Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Fabrice Vandeput is active.

Publication


Featured researches published by Fabrice Vandeput.


Journal of Biological Chemistry | 2007

Cyclic Nucleotide Phosphodiesterase PDE1C1 in Human Cardiac Myocytes

Fabrice Vandeput; Sharon L. Wolda; Judith Krall; Ryan Hambleton; Lothar Uher; Kim N. McCaw; Przemysław B. Radwański; Vincent Florio; Matthew A. Movsesian

Isoforms in the PDE1 family of cyclic nucleotide phosphodiesterases were recently found to comprise a significant portion of the cGMP-inhibited cAMP hydrolytic activity in human hearts. We examined the expression of PDE1 isoforms in human myocardium, characterized their catalytic activity, and quantified their contribution to cAMP hydrolytic and cGMP hydrolytic activity in subcellular fractions of this tissue. Western blotting with isoform-selective anti-PDE1 monoclonal antibodies showed PDE1C1 to be the principal isoform expressed in human myocardium. Immunohistochemical analysis showed that PDE1C1 is distributed along the Z-lines and M-lines of cardiac myocytes in a striated pattern that differs from that of the other major dual-specificity cyclic nucleotide phosphodiesterase in human myocardium, PDE3A. Most of the PDE1C1 activity was recovered in soluble fractions of human myocardium. It binds both cAMP and cGMP with Km values of ∼1 μm and hydrolyzes both substrates with similar catalytic rates. PDE1C1 activity in subcellular fractions was quantified using a new PDE1-selective inhibitor, IC295. At substrate concentrations of 0.1 μm, PDE1C1 constitutes the great majority of cAMP hydrolytic and cGMP hydrolytic activity in soluble fractions and the majority of cGMP hydrolytic activity in microsomal fractions, whereas PDE3 constitutes the majority of cAMP hydrolytic activity in microsomal fractions. These results indicate that PDE1C1 is expressed at high levels in human cardiac myocytes with an intracellular distribution distinct from that of PDE3A and that it may have a role in the integration of cGMP-, cAMP- and Ca2+-mediated signaling in these cells.


Nature Genetics | 2015

PDE3A mutations cause autosomal dominant hypertension with brachydactyly

Philipp G. Maass; Atakan Aydin; Friedrich C. Luft; Carolin Schächterle; Anja Weise; Sigmar Stricker; Carsten Lindschau; Martin Vaegler; Fatimunnisa Qadri; Hakan R. Toka; Herbert Schulz; Peter Krawitz; Dmitri Parkhomchuk; Jochen Hecht; Irene Hollfinger; Yvette Wefeld-Neuenfeld; Eireen Bartels-Klein; Astrid Mühl; Martin Kann; Herbert Schuster; David Chitayat; Martin G. Bialer; Thomas F. Wienker; Jurg Ott; Katharina Rittscher; Thomas Liehr; Jens Jordan; Ghislaine Plessis; Jens Tank; Knut Mai

Cardiovascular disease is the most common cause of death worldwide, and hypertension is the major risk factor. Mendelian hypertension elucidates mechanisms of blood pressure regulation. Here we report six missense mutations in PDE3A (encoding phosphodiesterase 3A) in six unrelated families with mendelian hypertension and brachydactyly type E (HTNB). The syndrome features brachydactyly type E (BDE), severe salt-independent but age-dependent hypertension, an increased fibroblast growth rate, neurovascular contact at the rostral-ventrolateral medulla, altered baroreflex blood pressure regulation and death from stroke before age 50 years when untreated. In vitro analyses of mesenchymal stem cell–derived vascular smooth muscle cells (VSMCs) and chondrocytes provided insights into molecular pathogenesis. The mutations increased protein kinase A–mediated PDE3A phosphorylation and resulted in gain of function, with increased cAMP-hydrolytic activity and enhanced cell proliferation. Levels of phosphorylated VASP were diminished, and PTHrP levels were dysregulated. We suggest that the identified PDE3A mutations cause the syndrome. VSMC-expressed PDE3A deserves scrutiny as a therapeutic target for the treatment of hypertension.


Journal of Pharmacology and Experimental Therapeutics | 2009

cGMP-Hydrolytic Activity and Its Inhibition by Sildenafil in Normal and Failing Human and Mouse Myocardium

Fabrice Vandeput; Judith Krall; Ramzi Ockaili; Fadi N. Salloum; Vincent A. Florio; Jackie D. Corbin; Sharron H. Francis; Rakesh C. Kukreja; Matthew A. Movsesian

In mouse models of cardiac disease, the type 5 (PDE5)-selective cyclic nucleotide phosphodiesterase inhibitor sildenafil has antihypertrophic and cardioprotective effects attributable to the inhibition of cGMP hydrolysis. To investigate the relevance of these findings to humans, we quantified cGMP-hydrolytic activity and its inhibition by sildenafil in cytosolic and microsomal preparations from the left ventricular myocardium of normal and failing human hearts. The vast majority of cGMP-hydrolytic activity was attributable to PDE1 and PDE3. Sildenafil had no measurable effect on cGMP hydrolysis at 10 nM, at which it is selective for PDE5, but it had a marked effect on cGMP and cAMP hydrolysis at 1 μM, at which it inhibits PDE1. In contrast, in preparations from the left ventricles of normal mice and mice with heart failure resulting from coronary artery ligation, the effects of sildenafil on cGMP hydrolysis were attributable to inhibition of both PDE5 and PDE1; PDE5 comprised ∼22 and ∼43% of the cytosolic cGMP-hydrolytic activity in preparations from normal and failing mouse hearts, respectively. These differences in PDE5 activities in human and mouse hearts call into question the extent to which the effects of sildenafil in mouse models are likely to be applicable in humans and raise the possibility of PDE1 as an alternative therapeutic target.


Heart Failure Reviews | 2009

Phosphodiesterase inhibition in heart failure.

Matthew A. Movsesian; Josef Stehlik; Fabrice Vandeput; Michael R. Bristow

Drugs that inhibit cyclic nucleotide phosphodiesterase activity act to increase intracellular cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP) content. In total, 11 families of these enzymes—which differ with respect to affinity for cAMP and cGMP, cellular expression, intracellular localization, and mechanisms of regulation—have been identified. Inhibitors of enzymes in the PDE3 family of cyclic nucleotide phosphodiesterases raise intracellular cAMP content in cardiac and vascular smooth muscle, with inotropic and, to a lesser extent, vasodilatory actions. These drugs have been used for many years in the treatment of patients with heart failure, but their long-term use has generally been shown to increase mortality through mechanisms that remain unclear. More recently, inhibitors of PDE5 cyclic nucleotide phosphodiesterases have been used as cGMP-raising agents in vascular smooth muscle. With respect to cardiovascular disease, there is evidence that these drugs are more efficacious in the pulmonary than in the systemic vasculature, for which reason they are used principally in patients with pulmonary hypertension. Effects attributable to inhibition of myocardial PDE5 activity are less well characterized. New information indicating that enzymes from the PDE1 family of cyclic nucleotide phosphodiesterases constitute the majority of cAMP- and cGMP-hydrolytic activity in human myocardium raises questions as to their role in regulating these signaling pathways in heart failure.


Journal of Biological Chemistry | 2015

Regulation of sarcoplasmic reticulum Ca2+ ATPase 2 (SERCA2) activity by phosphodiesterase 3A (PDE3A) in human myocardium: phosphorylation-dependent interaction of PDE3A1 with SERCA2.

Faiyaz Ahmad; Weixing Shen; Fabrice Vandeput; Nicolas Szabo-Fresnais; Judith Krall; Eva Degerman; Frank Goetz; Enno Klussmann; Matthew A. Movsesian; Vincent C. Manganiello

Background: PDE3A is part of a SERCA2 signaling complex in cardiac myocytes. Results: PDE3, not PDE4, regulates the activation of SERCA2 by PKA in human myocardium; phosphorylation of PDE3A1 at Ser-292/Ser-293 promotes its integration into the SERCA2 signaling complex. Conclusion: PDE3A1 regulates cAMP-mediated control of SERCA2 through its phosphorylation-dependent interaction with SERCA2. Significance: Targeted disruption of the PDE3A1-SERCA2 interaction may provide a new therapeutic approach for heart failure. Cyclic nucleotide phosphodiesterase 3A (PDE3) regulates cAMP-mediated signaling in the heart, and PDE3 inhibitors augment contractility in patients with heart failure. Studies in mice showed that PDE3A, not PDE3B, is the subfamily responsible for these inotropic effects and that murine PDE3A1 associates with sarcoplasmic reticulum Ca2+ ATPase 2 (SERCA2), phospholamban (PLB), and AKAP18 in a multiprotein signalosome in human sarcoplasmic reticulum (SR). Immunohistochemical staining demonstrated that PDE3A co-localizes in Z-bands of human cardiac myocytes with desmin, SERCA2, PLB, and AKAP18. In human SR fractions, cAMP increased PLB phosphorylation and SERCA2 activity; this was potentiated by PDE3 inhibition but not by PDE4 inhibition. During gel filtration chromatography of solubilized SR membranes, PDE3 activity was recovered in distinct high molecular weight (HMW) and low molecular weight (LMW) peaks. HMW peaks contained PDE3A1 and PDE3A2, whereas LMW peaks contained PDE3A1, PDE3A2, and PDE3A3. Western blotting showed that endogenous HMW PDE3A1 was the principal PKA-phosphorylated isoform. Phosphorylation of endogenous PDE3A by rPKAc increased cAMP-hydrolytic activity, correlated with shift of PDE3A from LMW to HMW peaks, and increased co-immunoprecipitation of SERCA2, cav3, PKA regulatory subunit (PKARII), PP2A, and AKAP18 with PDE3A. In experiments with recombinant proteins, phosphorylation of recombinant human PDE3A isoforms by recombinant PKA catalytic subunit increased co-immunoprecipitation with rSERCA2 and rat rAKAP18 (recombinant AKAP18). Deletion of the recombinant human PDE3A1/PDE3A2 N terminus blocked interactions with recombinant SERCA2. Serine-to-alanine substitutions identified Ser-292/Ser-293, a site unique to human PDE3A1, as the principal site regulating its interaction with SERCA2. These results indicate that phosphorylation of human PDE3A1 at a PKA site in its unique N-terminal extension promotes its incorporation into SERCA2/AKAP18 signalosomes, where it regulates a discrete cAMP pool that controls contractility by modulating phosphorylation-dependent protein-protein interactions, PLB phosphorylation, and SERCA2 activity.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Selective regulation of cyclic nucleotide phosphodiesterase PDE3A isoforms

Fabrice Vandeput; Nicolas Szabo-Fresnais; Faiyaz Ahmad; Changwon Kho; Ahyoung Lee; Judith Krall; Allan J. Dunlop; Mark Hazel; James A. Wohlschlegel; Roger J. Hajjar; Miles D. Houslay; Vincent C. Manganiello; Matthew A. Movsesian

Significance Inhibitors of cyclic nucleotide phosphodiesterase (PDE) PDE3A increase cardiac contractility in patients with heart failure, but their long-term use increases mortality. Two isoforms expressed in cardiac myocytes, PDE3A1 and PDE3A2, have amino acid sequences that are identical except for a unique N-terminal extension in PDE3A1. We found that PDE3A1 and PDE3A2 are selectively phosphorylated at alternative sites in response to the activation of PKA and PKC, respectively, resulting in differential regulation of their catalytic activity and protein interactomes. Existing PDE3 inhibitors thus target at least two functionally distinct cardiac isoforms likely with different roles in intracellular signaling. This raises the possibility that isoform-selective targeting may increase contractility in failing hearts without increasing mortality, providing a potential route for developing therapeutics. Inhibitors of cyclic nucleotide phosphodiesterase (PDE) PDE3A have inotropic actions in human myocardium, but their long-term use increases mortality in patients with heart failure. Two isoforms in cardiac myocytes, PDE3A1 and PDE3A2, have identical amino acid sequences except for a unique N-terminal extension in PDE3A1. We expressed FLAG-tagged PDE3A1 and PDE3A2 in HEK293 cells and examined their regulation by PKA- and PKC-mediated phosphorylation. PDE3A1, which is localized to intracellular membranes, and PDE3A2, which is cytosolic, were phosphorylated at different sites within their common sequence. Exposure to isoproterenol led to phosphorylation of PDE3A1 at the 14-3-3–binding site S312, whereas exposure to PMA led to phosphorylation of PDE3A2 at an alternative 14-3-3–binding site, S428. PDE3A2 activity was stimulated by phosphorylation at S428, whereas PDE3A1 activity was not affected by phosphorylation at either site. Phosphorylation of PDE3A1 by PKA and of PDE3A2 by PKC led to shifts in elution on gel-filtration chromatography consistent with increased interactions with other proteins, and 2D electrophoresis of coimmunoprecipitated proteins revealed that the two isoforms have distinct protein interactomes. A similar pattern of differential phosphorylation of endogenous PDE3A1 and PDE3A2 at S312 and S428 is observed in human myocardium. The selective phosphorylation of PDE3A1 and PDE3A2 at alternative sites through different signaling pathways, along with the different functional consequences of phosphorylation for each isoform, suggest they are likely to have distinct roles in cyclic nucleotide-mediated signaling in human myocardium, and raise the possibility that isoform-selective inhibition may allow inotropic responses without an increase in mortality.


Cellular Signalling | 2014

Phosphodiesterase 4 interacts with the 5-HT4(b) receptor to regulate cAMP signaling

Sabine Weninger; K. Van Craenenbroeck; Ryan T. Cameron; Fabrice Vandeput; Matthew A. Movsesian; George S. Baillie; Romain Lefebvre

Phosphodiesterase (PDE) 3 and PDE4, which degrade cyclic adenosine monophosphate (cAMP), are important regulators of 5-hydroxytryptamine (5-HT) 4 receptor signaling in cardiac tissue. Therefore, we investigated whether they interact with the 5-HT4(b) receptor, and whether A-kinase anchoring proteins (AKAPs), scaffolding proteins that bind to the regulatory subunit of protein kinase A (PKA) and contribute to the spacial-temporal control of cAMP signaling, are involved in the regulation of 5-HT4(b) receptor signaling. By measuring PKA activity in the absence and presence of PDE3 and PDE4 inhibitiors, we found that constitutive signaling of the overexpressed HA-tagged 5-HT4(b) receptor in HEK293 cells is regulated predominantly by PDE4, with a secondary role for PDE3 that is unmasked in the presence of PDE4 inhibition. Overexpressed PDE4D3 and PDE3A1, and to a smaller extent PDE4D5 co-immunoprecipitate constitutively with the 5-HT4(b) receptor. PDE activity measurements in immunoprecipitates of the 5-HT4(b) receptor confirm the association of PDE4D3 with the receptor and provide evidence that the activity of this PDE may be increased upon receptor stimulation with 5-HT. A possible involvement of AKAPs in 5-HT4(b) receptor signaling was uncovered in experiments using the St-Ht31 inhibitor peptide, which disrupts the interaction of AKAPs with PKA. However, St-Ht31 did not influence 5-HT4(b) receptor-stimulated PKA activity, and endogenous AKAP79 and gravin were not found in immunoprecipitates of the 5-HT4(b) receptor. In conclusion, we found that both PDE3A1 and PDE4D3 are integrated into complexes that contain the 5-HT4(b) receptor and may thereby regulate 5-HT4(b) receptor-mediated signaling.


Journal of Medicinal Chemistry | 2018

Discovery of Potent and Selective Periphery-Restricted Quinazoline Inhibitors of the Cyclic Nucleotide Phosphodiesterase PDE1

John Michael Humphrey; Matthew A. Movsesian; Christopher W. am Ende; Stacey L. Becker; Thomas A. Chappie; Stephen Jenkinson; Jennifer Liras; Spiros Liras; Orozco Cc; Jayvardhan Pandit; Felix Vajdos; Fabrice Vandeput; Eddie Yang; Frank S. Menniti

We disclose the discovery and X-ray cocrystal data of potent, selective quinazoline inhibitors of PDE1. Inhibitor ( S)-3 readily attains free plasma concentrations above PDE1 IC50 values and has restricted brain access. The racemic compound 3 inhibits >75% of PDE hydrolytic activity in soluble samples of human myocardium, consistent with heightened PDE1 activity in this tissue. These compounds represent promising new tools to probe the value of PDE1 inhibition in the treatment of cardiovascular disease.


Circulation Research | 2015

Cardiac Hypertrophy Is Inhibited by a Local Pool of cAMP Regulated by Phosphodiesterase 2.

Anna Zoccarato; Nicoletta C. Surdo; Jan Magnus Aronsen; Laura Ashley Fields; Luisa Mancuso; Giuliano Dodoni; Alessandra Stangherlin; Craig Livie; He Jiang; Yuan Yan Sin; F. Gesellchen; Anna Terrin; George S. Baillie; Stuart A. Nicklin; Delyth Graham; Nicolas Szabo-Fresnais; Judith Krall; Fabrice Vandeput; Matthew A. Movsesian; Leonardo Furlan; Veronica Corsetti; Graham Hamilton; Konstantinos Lefkimmiatis; Ivar Sjaastad; Manuela Zaccolo


Current Opinion in Pharmacology | 2011

PDE3 inhibition in dilated cardiomyopathy

Matthew A. Movsesian; Omar Wever-Pinzon; Fabrice Vandeput

Collaboration


Dive into the Fabrice Vandeput's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Faiyaz Ahmad

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ahyoung Lee

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Changwon Kho

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Roger J. Hajjar

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Weixing Shen

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge