phosphodiesterases: important signaling modulators and therapeutic targets. Oral Dis. 21, e25–e50 (2015).

    CAS  PubMed  Google Scholar 

  301. Zuo, H., Cattani-Cavalieri, I., Musheshe, N., Nikolaev, V. O. & Schmidt, M. Phosphodiesterases as therapeutic targets for respiratory diseases. Pharmacol. Ther. 197, 225–242 (2019).

    CAS  PubMed  Google Scholar 

  302. Pincelli, C., Schafer, P. H., French, L. E., Augustin, M. & Krueger, J. G. Mechanisms underlying the clinical effects of apremilast for psoriasis. J. Drugs Dermatol. 17, 835–840 (2018).

    CAS  PubMed  Google Scholar 

  303. Gupta, A., Tiwari, M., Prasad, S. & Chaube, S. K. Role of cyclic nucleotide phosphodiesterases during meiotic resumption from diplotene arrest in mammalian oocytes. J. Cell. Biochem. 118, 446–452 (2017).

    CAS  PubMed  Google Scholar 

  304. Drobnis, E. Z. & Nangia, A. K. Phosphodiesterase inhibitors (PDE inhibitors) and male reproduction. Adv. Exp. Med. Biol. 1034, 29–38 (2017).

    PubMed  Google Scholar 

  305. Vasta, V. et al. Identification of a new variant of PDE1A calmodulin-stimulated cyclic nucleotide phosphodiesterase expressed in mouse sperm. Biol. Reprod. 73, 598–609 (2005).

    CAS  PubMed  Google Scholar 

  306. Alves de Inda, M. et al. Validation of cyclic adenosine monophosphate phosphodiesterase-4D7 for its independent contribution to risk stratification in a prostate cancer patient cohort with longitudinal biological outcomes. Eur. Urol. Focus 4, 376–384 (2018).

    PubMed  Google Scholar 

  307. van Strijp, D. et al. The prognostic PDE4D7 score in a diagnostic biopsy prostate cancer patient cohort with longitudinal biological outcomes. Prostate Cancer 2018, 5821616 (2018).

    PubMed  PubMed Central  Google Scholar 

  308. Bottcher, R. et al. Human phosphodiesterase 4D7 (PDE4D7) expression is increased in TMPRSS2-ERG-positive primary prostate cancer and independently adds to a reduced risk of post-surgical disease progression. Br. J. Cancer 113, 1502–1511 (2015). This work is the first of three key studies (see the two additional studies by Alves de India [2018] and van Strijp [2018]) that led to the development of a PDE4D7-based prostate cancer biomarker.

    CAS  PubMed  PubMed Central  Google Scholar 

  309. Bottcher, R. et al. Human PDE4D isoform composition is deregulated in primary prostate cancer and indicative for disease progression and development of distant metastases. Oncotarget 7, 70669–70684 (2016).

    PubMed  PubMed Central  Google Scholar 

  310. MDxHealth. MDxHealth launch agreement with Philips for prognostic prostate cancer biomarker. MDxHealth https://mdxhealth.com/press-release/mdxhealth-launch-agreement-philips-prognostic-prostate-cancer-biomarker (2018).

  311. Nazir, M. et al. Targeting tumor cells based on phosphodiesterase 3A expression. Exp. Cell Res. 361, 308–315 (2017).

    CAS  PubMed  Google Scholar 

  312. Vandenberghe, P. et al. Phosphodiesterase 3A: a new player in development of interstitial cells of Cajal and a prospective target in gastrointestinal stromal tumors (GIST). Oncotarget 8, 41026–41043 (2017).

    PubMed  PubMed Central  Google Scholar 

  313. Fryknas, M. et al. Phenotype-based screening of mechanistically annotated compounds in combination with gene expression and pathway analysis identifies candidate drug targets in a human squamous carcinoma cell model. J. Biomol. Screen 11, 457–468 (2006).

    PubMed  Google Scholar 

  314. Ahmad, R. et al. PET imaging shows loss of striatal PDE10A in patients with Huntington disease. Neurology 82, 279–281 (2014).

