73 results on '"Maurice DH"'
Search Results
2. Cyclic nucleotide-mediated regulation of vascular smooth muscle cell cyclic nucleotide phosphodiesterase activity. Selective effect of cyclic AMP
- Author
-
Maurice Dh
- Subjects
Exonucleases ,Nitroprusside ,medicine.medical_specialty ,Vascular smooth muscle ,Phosphodiesterase 3 ,Biophysics ,8-Bromo Cyclic Adenosine Monophosphate ,PDE1 ,Biochemistry ,Muscle, Smooth, Vascular ,Cyclic nucleotide ,chemistry.chemical_compound ,Internal medicine ,medicine ,Cyclic AMP ,Animals ,heterocyclic compounds ,Cyclic GMP ,Aorta ,Cells, Cultured ,Forskolin ,Cyclic nucleotide phosphodiesterase ,Phosphoric Diester Hydrolases ,Colforsin ,Phosphodiesterase ,Cell Biology ,General Medicine ,musculoskeletal system ,Cyclic Nucleotide Phosphodiesterases, Type 3 ,Cyclic Nucleotide Phosphodiesterases, Type 4 ,Rats ,Enzyme Activation ,enzymes and coenzymes (carbohydrates) ,Endocrinology ,chemistry ,3',5'-Cyclic-AMP Phosphodiesterases ,sense organs ,PDE10A ,circulatory and respiratory physiology - Abstract
Cultured rat aortic vascular smooth muscle cells (VSMC) express both cGMP- inhibited cAMP phosphodiesterase (PDE-3) and Ro,20-1724-inhibited cAMP phosphodiesterase (PDE-4) activities. Utilizing a PDE-3-selective inhibitor (cilostamide) and a PDE-4-selective inhibitor (Ro,20-1724), PDE-3 and PDE-4 activities were shown to account for 15 and 55% of total VSMC cAMP phosphodiesterase (PDE) activity. Incubations of VSMC with either forskolin or 8-bromo-cAMP caused a concentration- and time-dependent increase in total cellular cAMP PDE activity. In these cells, both PDE-3 and PDE-4 activities were increased, with a relatively larger effect observed on PDE-3 activity. Similar incubations with an activator of soluble guanylyl cyclase (sodium nitroprusside) or with 8-bromo-cGMP did not increase cAMP PDE activity. cAMP-induced increases in cAMP PDE activity were inhibited with actinomycin D or cycloheximide, demonstrating that new mRNA and protein synthesis were required. We conclude that VSMC cAMP PDE activity is elevated following long-term elevation of cAMP, and that increases in PDE-3 and PDE-4 activities account for more than 70% of this increase. These results may have implications for long-term use of cAMP PDE inhibitors as therapeutic agents.
- Published
- 1998
3. PDEs and Endothelial Cell Function
- Author
-
Maurice, DH, primary
- Published
- 2012
- Full Text
- View/download PDF
4. Phosphodiesterase 4 activity uniquely regulates ciliary cAMP-dependent 3T3-L1 adipogenesis.
- Author
-
Erdelsky MR, Groves SA, Shah C, Delios SB, Umana MB, and Maurice DH
- Subjects
- Mice, Animals, 3T3-L1 Cells, Cell Differentiation, Docosahexaenoic Acids, PPAR gamma, Adipogenesis, Cyclic Nucleotide Phosphodiesterases, Type 4
- Abstract
Recent evidence indicates that the presence of a primary cilium (PC), and of selective cAMP signaling within this smallest of organelles, promotes adipogenic differentiation of 3T3-L1 preadipocytes incubated in media supplemented with either a natural (docosahexaenoic acid, DHA), or a synthetic (TUG-891), free fatty acid receptor 4 (FFAR4) agonist. Indeed, in this earlier work, activation of ciliary FFAR4 in 3T3-L1 cells was correlated with selective increases in PC cAMP and adipogenesis in these cells. However, this study was silent on the role of local PC cAMP phosphodiesterases (PDEs)-mediated events in regulating these adipogenic responses and on the identity of cAMP PDEs that could regulate the "pool" of ciliary cAMP accessed by FFAR4 agonists. In this context, we have identified the PDEs expressed by 3T3-L1 preadipocytes and showed that of these, only PDE4 inhibition promotes FFAR4-mediated adipogenesis. We propose that this work will identify more selective therapeutic targets through which to control adipogenesis, and perhaps the differentiation of other stem cells in which ciliary cAMP is critical., Competing Interests: Declaration of Competing Interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: Donald Maurice reports financial support was provided by Canadian Institutes of Health Research. If there are other authors, they declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
5. Macrophage-NLRP3 Activation Promotes Right Ventricle Failure in Pulmonary Arterial Hypertension.
- Author
-
Al-Qazazi R, Lima PDA, Prisco SZ, Potus F, Dasgupta A, Chen KH, Tian L, Bentley RET, Mewburn J, Martin AY, Wu D, Jones O, Maurice DH, Bonnet S, Provencher S, Prins KW, and Archer SL
- Subjects
- Animals, Atrial Natriuretic Factor, Cytokine Receptor gp130, Disease Models, Animal, Familial Primary Pulmonary Hypertension, Fibrosis, Heart Ventricles, Hypertrophy, Right Ventricular etiology, Inflammasomes, Macrophage Activation, Macrophages metabolism, Monocrotaline, NLR Family, Pyrin Domain-Containing 3 Protein, Rats, Heart Failure, Hypertension, Pulmonary, Pulmonary Arterial Hypertension etiology, Ventricular Dysfunction, Right
- Abstract
Rationale: Pulmonary arterial hypertension (PAH) often results in death from right ventricular failure (RVF). NLRP3 (nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3)-macrophage activation may promote RVF in PAH. Objectives: Evaluating the contribution of the NLRP3 inflammasome in RV macrophages to PAH RVF. Methods: Rats with decompensated RV hypertrophy (monocrotaline [MCT] and Sugen-5416 hypoxia [SuHx]) were compared with compensated RV hypertrophy rats (pulmonary artery banding). Echocardiography and right heart catheterization were performed. Macrophages, atrial natriuretic peptides, and fibrosis were evaluated by microscopy or flow cytometry. NLRP3 inflammasome activation and cardiotoxicity were confirmed by immunoblot and in vitro strategies. MCT rats were treated with SC-144 (a GP130 antagonist) or MCC950 (an NLRP3 inhibitor). Macrophage-NLRP3 activity was evaluated in patients with PAH RVF. Measurements and Main Results: Macrophages, fibrosis, and atrial natriuretic peptides were increased in MCT and SuHx RVs but not in left ventricles or pulmonary artery banding rats. Although MCT RV macrophages were inflammatory, lung macrophages were antiinflammatory. CCR2
+ macrophages (monocyte-derived) were increased in MCT and SuHx RVs and highly expressed NLRP3. The macrophage-NLRP3 pathway was upregulated in patients with PAH with decompensated RVs. Cultured MCT monocytes showed NLRP3 activation, and in coculture experiments resulted in cardiomyocyte mitochondrial damage, which MCC950 prevented. In vivo , MCC950 reduced NLRP3 activation and regressed pulmonary vascular disease and RVF. SC-144 reduced RV macrophages and NLRP3 content, prevented STAT3 (signal transducer and activator of transcription 3) activation, and improved RV function without regressing pulmonary vascular disease. Conclusions: NLRP3-macrophage activation occurs in the decompensated RV in preclinical PAH models and patients with PAH. Inhibiting GP130 or NLRP3 signaling improves RV function. The concept that PAH RVF results from RV inflammation rather than solely from elevated RV afterload suggests a new therapeutic paradigm.- Published
- 2022
- Full Text
- View/download PDF
6. Phosphodiesterase 4D7 selectively regulates cAMP-mediated control of human arterial endothelial cell transcriptomic responses to fluid shear stress.
- Author
-
Burke-Kleinman J and Maurice DH
- Abstract
Human arterial endothelial cells (HAECs) regulate their phenotype by integrating signals encoded in the frictional forces exerted by flowing blood, fluid shear stress (FSS). High laminar FSS promotes establishment of adaptive HAEC phenotype protective against atherosclerosis, whereas low or disturbed FSS cause HAECs to adopt atheroprone phenotypes. A vascular endothelial cadherin (VE cadherin)-based mechanosensory complex allows HAECs to regulate barrier function, cell morphology,/ and gene expression in response to FSS. Previously, we reported that this mechanosensor integrated exchange protein activated by cAMP (EPAC1) and a PDE4D gene derived cyclic nucleotide phosphodiesterase (PDE), but had not identified the PDE4D variant involved. Our hypothesis here was that only one of the two ∼100 kDa PDE4D variants expressed in HAECs coordinated these responses. Now, we show one unique PDE4D splice variant, PDE4D7, controls transcriptional responses of HAECs to FSS while another, PDE4D5, does not. Adaptive transcriptional responses of HAECs subjected to laminar FSS in vitro were blunted in cells in which PDE4D7 was silenced, but unaffected in cells with silenced PDE4D5. This work identifies a specific therapeutic target for the treatment or prevention of atherosclerosis and improves our understanding of the role of cAMP signaling in modulating mechanosensory signal transduction in the vascular endothelium.
- Published
- 2021
- Full Text
- View/download PDF
7. Phosphodiesterase 1C integrates store-operated calcium entry and cAMP signaling in leading-edge protrusions of migrating human arterial myocytes.
- Author
-
Brzezinska P, Simpson NJ, Hubert F, Jacobs AN, Umana MB, MacKeil JL, Burke-Kleinman J, Payne DM, Ferguson AV, and Maurice DH
- Subjects
- Biological Transport, Cell Movement, Gene Expression Regulation, Enzymologic, Humans, Kinetics, Arteries cytology, Calcium metabolism, Cyclic AMP metabolism, Cyclic Nucleotide Phosphodiesterases, Type 1 metabolism, Muscle Cells cytology, Signal Transduction
- Abstract
In addition to maintaining cellular ER Ca
2+ stores, store-operated Ca2+ entry (SOCE) regulates several Ca2+ -sensitive cellular enzymes, including certain adenylyl cyclases (ADCYs), enzymes that synthesize the secondary messenger cyclic AMP (cAMP). Ca2+ , acting with calmodulin, can also increase the activity of PDE1-family phosphodiesterases (PDEs), which cleave the phosphodiester bond of cAMP. Surprisingly, SOCE-regulated cAMP signaling has not been studied in cells expressing both Ca2+ -sensitive enzymes. Here, we report that depletion of ER Ca2+ activates PDE1C in human arterial smooth muscle cells (HASMCs). Inhibiting the activation of PDE1C reduced the magnitude of both SOCE and subsequent Ca2+ /calmodulin-mediated activation of ADCY8 in these cells. Because inhibiting or silencing Ca2+ -insensitive PDEs had no such effects, these data identify PDE1C-mediated hydrolysis of cAMP as a novel and important link between SOCE and its activation of ADCY8. Functionally, we showed that PDE1C regulated the formation of leading-edge protrusions in HASMCs, a critical early event in cell migration. Indeed, we found that PDE1C populated the tips of newly forming leading-edge protrusions in polarized HASMCs, and co-localized with ADCY8, the Ca2+ release activated Ca2+ channel subunit, Orai1, the cAMP-effector, protein kinase A, and an A-kinase anchoring protein, AKAP79. Because this polarization could allow PDE1C to control cAMP signaling in a hyper-localized manner, we suggest that PDE1C-selective therapeutic agents could offer increased spatial specificity in HASMCs over agents that regulate cAMP globally in cells. Similarly, such agents could also prove useful in regulating crosstalk between Ca2+ /cAMP signaling in other cells in which dysregulated migration contributes to human pathology, including certain cancers., Competing Interests: Conflict of interest None declared., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
8. Formulation parameters governing sustained protein delivery from degradable viscous liquid aliphatic polycarbonates.
- Author
-
Mohajeri S, Burke-Kleinman J, Maurice DH, and Amsden BG
- Subjects
- Drug Delivery Systems, Polycarboxylate Cement, Viscosity, Polymers, Vascular Endothelial Growth Factor A
- Abstract
Viscous liquid degradable polymers have advantages as drug depots for sustained protein delivery. We have created a new aliphatic polycarbonate for this purpose, poly(trimethylene carbonate-co-5-hydroxy trimethylene carbonate), which upon degradation retains a near neutral micro-environmental pH. As such, this copolymer is highly suited to the delivery of acid sensitive proteins. We show that the mechanism of protein release from this liquid copolymer is consistent with the formation of super-hydrated regions as a result of the osmotic activity of the solution formed upon distributed protein particle dissolution. Protein release can be manipulated by controlling polymer hydrophobicity which can be adjusted by molecular weight and choice of initiator. Moreover, protein release is highly dependent on protein solubility which impacts the osmotic activity of the solution formed upon dissolution of the protein particles while protein molecular size and isoelectric point are not as influential. As demonstrated by the release of highly bioactive vascular endothelial growth factor, formulations of this copolymer are suitable for prolonged delivery of protein therapeutics., (Copyright © 2020 Elsevier B.V. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
9. Endothelial BMPR2 Loss Drives a Proliferative Response to BMP (Bone Morphogenetic Protein) 9 via Prolonged Canonical Signaling.
- Author
-
Theilmann AL, Hawke LG, Hilton LR, Whitford MKM, Cole DV, Mackeil JL, Dunham-Snary KJ, Mewburn J, James PD, Maurice DH, Archer SL, and Ormiston ML
- Subjects
- Adult, Aged, Animals, Bone Morphogenetic Protein Receptors, Type II genetics, Case-Control Studies, Cells, Cultured, Endothelial Cells metabolism, Endothelial Cells pathology, Female, Growth Differentiation Factor 2 toxicity, Humans, Inhibitor of Differentiation Proteins genetics, Inhibitor of Differentiation Proteins metabolism, Male, Mice, Inbred C57BL, Mice, Knockout, Middle Aged, Pulmonary Arterial Hypertension genetics, Pulmonary Arterial Hypertension metabolism, Pulmonary Arterial Hypertension pathology, Signal Transduction, Young Adult, Bone Morphogenetic Protein Receptors, Type II deficiency, Cell Proliferation drug effects, Endothelial Cells drug effects, Growth Differentiation Factor 2 pharmacology, Pulmonary Arterial Hypertension drug therapy
- Abstract
Objective: Pulmonary arterial hypertension is a disease of proliferative vascular occlusion that is strongly linked to mutations in BMPR2 -the gene encoding the BMPR-II (BMP [bone morphogenetic protein] type II receptor). The endothelial-selective BMPR-II ligand, BMP9, reverses disease in animal models of pulmonary arterial hypertension and suppresses the proliferation of healthy endothelial cells. However, the impact of BMPR2 loss on the antiproliferative actions of BMP9 has yet to be assessed. Approach and Results: BMP9 suppressed proliferation in blood outgrowth endothelial cells from healthy control subjects but increased proliferation in blood outgrowth endothelial cells from pulmonary arterial hypertension patients with BMPR2 mutations. This shift from growth suppression to enhanced proliferation was recapitulated in control human pulmonary artery endothelial cells following siRNA-mediated BMPR2 silencing, as well as in mouse pulmonary endothelial cells isolated from endothelial-conditional Bmpr2 knockout mice ( Bmpr2
EC -/- ). BMP9-induced proliferation was not attributable to altered metabolic activity or elevated TGFβ (transforming growth factor beta) signaling but was linked to the prolonged induction of the canonical BMP target ID1 in the context of BMPR2 loss. In vivo, daily BMP9 administration to neonatal mice impaired both retinal and lung vascular patterning in control mice ( Bmpr2EC+/+ ) but had no measurable effect on mice bearing a heterozygous endothelial Bmpr2 deletion ( Bmpr2EC +/- ) and caused excessive angiogenesis in both vascular beds for Bmpr2EC -/- mice., Conclusions: BMPR2 loss reverses the endothelial response to BMP9, causing enhanced proliferation. This finding has potential implications for the proposed translation of BMP9 as a treatment for pulmonary arterial hypertension and suggests the need for focused patient selection in clinical trials.- Published
- 2020
- Full Text
- View/download PDF
10. Liquid Degradable Poly(trimethylene-carbonate- co -5-hydroxy-trimethylene carbonate): An Injectable Drug Delivery Vehicle for Acid-Sensitive Drugs.
- Author
-
Mohajeri S, Chen F, de Prinse M, Phung T, Burke-Kleinman J, Maurice DH, and Amsden BG
- Subjects
- 3T3 Cells, Animals, Carbon Dioxide chemistry, Chemistry, Pharmaceutical methods, Drug Delivery Systems methods, Excipients chemistry, Fibroblasts drug effects, Fibroblasts metabolism, Hydrogen-Ion Concentration, Macrophages drug effects, Macrophages metabolism, Mice, RAW 264.7 Cells, Vascular Endothelial Growth Factor A metabolism, Viscosity, Carbonates chemistry, Dioxanes chemistry, Pharmaceutical Preparations chemistry, Polymers chemistry, Water chemistry
- Abstract
Liquid, injectable hydrophobic polymers have advantages as degradable drug delivery vehicles; however, polymers examined for this purpose to date form acidic degradation products that may damage acid-sensitive drugs. Herein, we report on a new viscous liquid vehicle, poly(trimethylene carbonate- co -5-hydroxy-trimethylene carbonate), which degrades through intramolecular cyclization producing glycerol, carbon dioxide, and water-soluble trimethylene carbonate. Copolymer degradation durations from weeks to months were achieved with the 5-hydroxy-trimethylene carbonate (HTMC) content of the oligomer having the greatest impact on the degradation rate, with oligomers possessing a higher HTMC content degrading fastest. The degradation products were non-cytotoxic towards 3T3 fibroblasts and RAW 264.7 macrophages. These copolymers can be injected manually through standard gauge needles and, importantly, during in vitro degradation, the microenvironmental pH within the oligomers remained near neutral. Complete and sustained release of the acid-sensitive protein vascular endothelial growth factor was achieved, with the protein remaining highly bioactive throughout the release period. These copolymers represent a promising formulation for local and sustained release of acid sensitive drugs.
- Published
- 2020
- Full Text
- View/download PDF
11. An EPAC1/PDE1C-Signaling Axis Regulates Formation of Leading-Edge Protrusion in Polarized Human Arterial Vascular Smooth Muscle Cells.
- Author
-
Brzezinska P and Maurice DH
- Subjects
- Aorta, Thoracic drug effects, Cells, Cultured, Cyclic Nucleotide Phosphodiesterases, Type 1 antagonists & inhibitors, Guanine Nucleotide Exchange Factors antagonists & inhibitors, Humans, Myocytes, Smooth Muscle drug effects, Quinolines pharmacology, RNA, Small Interfering pharmacology, Aorta, Thoracic metabolism, Cyclic Nucleotide Phosphodiesterases, Type 1 metabolism, Guanine Nucleotide Exchange Factors metabolism, Myocytes, Smooth Muscle metabolism, Signal Transduction drug effects
- Abstract
Pharmacological activation of protein kinase A (PKA) reduces migration of arterial smooth muscle cells (ASMCs), including those isolated from human arteries (HASMCs). However, when individual migration-associated cellular events, including the polarization of cells in the direction of movement or rearrangements of the actin cytoskeleton, are studied in isolation, these individual events can be either promoted or inhibited in response to PKA activation. While pharmacological inhibition or deficiency of exchange protein activated by cAMP-1 (EPAC1) reduces the overall migration of ASMCs, the impact of EPAC1 inhibition or deficiency, or of its activation, on individual migration-related events has not been investigated. Herein, we report that EPAC1 facilitates the formation of leading-edge protrusions (LEPs) in HASMCs, a critical early event in the cell polarization that underpins their migration. Thus, RNAi-mediated silencing, or the selective pharmacological inhibition, of EPAC1 decreased the formation of LEPs by these cells. Furthermore, we show that the ability of EPAC1 to promote LEP formation by migrating HASMCs is regulated by a phosphodiesterase 1C (PDE1C)-regulated "pool" of intracellular HASMC cAMP but not by those regulated by the more abundant PDE3 or PDE4 activities. Overall, our data are consistent with a role for EPAC1 in regulating the formation of LEPs by polarized HASMCs and show that PDE1C-mediated cAMP hydrolysis controls this localized event.
- Published
- 2019
- Full Text
- View/download PDF
12. Phosphodiesterase 3B (PDE3B) antagonizes the anti-angiogenic actions of PKA in human and murine endothelial cells.
- Author
-
MacKeil JL, Brzezinska P, Burke-Kleinman J, Theilmann AL, Nicol CJB, Ormiston ML, and Maurice DH
- Subjects
- 3',5'-Cyclic-AMP Phosphodiesterases metabolism, 8-Bromo Cyclic Adenosine Monophosphate analogs & derivatives, 8-Bromo Cyclic Adenosine Monophosphate metabolism, Animals, Cyclic AMP genetics, Humans, Mice, Neovascularization, Pathologic metabolism, Neovascularization, Pathologic pathology, Phosphodiesterase 3 Inhibitors pharmacology, Cyclic AMP-Dependent Protein Kinases genetics, Cyclic Nucleotide Phosphodiesterases, Type 3 genetics, Endothelial Cells metabolism, Neovascularization, Pathologic genetics
- Abstract
Recent reports show that protein kinase A (PKA), but not exchange protein activated by cAMP (EPAC), acts in a cell autonomous manner to constitutively reduce the angiogenic sprouting capacity of murine and human endothelial cells. Specificity in the cellular actions of individual cAMP-effectors can be achieved when a cyclic nucleotide phosphodiesterase (PDE) enzyme acts locally to control the "pool" of cAMP that activates the cAMP-effector. Here, we examined whether PDEs coordinate the actions of PKA during endothelial cell sprouting. Inhibiting each of the cAMP-hydrolyzing PDEs expressed in human endothelial cells revealed that phosphodiesterase 3 (PDE3) inhibition with cilostamide reduced angiogenic sprouting in vitro, while inhibitors of PDE2 and PDE4 family enzymes had no such effect. Identifying a critical role for PDE3B in the anti-angiogenic effects of cilostamide, silencing this PDE3 variant, but not PDE3A, markedly impaired sprouting. Importantly, using both in vitro and ex vivo models of angiogenesis, we show the hypo-sprouting phenotype induced by PDE3 inhibition or PDE3B silencing was reversed by PKA inhibition. Examination of the individual cellular events required for sprouting revealed that PDE3B and PKA each regulated angiogenic sprouting by controlling the invasive capacity of endothelial cells, more specifically, by regulating podosome rosette biogenesis and matrix degradation. In support of the idea that PDE3B acts to inhibit angiogenic sprouting by limiting PKA-mediated reductions in active cdc42, the effects of PDE3B and/or PKA on angiogenic sprouting were negated in cells with reduced cdc42 expression or activity. Since PDE3B and PKA were co-localized in a perinuclear region in human ECs, could be co-immunoprecipitated from lysates of these cells, and silencing PDE3B activated the perinuclear pool of PKA in these cells, we conclude that PDE3B-mediated hydrolysis of cAMP acts to limit the anti-angiogenic potential of PKA in ECs., (Copyright © 2019 Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
13. A PKA/cdc42 Signaling Axis Restricts Angiogenic Sprouting by Regulating Podosome Rosette Biogenesis and Matrix Remodeling.
- Author
-
MacKeil JL, Brzezinska P, Burke-Kleinman J, Craig AW, Nicol CJB, and Maurice DH
- Subjects
- Cell Line, Cyclic AMP-Dependent Protein Kinases antagonists & inhibitors, Endothelial Cells cytology, Endothelial Cells pathology, Humans, Neovascularization, Physiologic physiology, cdc42 GTP-Binding Protein antagonists & inhibitors, rho Guanine Nucleotide Dissociation Inhibitor alpha pharmacology, src-Family Kinases antagonists & inhibitors, src-Family Kinases physiology, Cyclic AMP-Dependent Protein Kinases physiology, Endothelial Cells metabolism, Neovascularization, Pathologic, Neovascularization, Physiologic drug effects, Podosomes drug effects, cdc42 GTP-Binding Protein physiology
- Abstract
Angiogenic sprouting can contribute adaptively, or mal-adaptively, to a myriad of conditions including ischemic heart disease and cancer. While the cellular and molecular systems that regulate tip versus stalk endothelial cell (EC) specification during angiogenesis are known, those systems that regulate their distinct actions remain poorly understood. Pre-clinical and clinical findings support sustained adrenergic signaling in promoting angiogenesis, but links between adrenergic signaling and angiogenesis are lacking; importantly, adrenergic agents alter the activation status of the cAMP signaling system. Here, we show that the cAMP effector, PKA, acts in a cell autonomous fashion to constitutively reduce the in vitro and ex vivo angiogenic sprouting capacity of ECs. At a cellular level, we observed that silencing or inhibiting PKA in human ECs increased their invasive capacity, their generation of podosome rosettes and, consequently, their ability to degrade a collagen matrix. While inhibition of either Src-family kinases or of cdc42 reduced these events in control ECs, only cdc42 inhibition, or silencing, significantly impacted them in PKA(Cα)-silenced ECs. Consistent with these findings, cell-based measurements of cdc42 activity revealed that PKA activation inhibits EC cdc42 activity, at least in part, by promoting its interaction with the inhibitory regulator, guanine nucleotide dissociation inhibitor-α (RhoGDIα).
- Published
- 2019
- Full Text
- View/download PDF
14. Abnormal angiogenesis in blood outgrowth endothelial cells derived from von Willebrand disease patients.
- Author
-
Selvam SN, Casey LJ, Bowman ML, Hawke LG, Longmore AJ, Mewburn J, Ormiston ML, Archer SL, Maurice DH, and James P
- Subjects
- Animals, Case-Control Studies, Humans, Rabbits, von Willebrand Diseases genetics, von Willebrand Diseases metabolism, Angiogenesis Inducing Agents blood, Endothelial Cells metabolism, von Willebrand Diseases blood
- Abstract
: Bleeding associated with angiodysplasia is a common, often intractable complication in patients with von Willebrand disease (VWD). von Willebrand factor (VWF), the protein deficient or defective in VWD, is a negative regulator of angiogenesis, which may explain the pathologic blood vessel growth in VWD. This study explores the normal range of angiogenesis in blood outgrowth endothelial cells (BOECs) derived from healthy donors and compares this to angiogenesis in BOECs from VWD patients of all types and subtypes. BOECs were assessed for VWF and angiopoietin-2 (Ang-2) gene expression, secretion, and storage. To explore angiogenic potential, we characterized cellular proliferation, matrix protein adhesion, migration, and tubule formation. We found great angiogenic variability in VWD BOECs with respect to each of the angiogenesis parameters. However, type 1 and 3 VWD BOECs had higher Ang-2 secretion associated with impaired endothelial cell migration velocity and enhanced directionality. Type 2A and 2B BOECs were the most proliferative and multiple VWD BOECs had impaired tubule formation in Matrigel. This study highlights the angiogenic variability in BOECs derived from VWD patients. Abnormal cell proliferation, migration, and increased Ang-2 secretion are common features of VWD BOECs. Despite the many abnormalities of VWD BOECs, significant heterogeneity among individual VWD phenotypes precludes a simple description of relationship between VWD type and in vitro surrogates for angiodysplasia.
- Published
- 2017
- Full Text
- View/download PDF
15. Distinct phosphodiesterase 5A-containing compartments allow selective regulation of cGMP-dependent signalling in human arterial smooth muscle cells.
- Author
-
Wilson LS, Guo M, Umana MB, and Maurice DH
- Subjects
- Atrial Natriuretic Factor pharmacology, Cell Compartmentation, Cell Movement drug effects, Cell Proliferation drug effects, Cyclic AMP metabolism, Cyclic Nucleotide Phosphodiesterases, Type 3 metabolism, Enzyme Activation drug effects, Humans, Models, Biological, Myocytes, Smooth Muscle drug effects, Phosphorylation drug effects, Sildenafil Citrate pharmacology, Arteries cytology, Cyclic GMP metabolism, Cyclic Nucleotide Phosphodiesterases, Type 5 metabolism, Myocytes, Smooth Muscle metabolism, Signal Transduction drug effects
- Abstract
Cyclic GMP (cGMP) translates and integrates much of the information encoded by nitric oxide (NO
· ) and several natriuretic peptides, including the atrial natriuretic peptide (ANP). Previously, we reported that integration of a cGMP-specific cyclic nucleotide phosphodiesterase, namely phosphodiesterase 5A (PDE5A), into a protein kinase G (PKG)- and inositol-1,4,5-trisphosphate receptor (IP3 R)-containing endoplasmic reticulum (ER) signalosome allows localized control of PDE5A activity and of PKG-dependent inhibition of IP3 -mediated release of ER Ca2+ in human platelets. Herein, we report that PDE5A integrates into an analogous signalosome in human arterial smooth muscle cells (HASMC), wherein it regulates muscarinic agonist-dependent Ca2+ release and is activated selectively by PKG-dependent phosphorylation. In addition, we report that PDE5A also regulates HASMC functions via events independent of PKG, but rather through actions coordinated by competitive cGMP-mediated inhibition of cAMP hydrolysis by the so-called cGMP-inhibited cAMP PDE, namely phosphodiesterase 3A (PDE3A). Indeed, we show that ANP increases both cGMP and cAMP levels in HASMC and promotes phosphorylation of vasodilator-stimulated phospho-protein (VASP) at each the PKG and PKA phospho-acceptor sites. Since selective inhibition of PDE5 decreased DNA synthesis and chemotaxis of HASMC, and that PDE3A knockdown obviated these effects, our findings are consistent with a role for a PDE5A-PDE3A-PKA axis in their regulation. Our findings provide insight into the existence of distinct "pools" of PDE5A in HASMC and support the idea that these discrete compartments regulate distinct cGMP-dependent events. As a corollary, we suggest that it may be possible to target these distinct PDE5A-regulated pools and in so-doing differentially impact selected cGMP-regulated functions in these cells., (Copyright © 2017. Published by Elsevier Inc.)- Published
- 2017
- Full Text
- View/download PDF
16. Adaptive phenotypic modulation of human arterial endothelial cells to fluid shear stress-encoded signals: modulation by phosphodiesterase 4D-VE-cadherin signalling.
- Author
-
Rampersad SN, Wudwud A, Hubert F, and Maurice DH
- Subjects
- Cell Shape, Cyclic AMP metabolism, Gene Expression Regulation, Gene Knockdown Techniques, Humans, Intercellular Junctions metabolism, Mechanotransduction, Cellular, Phenotype, Platelet Endothelial Cell Adhesion Molecule-1 metabolism, Vascular Endothelial Growth Factor Receptor-2 metabolism, Antigens, CD metabolism, Aorta cytology, Cadherins metabolism, Cyclic Nucleotide Phosphodiesterases, Type 4 metabolism, Endothelial Cells metabolism, Shear Strength, Signal Transduction
- Abstract
Although cAMP-signalling regulates numerous functions of vascular endothelial cells (VECs), including their ability to impact vascular resistance in response to changes in blood flow dynamics, few of the mechanisms underlying these effects have yet to be described. In addition to forming stable adherens junctions (AJs) in static VEC cultures, VE-cadherin (VECAD) has emerged as a critical component in a key mechanosensor responsible for linking altered blood flow dynamics and the VEC-mediated control of vascular resistance. Previously, a cAMP phosphodiesterase, PDE4D, was shown to coordinate the VEC permeability limiting effects of cAMP-elevating agents in human arterial VECs (HAECs). Herein, we report that PDE4D acts to allow cAMP-elevating agents to regulate VECADs' role as a sensor of flow-associated fluid shear stress (FSS)-encoded information in HAECs. Thus, we report that PDE4 activity is increased in HAECs exposed to laminar FSS and that this effect contributes to controlling how FSS impacts the morphological and gene expression changes in HAECs exposed to flow. More specifically, we report that PDE4D regulates the efficiency with which VECAD, within its mechanosensor, controls VEGFR2 and Akt activities. Indeed, we show that PDE4D knockdown (KD) significantly blunts responses of HAECs to levels of FSS characteristically found in areas of the vasculature in which stenosis is prevalent. We propose that this effect may provide a new therapeutic avenue in modulating VEC behaviour at these sites by promoting an adaptive and vasculo-protective phenotype., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
17. EPAC1 promotes adaptive responses in human arterial endothelial cells subjected to low levels of laminar fluid shear stress: Implications in flow-related endothelial dysfunction.
- Author
-
Rampersad SN, Freitag SI, Hubert F, Brzezinska P, Butler N, Umana MB, Wudwud AR, and Maurice DH
- Subjects
- Adaptation, Physiological, Arteries cytology, Cells, Cultured, Endothelial Cells cytology, Endothelium, Vascular cytology, Endothelium, Vascular physiopathology, Gene Expression, Guanine Nucleotide Exchange Factors antagonists & inhibitors, Humans, Stress, Mechanical, Endothelial Cells metabolism, Endothelium, Vascular metabolism, Guanine Nucleotide Exchange Factors metabolism, Mechanotransduction, Cellular
- Abstract
Blood flow-associated fluid shear stress (FSS) dynamically regulates the endothelium's ability to control arterial structure and function. While arterial endothelial cells (AEC) subjected to high levels of laminar FSS express a phenotype resistant to vascular insults, those exposed to low levels of laminar FSS, or to the FSS associated with oscillatory blood flow, are less resilient. Despite numerous reports highlighting how the cAMP-signaling system controls proliferation, migration and permeability of human AECs (HAECs), its role in coordinating HAEC responses to FSS has received scant attention. Herein we show that the cAMP effector EPAC1 is required for HAECs to align and elongate in the direction of flow, and for the induction of several anti-atherogenic and anti-thrombotic genes associated with these events. Of potential therapeutic importance, EPAC1 is shown to play a dominant role the in response of HAECs to low levels of laminar FSS, such as would be found within atherosclerosis-prone areas of the vasculature. Moreover, we show that EPAC1 promotes these HAEC responses to flow by regulating Vascular Endothelial Growth Factor Receptor-2 and Akt activation, within a VE-cadherin (VECAD)/PECAM1-based mechanosensor. We submit that these findings are consistent with the novel proposition that promoting EPAC1-signaling represents a novel means through which to promote expression of an adaptive phenotype in HAECs exposed to non-adaptive FSS-encoded signals as a consequence of vascular disease., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
18. Leptin influences the excitability of area postrema neurons.
- Author
-
Smith PM, Brzezinska P, Hubert F, Mimee A, Maurice DH, and Ferguson AV
- Subjects
- Action Potentials, Animals, Area Postrema cytology, Area Postrema metabolism, Cells, Cultured, Cyclic AMP metabolism, Energy Metabolism drug effects, In Vitro Techniques, Islet Amyloid Polypeptide pharmacology, Male, Neurons metabolism, RNA, Messenger metabolism, Rats, Sprague-Dawley, Receptors, Leptin genetics, Receptors, Leptin metabolism, Second Messenger Systems drug effects, Time Factors, Area Postrema drug effects, Leptin pharmacology, Neurons drug effects, Receptors, Leptin agonists
- Abstract
The area postrema (AP) is a circumventricular organ with important roles in central autonomic regulation. This medullary structure has been shown to express the leptin receptor and has been suggested to have a role in modulating peripheral signals, indicating energy status. Using RT-PCR, we have confirmed the presence of mRNA for the leptin receptor, ObRb, in AP, and whole cell current-clamp recordings from dissociated AP neurons demonstrated that leptin influenced the excitability of 51% (42/82) of AP neurons. The majority of responsive neurons (62%) exhibited a depolarization (5.3 ± 0.7 mV), while the remaining affected cells (16/42) demonstrated hyperpolarizing effects (-5.96 ± 0.95 mV). Amylin was found to influence the same population of AP neurons. To elucidate the mechanism(s) of leptin and amylin actions in the AP, we used fluorescence resonance energy transfer (FRET) to determine the effect of these peptides on cAMP levels in single AP neurons. Leptin and amylin were found to elevate cAMP levels in the same dissociated AP neurons (leptin: % total FRET response 25.3 ± 4.9, n = 14; amylin: % total FRET response 21.7 ± 3.1, n = 13). When leptin and amylin were coapplied, % total FRET response rose to 53.0 ± 8.3 (n = 6). The demonstration that leptin and amylin influence a subpopulation of AP neurons and that these two signaling molecules have additive effects on single AP neurons to increase cAMP, supports a role for the AP as a central nervous system location at which these circulating signals may act through common intracellular signaling pathways to influence central control of energy balance., (Copyright © 2016 the American Physiological Society.)
- Published
- 2016
- Full Text
- View/download PDF
19. Cyclic nucleotide-based therapeutics for chronic obstructive pulmonary disease.
- Author
-
Giembycz MA and Maurice DH
- Subjects
- Adrenal Cortex Hormones therapeutic use, Adrenergic beta-2 Receptor Agonists therapeutic use, Animals, Humans, Nucleotides, Cyclic therapeutic use, Phosphodiesterase Inhibitors therapeutic use, Pulmonary Disease, Chronic Obstructive drug therapy
- Abstract
Chronic obstructive pulmonary disease (COPD) defines a group of chronic inflammatory disorders of the airways that are characterised by a progressive and largely irreversible decline in expiratory airflow. Drugs used to treat COPD through actions mediated by cyclic AMP (cAMP) are restricted to long-acting and short-acting β2-adrenoceptor agonists and, in a subset of patients with chronic bronchitis, a phosphodiesterase 4 inhibitor, roflumilast. These agents relax airway smooth muscle and suppress inflammation. At the molecular level, these effects in the airways are mediated by two cAMP effectors, cAMP-dependent protein kinase and exchange proteins activated by cAMP. The pharmacology of newer agents, acting through these systems, is discussed here with an emphasis on their potential to interact and increase therapeutic effectiveness., (Crown Copyright © 2014. Published by Elsevier Ltd. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
20. Advances in targeting cyclic nucleotide phosphodiesterases.
- Author
-
Maurice DH, Ke H, Ahmad F, Wang Y, Chung J, and Manganiello VC
- Subjects
- Animals, Cyclic AMP metabolism, Cyclic GMP metabolism, Humans, Molecular Targeted Therapy, Signal Transduction drug effects, Drug Design, Phosphodiesterase Inhibitors pharmacology, Phosphoric Diester Hydrolases metabolism
- Abstract
Cyclic nucleotide phosphodiesterases (PDEs) catalyse the hydrolysis of cyclic AMP and cyclic GMP, thereby regulating the intracellular concentrations of these cyclic nucleotides, their signalling pathways and, consequently, myriad biological responses in health and disease. Currently, a small number of PDE inhibitors are used clinically for treating the pathophysiological dysregulation of cyclic nucleotide signalling in several disorders, including erectile dysfunction, pulmonary hypertension, acute refractory cardiac failure, intermittent claudication and chronic obstructive pulmonary disease. However, pharmaceutical interest in PDEs has been reignited by the increasing understanding of the roles of individual PDEs in regulating the subcellular compartmentalization of specific cyclic nucleotide signalling pathways, by the structure-based design of novel specific inhibitors and by the development of more sophisticated strategies to target individual PDE variants.
- Published
- 2014
- Full Text
- View/download PDF
21. Cyclic nucleotide phosphodiesterases (PDEs): coincidence detectors acting to spatially and temporally integrate cyclic nucleotide and non-cyclic nucleotide signals.
- Author
-
Maurice DH, Wilson LS, Rampersad SN, Hubert F, Truong T, Kaczmarek M, Brzezinska P, Freitag SI, Umana MB, and Wudwud A
- Subjects
- Animals, Humans, Signal Transduction physiology, Cyclic AMP metabolism, Cyclic GMP metabolism, Phosphoric Diester Hydrolases metabolism
- Abstract
The cyclic nucleotide second messengers cAMP and cGMP each affect virtually all cellular processes. Although these hydrophilic small molecules readily diffuse throughout cells, it is remarkable that their ability to activate their multiple intracellular effectors is spatially and temporally selective. Studies have identified a critical role for compartmentation of the enzymes which hydrolyse and metabolically inactivate these second messengers, the PDEs (cyclic nucleotide phosphodiesterases), in this specificity. In the present article, we describe several examples from our work in which compartmentation of selected cAMP- or cGMP-hydrolysing PDEs co-ordinate selective activation of cyclic nucleotide effectors, and, as a result, selectively affect cellular functions. It is our belief that therapeutic strategies aimed at targeting PDEs within these compartments will allow greater selectivity than those directed at inhibiting these enzymes throughout the cells.
- Published
- 2014
- Full Text
- View/download PDF
22. PDE8A runs interference to limit PKA inhibition of Raf-1.
- Author
-
Maurice DH
- Subjects
- Animals, Humans, 3',5'-Cyclic-AMP Phosphodiesterases metabolism, MAP Kinase Signaling System physiology, Proto-Oncogene Proteins c-raf metabolism
- Published
- 2013
- Full Text
- View/download PDF
23. Potential therapeutic applications of phosphodiesterase inhibition in prostate cancer.
- Author
-
Hamilton TK, Hu N, Kolomitro K, Bell EN, Maurice DH, Graham CH, and Siemens DR
- Subjects
- 3',5'-Cyclic-GMP Phosphodiesterases, Adenocarcinoma drug therapy, Adenocarcinoma immunology, Animals, Antineoplastic Agents therapeutic use, Cell Line, Tumor, Colony-Forming Units Assay, Cyclic Nucleotide Phosphodiesterases, Type 1 metabolism, Cyclic Nucleotide Phosphodiesterases, Type 2 metabolism, Cyclic Nucleotide Phosphodiesterases, Type 4 metabolism, Cyclic Nucleotide Phosphodiesterases, Type 5 metabolism, Disease Models, Animal, Doxorubicin therapeutic use, Enzyme Assays, Histocompatibility Antigens Class I drug effects, Humans, Male, Mice, Prostatic Neoplasms drug therapy, Prostatic Neoplasms immunology, Tumor Escape physiology, Xenograft Model Antitumor Assays, Adenocarcinoma enzymology, Drug Resistance, Neoplasm drug effects, Histocompatibility Antigens Class I metabolism, Phosphodiesterase Inhibitors pharmacology, Phosphoric Diester Hydrolases metabolism, Prostatic Neoplasms enzymology, Tumor Escape drug effects
- Abstract
Objective: Phosphodiesterases (PDEs) play a role in controlling cyclic nucleotide action, including cyclic guanosine monophosphate (cGMP). Previous studies have ascribed a protective role of cGMP signaling on hypoxia-mediated cancer progression. Herein, we determine their potential role in hypoxia-mediated chemoresistance and immune escape., Materials and Methods: Phosphodiesterase assays were used to measure PDE activity in prostate cancer cell lines (DU145, PC3). Immunoblots were performed to determine the presence of PDEs in human prostate tissue samples. The effect of PDE inhibition on hypoxia-induced chemoresistance (compared to normoxic controls, 20% O2) was determined using clonogenic assays. Flow cytometry was used to determine the effects of PDE inhibition on surface MHC class I-related chain A (MICA), a natural killer (NK) cell-activating ligand. A mouse model was used to evaluate the in vivo effects of PDE inhibition on the growth of human prostate cancer cells., Results: PDE5 and PDE11 were the most prominent PDEs in the cell lines, representing between 86 and 95% of the total cGMP-specific PDE activity. Treatment of DU-145 cells with a PDE inhibitor significantly reduced the hypoxia-associated acquisition of resistance to doxorubicin, with a mean 51% reduction in surviving fraction compared to controls (p < 0.001, ANOVA). As well, PDE inhibition completely reversed (p = 0.02, ANOVA) hypoxia-induced shedding of the immune stimulatory molecule, MICA, and attenuated the growth of human prostate tumor xenografts in an NK cell-competent murine model (p = 0.03, Wilcoxon, Mann-Whitney)., Conclusions: These results suggest a rationale for future studies on the potential therapeutic applications of PDE inhibitors in men with prostate cancer.
- Published
- 2013
- Full Text
- View/download PDF
24. Targeting protein-protein interactions within the cyclic AMP signaling system as a therapeutic strategy for cardiovascular disease.
- Author
-
Lee LC, Maurice DH, and Baillie GS
- Subjects
- Animals, Cardiovascular Diseases metabolism, Cardiovascular System drug effects, Cardiovascular System metabolism, Humans, Peptides therapeutic use, Small Molecule Libraries therapeutic use, Cardiovascular Diseases drug therapy, Cyclic AMP metabolism, Molecular Targeted Therapy methods, Peptides pharmacology, Protein Interaction Maps drug effects, Signal Transduction drug effects, Small Molecule Libraries pharmacology
- Abstract
The cAMP signaling system can trigger precise physiological cellular responses that depend on the fidelity of many protein-protein interactions, which act to bring together signaling intermediates at defined locations within cells. In the heart, cAMP participates in the fine control of excitation-contraction coupling, hence, any disregulation of this signaling cascade can lead to cardiac disease. Due to the ubiquitous nature of the cAMP pathway, general inhibitors of cAMP signaling proteins such as PKA, EPAC and PDEs would act non-specifically and universally, increasing the likelihood of serious 'off target' effects. Recent advances in the discovery of peptides and small molecules that disrupt the protein-protein interactions that underpin cellular targeting of cAMP signaling proteins are described and discussed.
- Published
- 2013
- Full Text
- View/download PDF
25. Subcellular signaling in the endothelium: cyclic nucleotides take their place.
- Author
-
Maurice DH
- Subjects
- 3',5'-Cyclic-AMP Phosphodiesterases metabolism, 3',5'-Cyclic-GMP Phosphodiesterases metabolism, Animals, Cell Adhesion, Cell Membrane Permeability, Cell Movement, Cyclic AMP physiology, Cyclic GMP physiology, Endothelium, Vascular cytology, Endothelium, Vascular enzymology, Guanine Nucleotide Exchange Factors metabolism, Humans, Isoenzymes metabolism, rap1 GTP-Binding Proteins metabolism, ras Guanine Nucleotide Exchange Factors metabolism, Endothelium, Vascular physiology, Nucleotides, Cyclic physiology, Second Messenger Systems
- Abstract
When lecturing on the topic of cellular signaling I have had occasion to ask the class for examples of cellular processes NOT impacted by cyclic AMP (cAMP) and am struck by how few examples exist. Indeed, studies spanning the past 60 years have detailed how this ubiquitous second messenger impacts virtually all cellular processes, including intermediary metabolism, contractility, motility, proliferation, and gene expression in most mammalian cells. Since the hydrophobic cAMP could in principle diffuse rapidly throughout the cell once formed, the remarkable spatial and temporal specificity of its numerous actions in cells is truly impressive. Herein I introduce the main players involved in coordinating actions of cAMP in vascular endothelial cells (VECs), and focus on the increasing awareness of the dominant role that cyclic nucleotide phosphodiesterases (PDEs), the sole cellular enzymes capable of hydrolytically inactivating cAMP, play in fostering this specificity., (Copyright © 2011 Elsevier Ltd. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
26. Phosphodiesterase 4D regulates baseline sarcoplasmic reticulum Ca2+ release and cardiac contractility, independently of L-type Ca2+ current.
- Author
-
Beca S, Helli PB, Simpson JA, Zhao D, Farman GP, Jones P, Tian X, Wilson LS, Ahmad F, Chen SRW, Movsesian MA, Manganiello V, Maurice DH, Conti M, and Backx PH
- Subjects
- Animals, Calcium-Binding Proteins metabolism, Cardiomyopathy, Dilated metabolism, Cardiomyopathy, Dilated pathology, Cyclic AMP metabolism, Cyclic Nucleotide Phosphodiesterases, Type 4 genetics, Female, Heart Ventricles metabolism, Heart Ventricles pathology, Humans, Male, Mice, Mice, Knockout, Models, Animal, Myocytes, Cardiac pathology, Phosphatidylinositol 3-Kinases metabolism, Ryanodine Receptor Calcium Release Channel metabolism, Sarcoplasmic Reticulum Calcium-Transporting ATPases metabolism, Calcium metabolism, Calcium Channels, L-Type physiology, Cyclic Nucleotide Phosphodiesterases, Type 4 metabolism, Myocardial Contraction physiology, Myocytes, Cardiac metabolism, Sarcoplasmic Reticulum metabolism
- Abstract
Rationale: Baseline contractility of mouse hearts is modulated in a phosphatidylinositol 3-kinase-γ-dependent manner by type 4 phosphodiesterases (PDE4), which regulate cAMP levels within microdomains containing the sarcoplasmic reticulum (SR) calcium ATPase type 2a (SERCA2a)., Objective: The goal of this study was to determine whether PDE4D regulates basal cardiac contractility., Methods and Results: At 10 to 12 weeks of age, baseline cardiac contractility in PDE4D-deficient (PDE4D(-/-)) mice was elevated mice in vivo and in Langendorff perfused hearts, whereas isolated PDE4D(-/-) cardiomyocytes showed increased whole-cell Ca2+ transient amplitudes and SR Ca2+content but unchanged L-type calcium current, compared with littermate controls (WT). The protein kinase A inhibitor R(p)-adenosine-3',5' cyclic monophosphorothioate (R(p)-cAMP) lowered whole-cell Ca2+ transient amplitudes and SR Ca2+ content in PDE4D(-/-) cardiomyocytes to WT levels. The PDE4 inhibitor rolipram had no effect on cardiac contractility, whole-cell Ca2+ transients, or SR Ca2+ content in PDE4D(-/-) preparations but increased these parameters in WT myocardium to levels indistinguishable from those in PDE4D(-/-). The functional changes in PDE4D(-/-) myocardium were associated with increased PLN phosphorylation but not cardiac ryanodine receptor phosphorylation. Rolipram increased PLN phosphorylation in WT cardiomyocytes to levels indistinguishable from those in PDE4D(-/-) cardiomyocytes. In murine and failing human hearts, PDE4D coimmunoprecipitated with SERCA2a but not with cardiac ryanodine receptor., Conclusions: PDE4D regulates basal cAMP levels in SR microdomains containing SERCA2a-PLN, but not L-type Ca2+ channels or ryanodine receptor. Because whole-cell Ca2+ transient amplitudes are reduced in failing human myocardium, these observations may have therapeutic implications for patients with heart failure.
- Published
- 2011
- Full Text
- View/download PDF
27. A phosphodiesterase 3B-based signaling complex integrates exchange protein activated by cAMP 1 and phosphatidylinositol 3-kinase signals in human arterial endothelial cells.
- Author
-
Wilson LS, Baillie GS, Pritchard LM, Umana B, Terrin A, Zaccolo M, Houslay MD, and Maurice DH
- Subjects
- Arteries cytology, Arteries metabolism, Cell Adhesion, Cells, Cultured, Cyclic AMP metabolism, Cyclic AMP-Dependent Protein Kinases metabolism, Endothelial Cells cytology, Humans, Cyclic Nucleotide Phosphodiesterases, Type 3 metabolism, Endothelial Cells metabolism, Guanine Nucleotide Exchange Factors metabolism, Neovascularization, Physiologic physiology, Phosphatidylinositol 3-Kinases metabolism
- Abstract
Enzymes of the phosphodiesterase 3 (PDE3) and PDE4 families each regulate the activities of both protein kinases A (PKAs) and exchange proteins activated by cAMP (EPACs) in cells of the cardiovascular system. At present, the mechanisms that allow selected PDEs to individually regulate the activities of these two effectors are ill understood. The objective of this study was to determine how a specific PDE3 variant, namely PDE3B, interacts with and regulates EPAC1-based signaling in human arterial endothelial cells (HAECs). Using several biochemical approaches, we show that PDE3B and EPAC1 bind directly through protein-protein interactions. By knocking down PDE3B expression or by antagonizing EPAC1 binding with PDE3B, we show that PDE3B regulates cAMP binding by its tethered EPAC1. Interestingly, we also show that PDE3B binds directly to p84, a PI3Kγ regulatory subunit, and that this interaction allows PI3Kγ recruitment to the PDE3B-EPAC1 complex. Of potential cardiovascular importance, we demonstrate that PDE3B-tethered EPAC1 regulates HAEC PI3Kγ activity and that this allows dynamic cAMP-dependent regulation of HAEC adhesion, spreading, and tubule formation. We identify and molecularly characterize a PDE3B-based "signalosome" that integrates cAMP- and PI3Kγ-encoded signals and show how this signal integration regulates HAEC functions of importance in angiogenesis.
- Published
- 2011
- Full Text
- View/download PDF
28. Cyclic AMP phosphodiesterase 4D (PDE4D) Tethers EPAC1 in a vascular endothelial cadherin (VE-Cad)-based signaling complex and controls cAMP-mediated vascular permeability.
- Author
-
Rampersad SN, Ovens JD, Huston E, Umana MB, Wilson LS, Netherton SJ, Lynch MJ, Baillie GS, Houslay MD, and Maurice DH
- Subjects
- Amino Acid Motifs, Atherosclerosis metabolism, Cells, Cultured, Cyclic Nucleotide Phosphodiesterases, Type 4, Humans, Intercellular Junctions metabolism, Macromolecular Substances, Peptides chemistry, Permeability, Signal Transduction, beta Catenin metabolism, Antigens, CD metabolism, Cadherins metabolism, Cyclic AMP metabolism, Cyclic Nucleotide Phosphodiesterases, Type 3 metabolism, Endothelium, Vascular metabolism, Guanine Nucleotide Exchange Factors metabolism
- Abstract
Vascular endothelial cell (VEC) permeability is largely dependent on the integrity of vascular endothelial cadherin (VE-cadherin or VE-Cad)-based intercellular adhesions. Activators of protein kinase A (PKA) or of exchange protein activated by cAMP (EPAC) reduce VEC permeability largely by stabilizing VE-Cad-based intercellular adhesions. Currently, little is known concerning the nature and composition of the signaling complexes that allow PKA or EPAC to regulate VE-Cad-based structures and through these actions control permeability. Using pharmacological, biochemical, and cell biological approaches we identified and determined the composition and functionality of a signaling complex that coordinates cAMP-mediated control of VE-Cad-based adhesions and VEC permeability. Thus, we report that PKA, EPAC1, and cyclic nucleotide phosphodiesterase 4D (PDE4D) enzymes integrate into VE-Cad-based signaling complexes in human arterial endothelial cells. Importantly, we show that protein-protein interactions between EPAC1 and PDE4D serve to foster their integration into VE-Cad-based complexes and allow robust local regulation of EPAC1-based stabilization of VE-Cad-based adhesions. Of potential translational importance, we mapped the EPAC1 peptide motif involved in binding PDE4D and show that a cell-permeable variant of this peptide antagonizes EPAC1-PDE4D binding and directly alters VEC permeability. Collectively, our data indicate that PDE4D regulates both the activity and subcellular localization of EPAC1 and identify a novel mechanism for regulated EPAC1 signaling in these cells.
- Published
- 2010
- Full Text
- View/download PDF
29. Distinct phosphodiesterase-4D variants integrate into protein kinase A-based signaling complexes in cardiac and vascular myocytes.
- Author
-
Raymond DR, Carter RL, Ward CA, and Maurice DH
- Subjects
- Actins metabolism, Animals, Cell Line, Cell Movement, Cells, Cultured, Cyclic AMP metabolism, Cyclic Nucleotide Phosphodiesterases, Type 4 genetics, Humans, Isoenzymes, Male, Muscle, Smooth, Vascular drug effects, Myocytes, Cardiac drug effects, Myocytes, Smooth Muscle drug effects, Phosphodiesterase 4 Inhibitors, Phosphodiesterase Inhibitors pharmacology, Pseudopodia enzymology, Rats, Rats, Sprague-Dawley, Recombinant Fusion Proteins metabolism, Transfection, A Kinase Anchor Proteins metabolism, Cyclic AMP-Dependent Protein Kinases metabolism, Cyclic Nucleotide Phosphodiesterases, Type 4 metabolism, Muscle, Smooth, Vascular enzymology, Myocytes, Cardiac enzymology, Myocytes, Smooth Muscle enzymology, Signal Transduction drug effects
- Abstract
Numerous cAMP-elevating agents regulate events required for efficient migration of arterial vascular smooth muscle cells (VSMCs). Interestingly, when the impact of cAMP-elevating agents on individual migration-related events is studied, these agents have been shown to have distinct, and sometimes unexpected, effects. For example, although cAMP-elevating agents inhibit overall migration, they promote VSMC adhesion to extracellular matrix proteins and the formation of membrane extensions, which are both events that are essential for and promote migration. Herein, we extend previous observations that identified phosphodiesterase-4D3 (PDE4D3) as an integral component of a PKA/A kinase-anchoring protein (AKAP) complex in cultured/hypertrophied rat cardiac myocytes to the case for nonhypertrophied cardiac myocytes. Moreover, we show that while rat aortic VSMCs also express PDE4D3, this protein is not detected in PKA/AKAP complexes isolated from these cells. In contrast, we show that another PDE4D splice variant expressed in arterial vascular myocytes, namely, PDE4D8, integrates into PKA/AKAP-based signaling complexes in VSMCs. Consistent with the idea that a PDE4D8/PKA/AKAP complex regulates specific VSMC functions, PKA and PDE4D8 were each recruited to leading-edge structures in migrating VSMCs, and inhibition of PDE4D8 recruitment to pseudopodia of migrating cells caused localized changes in actin dynamics. Our data are presented in the context that cardiac myocytes and arterial VSMCs may use distinct PDE4D variants to regulate selected pools of targeted PKA activity and that disruption of this complex may allow selective regulation of cAMP-dependent events between these two cardiovascular cell types.
- Published
- 2009
- Full Text
- View/download PDF
30. Compartmentation and compartment-specific regulation of PDE5 by protein kinase G allows selective cGMP-mediated regulation of platelet functions.
- Author
-
Wilson LS, Elbatarny HS, Crawley SW, Bennett BM, and Maurice DH
- Subjects
- Blood Platelets drug effects, Calcium metabolism, Endoplasmic Reticulum metabolism, Enzyme Activation, Humans, Inositol 1,4,5-Trisphosphate Receptors metabolism, Nitroprusside pharmacology, Phosphorylation, Piperazines pharmacology, Platelet Aggregation drug effects, Protein Binding, Purines pharmacology, Signal Transduction, Sildenafil Citrate, Substrate Specificity, Sulfones pharmacology, Thrombin metabolism, Blood Platelets metabolism, Cyclic GMP metabolism, Cyclic GMP-Dependent Protein Kinases metabolism, Cyclic Nucleotide Phosphodiesterases, Type 5 metabolism
- Abstract
It is generally accepted that nitric oxide (NO) donors, such as sodium nitroprusside (SNP), or phosphodiesterase 5 (PDE5) inhibitors, including sildenafil, each impact human platelet function. Although a strong correlation exists between the actions of NO donors in platelets and their impact on cGMP, agents such as sildenafil act without increasing global intra-platelet cGMP levels. This study was undertaken to identify how PDE5 inhibitors might act without increasing cGMP. Our data identify PDE5 as an integral component of a protein kinase G1beta (PKG1beta)-containing signaling complex, reported previously to coordinate cGMP-mediated inhibition of inositol-1, 4, 5-trisphosphate receptor type 1 (IP(3)R1)-mediated Ca(2+)-release. PKG1beta and PDE5 did not interact in subcellular fractions devoid of IP(3)R1 and were not recruited to IP(3)R1-enriched membranes in response to cGMP-elevating agents. Activation of platelet PKG promoted phosphorylation and activation of the PDE5 fraction tethered to the IP(3)R1-PKG complex, an effect not observed for the nontethered PDE5. Based on these findings, we elaborate a model in which PKG selectively activates PDE5 within a defined microdomain in platelets and propose that this mechanism allows spatial and temporal regulation of cGMP signaling in these cells. Recent reports indicate that sildenafil might prove useful in limiting in-stent thrombosis and the thrombotic events associated with the acute coronary syndromes (ACS), situations poorly regulated with currently available therapeutics. We submit that our findings may define a molecular mechanism by which PDE5 inhibition can differentially impact selected cellular functions of platelets, and perhaps of other cell types.
- Published
- 2008
- Full Text
- View/download PDF
31. Numerous distinct PKA-, or EPAC-based, signalling complexes allow selective phosphodiesterase 3 and phosphodiesterase 4 coordination of cell adhesion.
- Author
-
Raymond DR, Wilson LS, Carter RL, and Maurice DH
- Subjects
- 4-(3-Butoxy-4-methoxybenzyl)-2-imidazolidinone pharmacology, Adenylyl Cyclases metabolism, Cell Line, Colforsin pharmacology, Cyclic AMP analogs & derivatives, Cyclic AMP metabolism, Cyclic Nucleotide Phosphodiesterases, Type 3 chemistry, Cyclic Nucleotide Phosphodiesterases, Type 3 genetics, Cyclic Nucleotide Phosphodiesterases, Type 4, Enzyme Activators pharmacology, Guanine Nucleotide Exchange Factors genetics, Humans, Multiprotein Complexes metabolism, Peptide Fragments metabolism, Phosphodiesterase 3 Inhibitors, Phosphodiesterase Inhibitors pharmacology, Protein Structure, Tertiary, Quinolones pharmacology, Transfection, Cell Adhesion drug effects, Cyclic AMP-Dependent Protein Kinases metabolism, Cyclic Nucleotide Phosphodiesterases, Type 3 metabolism, Guanine Nucleotide Exchange Factors metabolism, Signal Transduction drug effects
- Abstract
By activating two distinct classes of effector enzymes, namely Protein Kinases A [PKA] or Exchange Proteins Activated by cAMP [EPAC], the ubiquitous second messenger cAMP selectively coordinates numerous events simultaneously in virtually all cells. Studies focused on dissecting the manner by which cAMP simultaneously regulates multiple cellular events have shown that cAMP activates its effectors non-uniformly in cells and that this localized cAMP-mediated signalling is made possible, at least in part, by anchoring of cAMP effectors to selected subcellular structures. In the work described here, we report that HEK293T cells ["293T"] contain several PKA- and EPAC1-based signalling complexes. Interestingly, our data do not identify signalling complexes in which both PKA and EPAC are each present but rather are consistent with the idea that these two effectors operate in distinct complexes in these cells. Similarly, we report that while individual PKA- or EPAC-containing complexes can contain either phosphodiesterase 3B, [PDE3B] or phosphodiesterase 4D [PDE4D], they do not contain both these phosphodiesterases. Indeed, although PDE4D enzymes were identified in both PKA- and EPAC-based complexes, PDE3B was largely identified in EPAC-based complexes. Using a combination of approaches, we identified that integration of PDE3B into EPAC-based complexes occurred through its amino terminal fragment [PDE3B(AT)]. Consistent with the idea that integration of PDE3B within EPAC-based complexes was dynamic and regulated PDE3 inhibitor-mediated effects on cellular functions, expression of PDE3B(AT) competed with endogenous PDE3B for integration into EPAC-based complexes and antagonized PDE3 inhibitor-based cell adhesion. Our data support the concept that cells can contain several non-overlapping PKA- and EPAC-based signalling complexes and that these complexes may also represent sites within cells were the effects of family-selective PDE inhibitors could be integrated to affect cell functions, including adhesion.
- Published
- 2007
- Full Text
- View/download PDF
32. Both protein kinase A and exchange protein activated by cAMP coordinate adhesion of human vascular endothelial cells.
- Author
-
Netherton SJ, Sutton JA, Wilson LS, Carter RL, and Maurice DH
- Subjects
- Antiporters physiology, Cell Adhesion physiology, Cells, Cultured, Cyclic AMP-Dependent Protein Kinases physiology, Endothelial Cells enzymology, Endothelium, Vascular cytology, Endothelium, Vascular enzymology, Endothelium, Vascular pathology, Humans, Antiporters metabolism, Cyclic AMP physiology, Cyclic AMP-Dependent Protein Kinases metabolism, Endothelial Cells metabolism, Endothelial Cells pathology
- Abstract
cAMP regulates integrin-dependent adhesions of vascular endothelial cells (VECs) to extracellular matrix proteins, their vascular endothelial cadherin-dependent intercellular adhesions, and their proliferation and migration in response to growth and chemotactic factors. Previously, we reported that cAMP-elevating agents differentially inhibited migration of human VECs isolated from large vascular structures (macro-VECs, human aortic endothelial cells [HAECs]) or small vascular structures (micro-VECs, human microvascular endothelial cells [HMVECs]) and that cAMP hydrolysis by phosphodiesterase (PDE)3 and PDE4 enzymes was important in coordinating this difference. Here we report that 2 cAMP-effector enzymes, namely protein kinase (PK)A and exchange protein activated by cAMP (EPAC), each regulate extracellular matrix protein-based adhesions of both macro- and micro-VECs. Of interest and potential therapeutic importance, we report that although specific pharmacological activation of EPAC markedly stimulated adhesion of micro-VECs to extracellular matrix proteins when PKA was inhibited, this treatment only modestly promoted adhesion of macro-VECs. Consistent with an important role for cAMP PDEs in this difference, PDE3 or PDE4 inhibitors promoted EPAC-dependent adhesions in micro-VECs when PKA was inhibited but not in macro-VECs. At a molecular level, we identify multiple, nonoverlapping, PKA- or EPAC-based signaling protein complexes in both macro- and micro-VECs and demonstrate that each of these complexes contains either PDE3B or PDE4D but not both of these PDEs. Taken together, our data support the concept that adhesion of macro- and micro-VECs is differentially regulated by cAMP and that these differences are coordinated through selective actions of cAMP at multiple nonoverlapping signaling complexes that contain PKA or EPAC and distinct PDE variants.
- Published
- 2007
- Full Text
- View/download PDF
33. PI3Kgamma is required for PDE4, not PDE3, activity in subcellular microdomains containing the sarcoplasmic reticular calcium ATPase in cardiomyocytes.
- Author
-
Kerfant BG, Zhao D, Lorenzen-Schmidt I, Wilson LS, Cai S, Chen SR, Maurice DH, and Backx PH
- Subjects
- 3',5'-Cyclic-AMP Phosphodiesterases antagonists & inhibitors, Animals, Calcium metabolism, Calcium Channels, L-Type metabolism, Cell Compartmentation physiology, Class Ib Phosphatidylinositol 3-Kinase, Cyclic AMP metabolism, Cyclic Nucleotide Phosphodiesterases, Type 3, Cyclic Nucleotide Phosphodiesterases, Type 4, Enzyme Inhibitors pharmacology, Heart Diseases metabolism, Isoenzymes genetics, Isoenzymes metabolism, Mice, Mice, Mutant Strains, Myocardial Contraction physiology, Myocytes, Cardiac cytology, Phosphatidylinositol 3-Kinases genetics, Ryanodine Receptor Calcium Release Channel metabolism, 3',5'-Cyclic-AMP Phosphodiesterases metabolism, Myocytes, Cardiac enzymology, Phosphatidylinositol 3-Kinases metabolism, Sarcoplasmic Reticulum Calcium-Transporting ATPases metabolism
- Abstract
We recently showed that phosphoinositide-3-kinase-gamma-deficient (PI3Kgamma(-/-)) mice have enhanced cardiac contractility attributable to cAMP-dependent increases in sarcoplasmic reticulum (SR) Ca(2+) content and release but not L-type Ca(2+) current (I(Ca,L)), demonstrating PI3Kgamma locally regulates cAMP levels in cardiomyocytes. Because phosphodiesterases (PDEs) can contribute to cAMP compartmentation, we examined whether the PDE activity was altered by PI3Kgamma ablation. Selective inhibition of PDE3 or PDE4 in wild-type (WT) cardiomyocytes elevated Ca(2+) transients, SR Ca(2+) content, and phospholamban phosphorylation (PLN-PO(4)) by similar amounts to levels observed in untreated PI3Kgamma(-/-) myocytes. Combined PDE3 and PDE4 inhibition caused no further increases in SR function. By contrast, only PDE3 inhibition affected Ca(2+) transients, SR Ca(2+) loads, and PLN-PO(4) levels in PI3Kgamma(-/-) myocytes. On the other hand, inhibition of PDE3 or PDE4 alone did not affect I(Ca,L) in either PI3Kgamma(-/-) or WT cardiomyocytes, whereas simultaneous PDE3 and PDE4 inhibition elevated I(Ca,L) in both groups. Ryanodine receptor (RyR(2)) phosphorylation levels were not different in basal conditions between PI3Kgamma(-/-) and WT myocytes and increased in both groups with PDE inhibition. Our results establish that L-type Ca(2+) channels, RyR(2), and SR Ca(2+) pumps are regulated differently in distinct subcellular compartments by PDE3 and PDE4. In addition, the loss of PI3Kgamma selectively abolishes PDE4 activity, not PDE3, in subcellular compartments containing the SR Ca(2+)-ATPase but not RyR(2) or L-type Ca(2+) channels.
- Published
- 2007
- Full Text
- View/download PDF
34. Formation of extracellular matrix-digesting invadopodia by primary aortic smooth muscle cells.
- Author
-
Furmaniak-Kazmierczak E, Crawley SW, Carter RL, Maurice DH, and Côté GP
- Subjects
- Animals, Cell Movement, Cells, Cultured, Collagen metabolism, Microscopy, Fluorescence, Muscle, Smooth, Vascular metabolism, Phorbol 12,13-Dibutyrate pharmacology, Protein Serine-Threonine Kinases physiology, Rats, cdc42 GTP-Binding Protein physiology, p21-Activated Kinases, rac GTP-Binding Proteins physiology, src-Family Kinases physiology, Aorta cytology, Cell Surface Extensions metabolism, Extracellular Matrix metabolism, Muscle, Smooth, Vascular cytology, Myocytes, Smooth Muscle physiology
- Abstract
Invasion of the subendothelial space by vascular smooth muscle cells (VSMCs) contributes to the development and progression of diverse cardiovascular diseases. In this report we show that the expression of activated versions of Src, Cdc42 and Rac1, or a kinase-dead but open form of the p21-activated kinase (PAK1), induces primary rat aorta VSMCs to form extracellular matrix-degrading actin-rich protrusions that are morphologically similar to the invadopodia formed by highly invasive tumor cells. The matrix-degrading structures are enriched in known markers for invadopodia, including cortactin and tyrosine-phosphorylated cortactin and contain the matrix metalloproteinases MMP-9 and MT1-MMP and the urokinase plasminogen activator receptor (uPAR). In contrast to other cell types, invadopodia formation in VSMCs is only weakly supported by the phorbol ester PBDu. Invadopodia formation by Src was dependent on Cdc42, Rac, and ERK, but not on p38 MAPK. Invadopodia formation induced by kinase-dead PAK1 required Src and ERK activity and a direct interaction with the exchange factor PIX. VSMCs embedded in a three-dimensional collagen matrix formed actin- and cortactin-rich extensions that penetrated through holes in the matrix, suggesting that invadopodia-like structures are formed in a three-dimensional environment.
- Published
- 2007
- Full Text
- View/download PDF
35. cAMP-Specific phosphodiesterase-4 enzymes in the cardiovascular system: a molecular toolbox for generating compartmentalized cAMP signaling.
- Author
-
Houslay MD, Baillie GS, and Maurice DH
- Subjects
- 3',5'-Cyclic-AMP Phosphodiesterases chemistry, A Kinase Anchor Proteins, Adaptor Proteins, Signal Transducing metabolism, Animals, Arrestins metabolism, Cardiovascular System metabolism, Cyclic Nucleotide Phosphodiesterases, Type 4, Humans, Isoenzymes metabolism, Muscle, Smooth, Vascular cytology, Muscle, Smooth, Vascular enzymology, Myocytes, Cardiac enzymology, Myocytes, Smooth Muscle enzymology, Myocytes, Smooth Muscle physiology, Ryanodine Receptor Calcium Release Channel metabolism, Signal Transduction, Vasoconstriction, beta-Arrestins, 3',5'-Cyclic-AMP Phosphodiesterases metabolism, Cardiovascular System enzymology, Cyclic AMP metabolism
- Abstract
Cyclic AMP regulates a vast number of distinct events in all cells. Early studies established that its hydrolysis by cyclic nucleotide phosphodiesterases (PDEs) controlled both the magnitude and the duration of its influence. Recent evidence shows that PDEs also act as coincident detectors linking cyclic-nucleotide- and non-cyclic-nucleotide-based cellular signaling processes and are tethered with great selectively to defined intracellular structures, thereby integrating and spatially restricting their cellular effects in time and space. Although 11 distinct families of PDEs have been defined, and cells invariably express numerous individual PDE enzymes, a large measure of our increased appreciation of the roles of these enzymes in regulating cyclic nucleotide signaling has come from studies on the PDE4 family. Four PDE4 genes encode more than 20 isoforms. Alternative mRNA splicing and the use of different promoters allows cells the possibility of expressing numerous PDE4 enzymes, each with unique amino-terminal-targeting and/or regulatory sequences. Dominant negative and small interfering RNA-mediated knockdown strategies have proven that particular isoforms can uniquely control specific cellular functions. Thus the protein kinase A phosphorylation status of the beta(2) adrenoceptor and, thereby, its ability to switch its signaling to extracellular signal-regulated kinase activation, is uniquely regulated by PDE4D5 in cardiomyocytes. We describe how cardiomyocytes and vascular smooth muscle cells selectively vary both the expression and the catalytic activities of PDE4 isoforms to regulate their various functions and how altered regulation of these processes can influence the development, or resolution, of cardiovascular pathologies, such as heart failure, as well as various vasculopathies.
- Published
- 2007
- Full Text
- View/download PDF
36. Protein kinase A phosphorylation of human phosphodiesterase 3B promotes 14-3-3 protein binding and inhibits phosphatase-catalyzed inactivation.
- Author
-
Palmer D, Jimmo SL, Raymond DR, Wilson LS, Carter RL, and Maurice DH
- Subjects
- 3',5'-Cyclic-AMP Phosphodiesterases antagonists & inhibitors, 3',5'-Cyclic-AMP Phosphodiesterases physiology, Animals, Cell Line, Cyclic AMP-Dependent Protein Kinases metabolism, Cyclic Nucleotide Phosphodiesterases, Type 3, Humans, Mice, NIH 3T3 Cells, Phosphodiesterase Inhibitors metabolism, Phosphoric Monoester Hydrolases physiology, Phosphorylation, Protein Binding physiology, 14-3-3 Proteins metabolism, 3',5'-Cyclic-AMP Phosphodiesterases metabolism, Cyclic AMP-Dependent Protein Kinases physiology, Phosphoric Monoester Hydrolases antagonists & inhibitors, Phosphoric Monoester Hydrolases metabolism
- Abstract
Recent studies confirm that intracellular cAMP concentrations are nonuniform and that localized subcellular cAMP hydrolysis by cyclic nucleotide phosphodiesterases (PDEs) is important in maintaining these cAMP compartments. Human phosphodiesterase 3B (HSPDE3B), a member of the PDE3 family of PDEs, represents the dominant particulate cAMP-PDE activity in many cell types, including adipocytes and cells of hematopoietic lineage. Although several previous reports have shown that phosphorylation of HSPDE3B by either protein kinase A (PKA) or protein kinase B (PKB) activates this enzyme, the mechanisms that allow cells to distinguish these two activated forms of HSPDE3B are unknown. Here we report that PKA phosphorylates HSPDE3B at several distinct sites (Ser-73, Ser-296, and Ser-318), and we show that phosphorylation of HSPDE3B at Ser-318 activates this PDE and stimulates its interaction with 14-3-3 proteins. In contrast, although PKB-catalyzed phosphorylation of HSPDE3B activates this enzyme, it does not promote 14-3-3 protein binding. Interestingly, we report that the PKA-phosphorylated, 14-3-3 protein-bound, form of HSPDE3B is protected from phosphatase-dependent dephosphorylation and inactivation. In contrast, PKA-phosphorylated HSPDE3B that is not bound to 14-3-3 proteins is readily dephosphorylated and inactivated. Our data are presented in the context that a selective interaction between PKA-activated HSPDE3B and 14-3-3 proteins represents a mechanism by which cells can protect this enzyme from deactivation. Moreover, we propose that this mechanism may allow cells to distinguish between PKA- and PKB-activated HSPDE3B.
- Published
- 2007
- Full Text
- View/download PDF
37. Adiponectin, ghrelin, and leptin differentially influence human platelet and human vascular endothelial cell functions: implication in obesity-associated cardiovascular diseases.
- Author
-
Elbatarny HS, Netherton SJ, Ovens JD, Ferguson AV, and Maurice DH
- Subjects
- Capillary Permeability drug effects, Dose-Response Relationship, Drug, Endothelial Cells physiology, Ghrelin, Humans, Platelet Adhesiveness drug effects, Platelet Aggregation drug effects, Adiponectin pharmacology, Cardiovascular Diseases etiology, Endothelial Cells drug effects, Leptin pharmacology, Obesity complications, Peptide Hormones pharmacology, Platelet Activation drug effects
- Abstract
A very strong epidemiological link exists between obesity, the metabolic syndrome, diabetes and diabetes-associated cardiovascular pathologies. For this reason the peripheral effects of the centrally-acting satiety adipokines, adiponectin and leptin, and of non-adipose-derived hormones with similar effects, like ghrelin, have received considerable attention. In this report, we have extended our previous studies of the pro-thrombotic effects of leptin and determined the effects of adiponectin or ghrelin on human platelet activation. Thus, while leptin stimulated human platelet aggregation and adhesion, addition of adiponectin or of ghrelin did not affect either aggregation or adhesion of these cells; even at supra-physiological concentrations. In addition, we compared the impact of these three important hormones on microvascular endothelial cell permeability, an important parameter of endothelial function that when impaired contributes to several vascular pathologies. While physiologically relevant concentrations of either leptin or adiponectin increased the integrity of the diffusion barrier formed by a monolayer of human microvascular endothelial cells, only supra-physiological concentrations of ghrelin had this effect. None of these agents reduced microvascular endothelial barrier function. Taken together, our data are consistent with the ideas that leptin activates human platelets and limits transendothelial cell diffusion but that adiponectin only influences endothelial cell permeability. In contrast, ghrelin had neither of these effects. We propose that these data identify important differences in the effects of leptin, adiponectin or ghrelin on microvascular endothelial cells and platelets and may provide a basis on which to pharmacologically manipulate the selective effects of these peptides on these cell types in human cardiovascular or thrombotic diseases associated with obesity.
- Published
- 2007
- Full Text
- View/download PDF
38. Inhibition of phosphodiesterase 5 selectively reverses nitrate tolerance in the venous circulation.
- Author
-
MacPherson JD, Gillespie TD, Dunkerley HA, Maurice DH, and Bennett BM
- Subjects
- 3',5'-Cyclic-GMP Phosphodiesterases metabolism, Animals, Cyclic Nucleotide Phosphodiesterases, Type 5, Femoral Artery drug effects, Femoral Artery enzymology, Femoral Vein drug effects, Femoral Vein enzymology, Hemodynamics drug effects, Male, Quinazolines pharmacology, Rats, Rats, Sprague-Dawley, Drug Tolerance, Nitroglycerin pharmacology, Phosphodiesterase Inhibitors pharmacology, Phosphoric Diester Hydrolases metabolism, Vasodilation drug effects
- Abstract
An important component of the antianginal efficacy of glyceryl trinitrate (GTN) is attributable to its selective venodilator effect, resulting in decreased cardiac preload and myocardial oxygen demand. Tolerance to nitrates occurs during chronic exposure, and the current study assessed whether this was due to increased phosphodiesterase (PDE) activity in the venous circulation. Tolerance was induced in rats by continuous exposure to 0.4 mg/h GTN for 48 h. Tension recordings of isolated femoral artery and vein indicated that tolerance was more pronounced in femoral vein. 4-[[3,4-(Methylenedioxy)benzyl]amino]-6-chloroquinazoline (MBCQ), a selective PDE5 inhibitor, significantly decreased the EC(50) values for GTN-induced relaxation in both tolerant and nontolerant tissues, but with the greatest relative shift occurring in tolerant veins. MBCQ also increased the vasodilator potency of 1,1-diethyl-2-hydroxy-2-nitrosohydrazine (DEA/NO), a nitric oxide donor; however, cross-tolerance between DEA/NO and GTN was not observed. A significant increase in cGMP PDE activity was observed in tolerant femoral vein, whereas PDE activity was unchanged in femoral artery. Conscious rats treated with hexamethonium (30 mg/kg) to induce ganglionic blockade exhibited blunted central venous pressure (CVP) and mean arterial pressure (MAP) responses to bolus i.v. doses of GTN in GTN-tolerant animals. The cGMP PDE inhibitor zaprinast (1 mg/kg) selectively reversed the blunted CVP response to GTN in tolerant animals but had no effect on the CVP response to GTN in nontolerant animals or on the MAP response in either group. These results suggest that increased PDE5 activity in the venous circulation contributes to the altered hemodynamic response to GTN following chronic GTN exposure.
- Published
- 2006
- Full Text
- View/download PDF
39. Leptin-mediated activation of human platelets: involvement of a leptin receptor and phosphodiesterase 3A-containing cellular signaling complex.
- Author
-
Elbatarny HS and Maurice DH
- Subjects
- Blood Platelets drug effects, Cells, Cultured, Cyclic Nucleotide Phosphodiesterases, Type 3, Humans, Multienzyme Complexes metabolism, Platelet Activation drug effects, Receptors, Cell Surface administration & dosage, Receptors, Leptin, 3',5'-Cyclic-AMP Phosphodiesterases metabolism, Blood Platelets physiology, Leptin metabolism, Platelet Activation physiology, Receptors, Cell Surface metabolism, Signal Transduction physiology
- Abstract
An elevated circulating level of the adipocyte-derived satiety hormone leptin is an independent risk factor for cardiovascular disease. Because thrombus formation is a major cause of acute coronary events and leptin was shown previously to facilitate ADP-induced platelet aggregation, we chose to define the signaling events involved in leptin-mediated platelet activation. Using pharmacological, biochemical, and cell biological approaches, we show that leptin-induced platelet activation required activation of a signaling cascade that included the long form of the leptin receptor, three kinases [Janus kinase 2 (JAK2), phosphatidylinositol 3-kinase (PI3K), and protein kinase B (PKB/Akt)], the insulin receptor substrate-1 (IRS-1), and the major human platelet cAMP phosphodiesterase phosphodiesterase 3A (PDE3A). Moreover, we identify a role for an intraplatelet LEPR/JAK2/IRS-1/PI3K/PKB/PDE3A molecular complex that allows for the selective leptin-mediated activation of platelets. Our data demonstrate that leptin promotes platelet activation, provides a mechanistic basis for the prothrombotic effect of this hormone, and identifies a potentially novel therapeutic avenue to limit obesity-associated cardiovascular disease.
- Published
- 2005
- Full Text
- View/download PDF
40. Vascular smooth muscle cell phenotype-dependent phosphodiesterase 4D short form expression: role of differential histone acetylation on cAMP-regulated function.
- Author
-
Tilley DG and Maurice DH
- Subjects
- Acetylation, Animals, Base Sequence, Cyclic Nucleotide Phosphodiesterases, Type 4, DNA Primers, Introns, Male, Mice, Muscle, Smooth, Vascular cytology, NIH 3T3 Cells, Phenotype, Polymerase Chain Reaction, Rats, Rats, Wistar, 3',5'-Cyclic-AMP Phosphodiesterases metabolism, Cyclic AMP physiology, Histones metabolism, Muscle, Smooth, Vascular enzymology
- Abstract
Sustained activation of adenylyl cyclase in vascular smooth muscle cells (VSMCs) results in the activation of a series of complex regulatory systems designed to desensitize these cells to further cAMP-mediated events. Although an increase in phosphodiesterase (PDE) 4-mediated hydrolysis of cAMP forms an integral part of this desensitization program in both "contractile/quiescent" and "synthetic/activated" VSMCs, distinct PDE4D gene variants coordinate these events in these phenotypically distinct cells. Using a combination of pharmacological, biochemical, and molecular biological approaches, and both in vivo and in vitro systems, we have identified the molecular basis underlying this VSMC phenotype-selective expression of PDE4D in response to cAMP-elevating agents in these cells. Thus, whereas the protein kinase A/cAMP response element-binding protein/cAMP response element signaling cascade regulates PDE4D expression in each VSMC phenotype, elevated levels of histone acetylation of the intronic promoter regulating PDE4D1 and PDE4D2 expression allows selective cAMP-mediated induction of expression of these PDE4D variants in synthetic/activated VSMCs. In contrast, the newly described EPAC1/Rap1A cAMP-dependent signaling cascade plays no role in regulating PDE4D expression in either VSMC phenotype. Our data are presented in the context of PDE4-mediated desensitization to cAMP-elevating agents in VSMCs and with the recognition that cAMP-elevating agents are being considered as adjunctive pharmacotherapy in percutaneous coronary interventions, including stenting.
- Published
- 2005
- Full Text
- View/download PDF
41. Cyclic nucleotide phosphodiesterase-mediated integration of cGMP and cAMP signaling in cells of the cardiovascular system.
- Author
-
Maurice DH
- Subjects
- 3',5'-Cyclic-AMP Phosphodiesterases metabolism, 3',5'-Cyclic-GMP Phosphodiesterases metabolism, Animals, Cardiovascular System cytology, Cardiovascular System enzymology, Cyclic Nucleotide Phosphodiesterases, Type 2, Cyclic Nucleotide Phosphodiesterases, Type 3, Cyclic Nucleotide Phosphodiesterases, Type 5, Humans, Signal Transduction, Cardiovascular System metabolism, Cyclic AMP physiology, Cyclic GMP physiology, Phosphoric Diester Hydrolases metabolism
- Abstract
Numerous pharmacological and physiological agents acting via either cAMP- or cGMP-mediated impact the activities of cells of the cardiovascular system. While most define cAMP and cGMP signaling systems as separate and independent, recent advances in our understanding of cyclic nucleotide signaling, and more specifically, of the roles which cyclic nucleotide phosphodiesterases (PDEs) play in these events, have altered this view. In this short chapter, I will review the data identifying expression of several PDEs in cells of the cardiovascular system. In addition, I will review the data that identify PDEs as enzymes capable of allowing integration between cAMP and cGMP signaling in cells, and propose that cAMP and cGMP signaling systems can represent parallel and interdependent signaling systems. Moreover, I will propose that cGMP-mediated effects on the activities of variants of the Phosphodiesterase 2 (PDE2), PDE3 and PDE5 families may act to coordinate linkage between cAMP and cGMP signaling in these cells.
- Published
- 2005
- Full Text
- View/download PDF
42. Mechanism of tissue-selective drug action in the cardiovascular system.
- Author
-
Barrett TD, Triggle DJ, Walker MJ, and Maurice DH
- Subjects
- 3',5'-Cyclic-GMP Phosphodiesterases antagonists & inhibitors, Binding Sites, Calcium Channels chemistry, Chemistry, Pharmaceutical methods, Cyclic Nucleotide Phosphodiesterases, Type 5, Electrophysiology, Erectile Dysfunction drug therapy, Humans, Hypertension drug therapy, Ischemia drug therapy, Male, Models, Anatomic, Models, Biological, Myocardium pathology, Nifedipine pharmacology, Phosphodiesterase Inhibitors pharmacology, Verapamil pharmacology, Cardiovascular System drug effects
- Abstract
Analysis of the human genome project tells us that there may be as few as 3000 genes that are likely to be good drug targets. Although the number of targets is still very large, these data have been interpreted by some to mean that the pharmaceutical industry may someday run out of novel drug targets. Despite the doom and gloom of such analysis, there is considerable reason for optimism. Drugs may exhibit selectivity of action beyond that predicted by target expression alone. Drugs that act at a single molecular target may have very different pharmacology and, as a result, different therapeutic uses. Three well-characterized model systems are highlighted to illustrate this point. The first model system is exemplified by nifedipine and verapamil, both of which act on L-type calcium channels. Both drugs are used to treat hypertension, but only verapamil can be used to produce atrioventricular block in patients with atrial fibrillation. The second model system describes the therapeutic exploitation of unusual conditions that occur in the ischemic myocardium to produce drugs that are more effective for suppressing ischemia-induced arrhythmias. The third model system discusses the mechanisms through which phosphodiesterase-5 (PDE5) inhibitors act selectively to facilitate penile erection while having little effect in the non-penile vasculature that also expresses PDE5.
- Published
- 2005
- Full Text
- View/download PDF
43. Vascular endothelial cell cyclic nucleotide phosphodiesterases and regulated cell migration: implications in angiogenesis.
- Author
-
Netherton SJ and Maurice DH
- Subjects
- 1-Methyl-3-isobutylxanthine pharmacology, 3',5'-Cyclic-AMP Phosphodiesterases genetics, 3',5'-Cyclic-AMP Phosphodiesterases metabolism, 3',5'-Cyclic-GMP Phosphodiesterases genetics, 3',5'-Cyclic-GMP Phosphodiesterases metabolism, Aorta, Cyclic AMP metabolism, Cyclic Nucleotide Phosphodiesterases, Type 2, Cyclic Nucleotide Phosphodiesterases, Type 3, Cyclic Nucleotide Phosphodiesterases, Type 4, Cyclic Nucleotide Phosphodiesterases, Type 5, Endothelium, Vascular enzymology, Humans, Kinetics, Microcirculation physiology, Phosphoric Diester Hydrolases genetics, Phosphoric Diester Hydrolases metabolism, Quinolones pharmacology, RNA, Messenger genetics, Reverse Transcriptase Polymerase Chain Reaction, Umbilical Veins, 2',3'-Cyclic-Nucleotide Phosphodiesterases genetics, 2',3'-Cyclic-Nucleotide Phosphodiesterases metabolism, Cell Movement physiology, Cyclic AMP physiology, Endothelium, Vascular physiology, Neovascularization, Physiologic physiology
- Abstract
Angiogenesis is necessary during embryonic development and wound healing but can be detrimental in pathologies, including cancer. Because initiation of angiogenesis involves migration and proliferation of vascular endothelial cells (VECs) and cAMP-elevating agents inhibit these events, such agents may represent a novel therapeutic avenue to controlling angiogenesis. Intracellular cAMP levels are regulated by their synthesis by adenylyl cyclases and hydrolysis by cyclic nucleotide phosphodiesterases (PDEs). In this report, we show that human VECs express variants of PDE2, PDE3, PDE4, and PDE5 families and demonstrate that the levels of these enzymes differ in VECs derived from aorta, umbilical vein, and microvascular structures. Selective inhibition of PDE2 did not increase cAMP in any VECs, whether in the absence or presence of forskolin, but it did inhibit migration of all VECs studied. Inhibition of PDE4 activity decreased migration, and in conjunction with forskolin, increased cAMP in all VECs studied. PDE3 inhibition potentiated forskolin-induced increases in cAMP and inhibited migration in VECs derived from aorta and umbilical vein but not in microvascular VECs. In experiments with combinations of PDE2, PDE3, and PDE4 inhibitors, a complex interaction between the abilities of these agents to limit human VEC migration was observed. Overall, our data are consistent with the hypothesis that PDE subtype inhibition allows different effects in distinct VEC populations and indicate that these agents may represent novel therapeutic agents to limit angiogenesis in complex human diseases.
- Published
- 2005
- Full Text
- View/download PDF
44. Cardiovascular implications in the use of PDE5 inhibitor therapy.
- Author
-
Maurice DH
- Subjects
- Animals, Cardiovascular System cytology, Cardiovascular System enzymology, Cyclic AMP metabolism, Cyclic GMP metabolism, Cyclic Nucleotide Phosphodiesterases, Type 5, Endothelium, Vascular enzymology, Guanylate Cyclase metabolism, Humans, Male, Muscle, Smooth cytology, Muscle, Smooth drug effects, Phosphodiesterase Inhibitors therapeutic use, Phosphoric Diester Hydrolases metabolism, 3',5'-Cyclic-GMP Phosphodiesterases antagonists & inhibitors, Cardiovascular System drug effects, Phosphodiesterase Inhibitors adverse effects
- Abstract
Cardiovascular smooth muscle cells (SMCs) exist as resting or activated cells. Resting SMCs produce contractile proteins and are nearly transcriptionally inactive; activated SMCs are transcriptionally active and are involved in pathological processes such as atherosclerosis. Soluble guanylate cyclase, protein kinase G, and protein kinase A are present in SMCs, but their levels can be decreased in activated cells. Phosphodiesterase 3 (PDE3) activity is abundant in cardiovascular tissues; both PDE3A and PDE3B are involved in cyclic adenosine monophosphate (cAMP) hydrolysis in these tissues. Cyclic-AMP-hydrolyzing PDE activities are altered during the phenotypic transition of SMCs from the resting to the activated phenotype. Similar changes have been observed in cyclic guanosine monophosphate cGMP-hydrolyzing PDEs, although the impact of these alterations on PDE5 inhibitor-mediated effects requires further study. This report presents the changes in PDE expression that accompany phenotypic modulation of SMCs and discusses the potential impact of these events on PDE5-mediated cell functions.
- Published
- 2004
- Full Text
- View/download PDF
45. Cleavage of the matricellular protein SPARC by matrix metalloproteinase 3 produces polypeptides that influence angiogenesis.
- Author
-
Sage EH, Reed M, Funk SE, Truong T, Steadele M, Puolakkainen P, Maurice DH, and Bassuk JA
- Subjects
- Amino Acid Sequence, Animals, Cattle, Cell Division, Copper metabolism, Dose-Response Relationship, Drug, Endothelium, Vascular cytology, Humans, Molecular Sequence Data, Osteonectin chemistry, Recombinant Proteins metabolism, Matrix Metalloproteinase 3 physiology, Neovascularization, Physiologic drug effects, Osteonectin metabolism, Peptides pharmacology
- Abstract
SPARC, a matricellular protein that affects cellular adhesion and proliferation, is produced in remodeling tissue and in pathologies involving fibrosis and angiogenesis. In this study we have asked whether peptides generated from cleavage of SPARC in the extracellular milieu can regulate angiogenesis. Matrix metalloproteinase (MMP)-3, but not MMP-1 or 9, showed significant activity toward SPARC. Limited digestion of recombinant human (rhu)SPARC with purified catalytic domain of rhuMMP-3 produced three major fragments, which were sequenced after purification by HPLC. Three synthetic peptides (Z-1, Z-2, and Z-3) representing motifs from each fragment were tested in distinct assays of angiogenesis. Peptide Z-1 (3.9 kDa, containing a Cu2+-binding sequence KHGK) exhibited a biphasic effect on [3H]thymidine incorporation by cultured endothelial cells and stimulated vascular growth in the chick chorioallantoic membrane (CAM). In contrast, peptides Z-2 (6.1 kDa, containing Ca2+-binding EF hand-1) and Z-3 (2.2 kDa, containing neither Cu2+-binding motifs nor EF hands), inhibited cell proliferation in a concentration-dependent manner and exhibited no effects on vessel growth in the CAM. Reciprocal results were obtained in a migration assay in native collagen gels: peptide Z-1 was ineffective over a range of concentrations, whereas Z-2 or Z-3 stimulated cell migration. Therefore, proteolysis of SPARC by MMP-3 produced peptides that regulate endothelial cell proliferation and/or migration in vitro in a mutually exclusive manner. One of these peptides containing KHGK also demonstrated a concentration-dependent effect on angiogenesis.
- Published
- 2003
- Full Text
- View/download PDF
46. Cyclic nucleotide phosphodiesterase activity, expression, and targeting in cells of the cardiovascular system.
- Author
-
Maurice DH, Palmer D, Tilley DG, Dunkerley HA, Netherton SJ, Raymond DR, Elbatarny HS, and Jimmo SL
- Subjects
- 2',3'-Cyclic-Nucleotide Phosphodiesterases antagonists & inhibitors, Animals, Cardiovascular System drug effects, Enzyme Activation drug effects, Enzyme Activation physiology, Gene Expression Regulation, Enzymologic drug effects, Humans, 2',3'-Cyclic-Nucleotide Phosphodiesterases biosynthesis, Cardiovascular System enzymology, Drug Delivery Systems methods, Gene Expression Regulation, Enzymologic physiology
- Abstract
Cyclic AMP (cAMP) and cGMP regulate a myriad of cellular functions, such as metabolism, contractility, motility, and transcription in virtually all cell types, including those of the cardiovascular system. Considerable effort over the last 20 years has allowed identification of the cellular components involved in the synthesis of cyclic nucleotides, as well as effectors of cyclic nucleotide-mediated signaling. More recently, a central role for cyclic nucleotide phosphodiesterase (PDE) has also been elaborated in many cell types, including those involved in regulating the activities of the cardiovascular system. In this review, we introduce the PDE families whose members are expressed in cells of the cardiovascular system including cardiomyocytes, vascular smooth muscle cells, and vascular endothelial cells. Because cell behavior is a dynamic process influenced by numerous factors, we will attempt to emphasize how changes in the activity, expression, and targeting of PDE influence cyclic nucleotide-mediated regulation of the behavior of these cells.
- Published
- 2003
- Full Text
- View/download PDF
47. Does sildenafil indirectly inhibit phosphodiesterase 3 in vascular smooth muscle?
- Author
-
Maurice DH
- Subjects
- Cyclic AMP metabolism, Cyclic GMP metabolism, Cyclic Nucleotide Phosphodiesterases, Type 3, Forearm blood supply, Humans, Muscle, Smooth, Vascular drug effects, Purines, Regional Blood Flow, Sildenafil Citrate, Sulfones, Vasodilation drug effects, 3',5'-Cyclic-AMP Phosphodiesterases antagonists & inhibitors, Muscle, Smooth, Vascular enzymology, Phosphodiesterase Inhibitors pharmacology, Piperazines pharmacology, Vasodilator Agents pharmacology
- Published
- 2003
- Full Text
- View/download PDF
48. Dynamic regulation of cAMP signaling by cGMP in the cardiovascular system: roles of phosphodiesterase 2 and phosphodiesterase 3 enzymes.
- Author
-
Maurice DH
- Subjects
- Blood Platelets physiology, Cardiovascular System cytology, Cyclic Nucleotide Phosphodiesterases, Type 3, Humans, Muscle Cells physiology, Muscle, Smooth, Vascular cytology, Muscle, Smooth, Vascular physiology, 3',5'-Cyclic-AMP Phosphodiesterases physiology, Cardiovascular Physiological Phenomena, Cyclic AMP physiology, Cyclic GMP physiology, Exonucleases physiology, Signal Transduction physiology
- Published
- 2003
49. Vascular smooth muscle cell phosphodiesterase (PDE) 3 and PDE4 activities and levels are regulated by cyclic AMP in vivo.
- Author
-
Tilley DG and Maurice DH
- Subjects
- Animals, Aorta cytology, Cyclic Nucleotide Phosphodiesterases, Type 3, Cyclic Nucleotide Phosphodiesterases, Type 4, Femoral Artery drug effects, Femoral Artery enzymology, Male, Muscle, Smooth, Vascular enzymology, Rats, Rats, Wistar, 3',5'-Cyclic-AMP Phosphodiesterases metabolism, Bucladesine pharmacology, Cyclic AMP metabolism, Muscle, Smooth, Vascular drug effects
- Abstract
Prolonged incubation of several cell types, including cultured vascular smooth muscle cells (VSMC), with cyclic AMP-elevating agents increases cAMP phosphodiesterase (PDE) activity and levels. In this work, we describe for the first time an increase in arterial VSMC cAMP PDE activity and levels caused by cAMP-elevating agents when these agents are administered to rats in vivo. Injections of rats with dibutyryl cAMP (dbcAMP) or forskolin increased both PDE3 and PDE4 activities in aortic and femoral artery VSMC. Consistent with the idea that cAMP-elevating agents increased PDE3 and PDE4 activities by acting directly on VSMC, local delivery of dbcAMP or forskolin to femoral arteries using a pluronic gel-based approach increased femoral artery VSMC PDE3 and PDE4 activities to levels similar to those observed after injection of these agents. Consistent with a role for de novo mRNA and protein synthesis in the cAMP-elevating agent induced increase in PDE3 and PDE4, 1) systemic administration of forskolin increased PDE3A, PDE3B, and PDE4D mRNA levels in aortic VSMC and femoral artery VSMC, 2) local delivery of dbcAMP increased PDE3A, PDE3B, and PDE4D3 protein levels in femoral artery VSMC, and 3) local administration of either actinomycin D or cycloheximide attenuated the effect of dbcAMP. In addition, our results indicate that the PDE3 and PDE4 variants increased by cAMP-elevating agents in arterial VSMC in situ were distinct from those elevated by these agents in cultured arterial VSMC. Consistent with the effect of increased VSMC cAMP PDE on blood vessel function, inhibition of PDE3 and PDE4 activities potentiated the relaxant effect of forskolin in dbcAMP-treated femoral artery rings to a greater extent than in untreated control blood vessels. We propose that our findings are consistent with the concept that cAMP regulates VSMC cAMP PDE activity and levels in vivo and that VSMC phenotype influences the choice of cAMP PDE variant that is elevated. Our findings are discussed in the context that agents aimed at specific PDE3 or PDE4 variants could perhaps allow greater control of cAMP-mediated regulation of VSMC behaviors that are phenotype-dependent.
- Published
- 2002
- Full Text
- View/download PDF
50. Reduced phosphodiesterase 3 activity and phosphodiesterase 3A level in synthetic vascular smooth muscle cells: implications for use of phosphodiesterase 3 inhibitors in cardiovascular tissues.
- Author
-
Dunkerley HA, Tilley DG, Palmer D, Liu H, Jimmo SL, and Maurice DH
- Subjects
- 3',5'-Cyclic-AMP Phosphodiesterases genetics, Animals, Aorta cytology, Aorta drug effects, Cardiovascular System metabolism, Cells, Cultured, Cyclic AMP metabolism, Cyclic GMP metabolism, Cyclic Nucleotide Phosphodiesterases, Type 3, Drug Delivery Systems, Gene Expression, Male, Muscle, Smooth, Vascular cytology, Muscle, Smooth, Vascular drug effects, RNA, Messenger drug effects, RNA, Messenger metabolism, Rats, Rats, Wistar, Vasoconstriction, 3',5'-Cyclic-AMP Phosphodiesterases metabolism, Cardiovascular System drug effects, Muscle, Smooth, Vascular enzymology, Phosphodiesterase Inhibitors pharmacology
- Abstract
Vascular smooth muscle cells (VSMC) in situ function to control contraction and are said to express a contractile phenotype. However, during development or in response to vascular damage, VSMC proliferate and express a more synthetic phenotype. A survey of literature values for contractile and synthetic VSMC phosphodiesterase (PDE) 3 and PDE4 activities identified a marked difference in the PDE3 and PDE4 activities of these cells. In this study, a comparison of PDE3 and PDE4 activities in contractile and synthetic VSMC demonstrates that a reduced PDE3/PDE4 activity ratio in synthetic VSMC correlates with a reduced PDE3 activity and is associated with marked reductions in PDE3A mRNA and protein levels. Because we show that similar reductions in PDE3 activity and PDE3A levels occur upon culture of human aortic VSMC and that this phenomenon associates with the phenotypic switch that occurs to VSMC in response to vascular damage, our findings are presented in the context that PDE3 inhibition might be expected to selectively alter functions of contractile VSMC.
- Published
- 2002
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.