Show simple item record

dc.contributor.authorBaliga, RSen_US
dc.contributor.authorPreedy, MEJen_US
dc.contributor.authorDukinfield, MSen_US
dc.contributor.authorChu, SMen_US
dc.contributor.authorAubdool, AAen_US
dc.contributor.authorBubb, KJen_US
dc.contributor.authorMoyes, AJen_US
dc.contributor.authorTones, MAen_US
dc.contributor.authorHobbs, AJen_US
dc.date.accessioned2018-07-31T09:53:53Z
dc.date.available2018-06-26en_US
dc.date.issued2018-07-31en_US
dc.date.submitted2018-07-18T09:05:00.881Z
dc.identifier.urihttp://qmro.qmul.ac.uk/xmlui/handle/123456789/42747
dc.description.abstractHeart failure (HF) is a shared manifestation of several cardiovascular pathologies, including hypertension and myocardial infarction, and a limited repertoire of treatment modalities entails that the associated morbidity and mortality remain high. Impaired nitric oxide (NO)/guanylyl cyclase (GC)/cyclic guanosine-3',5'-monophosphate (cGMP) signaling, underpinned, in part, by up-regulation of cyclic nucleotide-hydrolyzing phosphodiesterase (PDE) isozymes, contributes to the pathogenesis of HF, and interventions targeted to enhancing cGMP have proven effective in preclinical models and patients. Numerous PDE isozymes coordinate the regulation of cardiac cGMP in the context of HF; PDE2 expression and activity are up-regulated in experimental and human HF, but a well-defined role for this isoform in pathogenesis has yet to be established, certainly in terms of cGMP signaling. Herein, using a selective pharmacological inhibitor of PDE2, BAY 60-7550, and transgenic mice lacking either NO-sensitive GC-1α (GC-1α-/-) or natriuretic peptide-responsive GC-A (GC-A-/-), we demonstrate that the blockade of PDE2 promotes cGMP signaling to offset the pathogenesis of experimental HF (induced by pressure overload or sympathetic hyperactivation), reversing the development of left ventricular hypertrophy, compromised contractility, and cardiac fibrosis. Moreover, we show that this beneficial pharmacodynamic profile is maintained in GC-A-/- mice but is absent in animals null for GC-1α or treated with a NO synthase inhibitor, revealing that PDE2 inhibition preferentially enhances NO/GC/cGMP signaling in the setting of HF to exert wide-ranging protection to preserve cardiac structure and function. These data substantiate the targeting of PDE2 in HF as a tangible approach to maximize myocardial cGMP signaling and enhancing therapy.en_US
dc.description.sponsorshipBritish Heart Foundation Grant PG/10/077/28554.en_US
dc.format.extentE7428 - E7437en_US
dc.languageengen_US
dc.language.isoenen_US
dc.relation.ispartofProc Natl Acad Sci U S Aen_US
dc.subjectcyclic GMPen_US
dc.subjectheart failureen_US
dc.subjectnatriuretic peptideen_US
dc.subjectnitric oxideen_US
dc.subjectphosphodiesteraseen_US
dc.subjectAnimalsen_US
dc.subjectCells, Cultureden_US
dc.subjectCyclic GMPen_US
dc.subjectCyclic Nucleotide Phosphodiesterases, Type 2en_US
dc.subjectGuanylate Cyclaseen_US
dc.subjectHeart Failureen_US
dc.subjectMaleen_US
dc.subjectMiceen_US
dc.subjectNitric Oxideen_US
dc.subjectPhosphodiesterase Inhibitorsen_US
dc.subjectSignal Transductionen_US
dc.titlePhosphodiesterase 2 inhibition preferentially promotes NO/guanylyl cyclase/cGMP signaling to reverse the development of heart failure.en_US
dc.typeArticle
dc.identifier.doi10.1073/pnas.1800996115en_US
pubs.author-urlhttps://www.ncbi.nlm.nih.gov/pubmed/30012589en_US
pubs.issue31en_US
pubs.notesNo embargoen_US
pubs.publication-statusPublisheden_US
pubs.volume115en_US


Files in this item

Thumbnail

This item appears in the following Collection(s)

Show simple item record