    PubMed  Google Scholar 

  315. Wilson, H. et al. Loss of extra-striatal phosphodiesterase 10A expression in early premanifest Huntington’s disease gene carriers. J. Neurol. Sci. 368, 243–248 (2016).

    CAS  PubMed  Google Scholar 

  316. Niccolini, F. et al. Loss of phosphodiesterase 10A expression is associated with progression and severity in Parkinson’s disease. Brain 138, 3003–3015 (2015). This work is an important study showing how in vivo imaging of a PDE may prove a useful biomarker for disease diagnosis and monitoring.

    PubMed  Google Scholar 

  317. Schroder, S., Wenzel, B., Deuther-Conrad, W., Scheunemann, M. & Brust, P. Novel radioligands for cyclic nucleotide phosphodiesterase imaging with positron emission tomography: an update on developments since 2012. Molecules 21, E650 (2016).

    PubMed  Google Scholar 

  318. Neves, S. R., Ram, P. T. & Iyengar, R. G protein pathways. Science 296, 1636–1639 (2002).

    CAS  PubMed  Google Scholar 

  319. Chen, J., Martinez, J., Milner, T. A., Buck, J. & Levin, L. R. Neuronal expression of soluble adenylyl cyclase in the mammalian brain. Brain Res. 1518, 1–8 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  320. Kobialka, M. & Gorczyca, W. A. Particulate guanylyl cyclases: multiple mechanisms of activation. Acta Biochim. Pol. 47, 517–528 (2000).

    CAS  PubMed  Google Scholar 

  321. Brand, T. The Popeye domain containing genes and their function as cAMP effector proteins in striated muscle. J. Cardiovasc. Dev. Dis. 5, E18 (2018).

    PubMed  Google Scholar 

  322. Gamanuma, M. et al. Comparison of enzymatic characterization and gene organization of cyclic nucleotide phosphodiesterase 8 family in humans. Cell. Signal. 15, 565–574 (2003).

    CAS  PubMed  Google Scholar 

  323. Goraya, T. A. & Cooper, D. M. Ca2+-calmodulin-dependent phosphodiesterase (PDE1): current perspectives. Cell. Signal. 17, 789–797 (2005).

    CAS  PubMed  Google Scholar 

  324. Pandit, J., Forman, M. D., Fennell, K. F., Dillman, K. S. & Menniti, F. S. Mechanism for the allosteric regulation of phosphodiesterase 2A deduced from the X-ray structure of a near full-length construct. Proc. Natl Acad. Sci. USA 106, 18225–18230 (2009). This seminal work reveals the three-dimensional nature by which cyclic nucleotide binding and protein–protein interactions at a PDE GAF domain can influence the ability of the regulatory N-terminal to control access of the catalytic pocket.

    CAS  PubMed  Google Scholar 

  325. Qureshi, B. M. et al. It takes two transducins to activate the cGMP-phosphodiesterase 6 in retinal rods. Open Biol. 8, 180075 (2018).

    PubMed  PubMed Central  Google Scholar 

  326. Francis, S. H., Houslay, M. D. & Conti, M. Phosphodiesterase inhibitors: factors that influence potency, selectivity, and action. Handb. Exp. Pharmacol. 204, 47–84 (2011).

    CAS  Google Scholar 

  327. Berthouze-Duquesnes, M. et al. Specific interactions between Epac1, beta-arrestin2 and PDE4D5 regulate beta-adrenergic receptor subtype differential effects on cardiac hypertrophic signaling. Cell. Signal. 25, 970–980 (2013).

    CAS  PubMed  Google Scholar 

  328. Bolger, G. B. RACK1 and beta-arrestin2 attenuate dimerization of PDE4 cAMP phosphodiesterase PDE4D5. Cell. Signal. 28, 706–712 (2016).

    CAS  PubMed  Google Scholar 

  329. Govindan, R. et al. A phase II study of carboplatin, etoposide, and exisulind in patients with extensive small cell lung cancer: CALGB 30104. J. Thorac. Oncol. 4, 220–226 (2009).

    PubMed  Google Scholar 

  330. Dawson, N. A. et al. A phase II study of estramustine, docetaxel, and exisulind in patients with hormone-refractory prostate cancer: results of cancer and leukemia group B trial 90004. Clin. Genitourin. Cancer 6, 110–116 (2008).

    CAS  PubMed  Google Scholar 

  331. Yu, E. Y. et al. A pilot study of high-dose exisulind in men with biochemical relapse (BCR) of prostate cancer after definitive local therapy treated with intermittent androgen deprivation (IAD). J. Clin. Oncol. 31, 209–209 (2013).

    Google Scholar 

  332. Curiel-Lewandrowski, C. et al. Randomized, double-blind, placebo-controlled trial of sulindac in individuals at risk for melanoma: evaluation of potential chemopreventive activity. Cancer 118, 5848–5856 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  333. Leung, D. G. et al. Sildenafil does not improve cardiomyopathy in Duchenne/Becker muscular dystrophy. Ann. Neurol. 76, 541–549 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  334. Victor, R. G. et al. A phase 3 randomized placebo-controlled trial of tadalafil for Duchenne muscular dystrophy. Neurology 89, 1811–1820 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  335. McDonald, C. M. et al. The 6-minute walk test as a new outcome measure in Duchenne muscular dystrophy. Muscle Nerve 41, 500–510 (2010).

    PubMed  Google Scholar 

  336. Ramirez, C. E. et al. Treatment with sildenafil improves insulin sensitivity in prediabetes: a randomized, controlled trial. J. Clin. Endocrinol. Metab. 100, 4533–4540 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  337. Frolich, L. et al. Evaluation of the efficacy, safety and tolerability of orally administered BI 409306, a novel phosphodiesterase type 9 inhibitor, in two randomised controlled phase II studies in patients with prodromal and mild Alzheimer’s disease. Alzheimers Res. Ther. 11, 18 (2019).

    PubMed  PubMed Central  Google Scholar 

  338. Targum, S. D. et al. Application of external review for subject selection in a schizophrenia trial. J. Clin. Psychopharmacol. 32, 825–826 (2012).

    PubMed  Google Scholar 

  339. Targum, S. D. et al. Impact of BPRS interview length on ratings reliability in a schizophrenia trial. Eur. Neuropsychopharmacol. 25, 312–318 (2015).

    CAS  PubMed  Google Scholar 

  340. Delnomdedieu, M. et al. In vivo measurement of PDE10A enzyme occupancy by positron emission tomography (PET) following single oral dose administration of PF-02545920 in healthy male subjects. Neuropharmacology 117, 171–181 (2017). This study is an example of how clinical trials are attempting to verify target engagement when testing PDE inhibitors in the context of nervous system disorders.

    CAS  PubMed  Google Scholar 

  341. Metz, V. E. et al. Effects of ibudilast on the subjective, reinforcing, and analgesic effects of oxycodone in recently detoxified adults with opioid dependence. Neuropsychopharmacology 42, 1825–1832 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  342. Zhang, F. et al. Vinpocetine inhibits NF-κB-dependent inflammation in acute ischemic stroke patients. Transl Stroke Res. 9, 174–184 (2018).

    CAS  PubMed  Google Scholar 

  343. Ma, X. W. et al. A randomized, open-label, multicentre study to evaluate plasma atherosclerotic biomarkers in patients with type 2 diabetes mellitus and arteriosclerosis obliterans when treated with probucol and cilostazol. J. Geriatr. Cardiol. 9, 228–236 (2012).

    PubMed  PubMed Central  Google Scholar 

  344. Lee, J. Y. et al. Efficacy of cilostazol administration in Alzheimer’s disease patients with white matter lesions: a positron-emission tomography study. Neurotherapeutics 16, 394–403 (2019).

    CAS  PubMed  Google Scholar 

  345. Heckman, P. R. A. et al. Acute administration of roflumilast enhances sensory gating in healthy young humans in a randomized trial. Psychopharmacology 235, 301–308 (2018).

    CAS  PubMed  Google Scholar 

  346. Wouters, E. F. et al. Effect of the phosphodiesterase 4 inhibitor roflumilast on glucose metabolism in patients with treatment-naive, newly diagnosed type 2 diabetes mellitus. J. Clin. Endocrinol. Metab. 97, E1720–E1725 (2012).

    CAS  PubMed  Google Scholar 

  347. Gonzalez-Ortiz, M., Martinez-Abundis, E., Hernandez-Corona, D. M. & Ramirez-Rodriguez, A. M. Effect of tadalafil administration on insulin secretion and insulin sensitivity in obese men. Acta Clin. Belg. 72, 326–330 (2017).

    PubMed  Google Scholar 

  348. Sbardella, E. et al. Cardiovascular features of possible autonomous cortisol secretion in patients with adrenal incidentalomas. Eur. J. Endocrinol. 178, 501–511 (2018).

    CAS  PubMed  Google Scholar 

  349. Pauls, M. M. H. et al. Perfusion by arterial spin labelling following single dose tadalafil in small vessel disease (PASTIS): study protocol for a randomised controlled trial. Trials 18, 229 (2017).

    PubMed  PubMed Central  Google Scholar 

  350. Charnigo, R. J. et al. PF-04447943, a phosphodiesterase 9A inhibitor, in stable sickle cell disease patients: a phase Ib randomized, placebo-controlled study. Clin. Transl Sci. 12, 180–188 (2019).

    CAS  PubMed  Google Scholar 

  351. Moschetti, V. et al. First-in-human study assessing safety, tolerability and pharmacokinetics of BI 409306, a selective phosphodiesterase 9A inhibitor, in healthy males. Br. J. Clin. Pharmacol. 82, 1315–1324 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  352. Boland, K. et al. A phase I, randomized, proof-of-clinical-mechanism study assessing the pharmacokinetics and pharmacodynamics of the oral PDE9A inhibitor BI 409306 in healthy male volunteers. Hum. Psychopharmacol. 32, e2569 (2017).

    Google Scholar 

  353. Fazio, P. et al. Patterns of age related changes for phosphodiesterase type-10A in comparison with dopamine D2/3 receptors and sub-cortical volumes in the human basal ganglia: a PET study with (18)F-MNI-659 and (11)C-raclopride with correction for partial volume effect. Neuroimage 152, 330–339 (2017).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Work supported by 1R01MH101130 and 1R01AG061200 (M.P.K.). G.S.B. is supported by grants from the British Heart Foundation (BHF/TARGETPDE/PG/17/26/32881) and Medical Research Council (MC-PC-13063 and MC-PC-15039). G.S.T. is a fellow of the AstraZeneca postdoctoral programme.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Michy P. Kelly.

Ethics declarations

Competing interests

G.S.B. is co-founder and director of Portage Glasgow Limited. The other authors declare no competing interests. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Glossary

Nanodomains

Subcellular compartments within which intracellular signalling is discretely localized largely due to anchoring of molecules within macromolecular complexes.

Acrodysostosis

A rare congenital disorder characterized by abnormal bone growth leading to very short fingers/toes, underdeveloped facial bones, a small nose and short stature, as well as developmental and intellectual disabilities.

Cerebral hypofusion

Reduces blood flow to/throughout the brain.

Coronary stent stenosis

A gradual re-narrowing of a coronary artery leading to restricted blood flow that occurs after angioplasty and stenting have been performed to relieve a prior blockage.

Dystonia

A movement disorder characterized by repetitive or twisting movements due to involuntary muscle contraction.

Lichen planus

An inflammatory condition characterized by purplish, itchy bumps on/around the skin, hair, nails and mucous membrane (due to unknown causes).

Dyskinesias

Diminished voluntary movements or uncontrollable involuntary movements (for example, tics or chorea).

Retinitis pigmentosa

A rare genetic disorder characterized by black pigmentation and retinal degeneration.

Trans-capping

The process whereby the regulatory domains from one phosphodiesterase (PDE) enzyme physically bind — and, thus, block substrate access to — the C-terminal catalytic domain of another PDE molecule.

Phosphodegron

A phosphorylation motif that alters the rate of degradation by attracting ubiquitin ligases to the phosphorylated protein.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Baillie, G.S., Tejeda, G.S. & Kelly, M.P. Therapeutic targeting of 3′,5′-cyclic nucleotide phosphodiesterases: inhibition and beyond. Nat Rev Drug Discov 18, 770–796 (2019). https://doi.org/10.1038/s41573-019-0033-4

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41573-019-0033-4

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer