Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
J Neuroinflammation ; 14(1): 114, 2017 06 05.
Article in English | MEDLINE | ID: mdl-28583195

ABSTRACT

BACKGROUND: Cerebral microbleeds (CMB) are MRI-demonstrable cerebral microhemorrhages (CMH) which commonly coexist with ischemic stroke. This creates a challenging therapeutic milieu, and a strategy that simultaneously protects the vessel wall and provides anti-thrombotic activity is an attractive potential approach. Phosphodiesterase 3A (PDE3A) inhibition is known to provide cerebral vessel wall protection combined with anti-thrombotic effects. As an initial step in the development of a therapy that simultaneously treats CMB and ischemic stroke, we hypothesized that inhibition of the PDE3A pathway is protective against CMH development. METHODS: The effect of PDE3A pathway inhibition was studied in the inflammation-induced and cerebral amyloid angiopathy (CAA)-associated mouse models of CMH. The PDE3A pathway was modulated using two approaches: genetic deletion of PDE3A and pharmacological inhibition of PDE3A by cilostazol. The effects of PDE3A pathway modulation on H&E- and Prussian blue (PB)-positive CMH development, BBB function (IgG, claudin-5, and fibrinogen), and neuroinflammation (ICAM-1, Iba-1, and GFAP) were investigated. RESULTS: Robust development of CMH in the inflammation-induced and CAA-associated spontaneous mouse models was observed. Inflammation-induced CMH were associated with markers of BBB dysfunction and inflammation, and CAA-associated spontaneous CMH were associated primarily with markers of neuroinflammation. Genetic deletion of the PDE3A gene did not alter BBB function, microglial activation, or CMH development, but significantly reduced endothelial and astrocyte activation in the inflammation-induced CMH mouse model. In the CAA-associated CMH mouse model, PDE3A modulation via pharmacological inhibition by cilostazol did not alter BBB function, neuroinflammation, or CMH development. CONCLUSIONS: Modulation of the PDE3A pathway, either by genetic deletion or pharmacological inhibition, does not alter CMH development in an inflammation-induced or in a CAA-associated mouse model of CMH. The role of microglial activation and BBB injury in CMH development warrants further investigation.


Subject(s)
Cerebral Hemorrhage/drug therapy , Cerebral Hemorrhage/genetics , Cyclic Nucleotide Phosphodiesterases, Type 3/deficiency , Cyclic Nucleotide Phosphodiesterases, Type 3/genetics , Microvessels/drug effects , Phosphodiesterase 3 Inhibitors/therapeutic use , Animals , Cerebral Hemorrhage/enzymology , Cilostazol , Gene Deletion , Mice , Mice, Knockout , Mice, Transgenic , Microvessels/enzymology , Microvessels/pathology , Phosphodiesterase 3 Inhibitors/pharmacology , Tetrazoles , Treatment Outcome
2.
Sci Rep ; 7: 40445, 2017 01 13.
Article in English | MEDLINE | ID: mdl-28084425

ABSTRACT

Understanding mechanisms by which a population of beige adipocytes is increased in white adipose tissue (WAT) reflects a potential strategy in the fight against obesity and diabetes. Cyclic adenosine monophosphate (cAMP) is very important in the development of the beige phenotype and activation of its thermogenic program. To study effects of cyclic nucleotides on energy homeostatic mechanisms, mice were generated by targeted inactivation of cyclic nucleotide phosphodiesterase 3b (Pde3b) gene, which encodes PDE3B, an enzyme that catalyzes hydrolysis of cAMP and cGMP and is highly expressed in tissues that regulate energy homeostasis, including adipose tissue, liver, and pancreas. In epididymal white adipose tissue (eWAT) of PDE3B KO mice on a SvJ129 background, cAMP/protein kinase A (PKA) and AMP-activated protein kinase (AMPK) signaling pathways are activated, resulting in "browning" phenotype, with a smaller increases in body weight under high-fat diet, smaller fat deposits, increased ß-oxidation of fatty acids (FAO) and oxygen consumption. Results reported here suggest that PDE3B and/or its downstream signaling partners might be important regulators of energy metabolism in adipose tissue, and potential therapeutic targets for treating obesity, diabetes and their associated metabolic disorders.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Adipose Tissue, Beige/metabolism , Adipose Tissue, White/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 3/deficiency , Signal Transduction , 3T3-L1 Cells , Adipocytes/metabolism , Animals , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism , Energy Metabolism , Enzyme Activation , Epididymis/metabolism , Female , Gene Knockdown Techniques , Male , Mice , Mice, Knockout , Mitochondria/metabolism , Obesity/metabolism , Obesity/prevention & control , Organelle Biogenesis , Phenotype , Thermogenesis , Weight Gain
3.
Sci Rep ; 6: 28056, 2016 06 20.
Article in English | MEDLINE | ID: mdl-27321128

ABSTRACT

Activation of inflammation in white adipose tissue (WAT), includes infiltration/expansion of WAT macrophages, contributes pathogenesis of obesity, insulin resistance, and metabolic syndrome. The inflammasome comprises an intracellular sensor (NLR), caspase-1 and the adaptor ASC. Inflammasome activation leads to maturation of caspase-1 and processing of IL1ß, contributing to many metabolic disorders and directing adipocytes to a more insulin-resistant phenotype. Ablation of PDE3B in WAT prevents inflammasome activation by reducing expression of NLRP3, caspase-1, ASC, AIM2, TNFα, IL1ß and proinflammatory genes. Following IP injection of lipopolysaccharide (LPS), serum levels of IL1ß and TNFα were reduced in PDE3B(-/-)mice compared to WT. Activation of signaling cascades, which mediate inflammasome responses, were modulated in PDE3B(-/-)mice WAT, including smad, NFAT, NFkB, and MAP kinases. Moreover, expression of chemokine CCL2, MCP-1 and its receptor CCR2, which play an important role in macrophage chemotaxis, were reduced in WAT of PDE3B(-/-)mice. In addition, atherosclerotic plaque formation was significantly reduced in the aorta of apoE(-/-)/PDE3B(-/-)and LDL-R(-/-)/PDE3B(-/-)mice compared to apoE(-/-)and LDL-R(-/-)mice, respectively. Obesity-induced changes in serum-cholesterol were blocked in PDE3B(-/-)mice. Collectively, these data establish a role for PDE3B in modulating inflammatory response, which may contribute to a reduced inflammatory state in adipose tissue.


Subject(s)
Adipose Tissue, White/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , Aorta/metabolism , Aorta/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Caspase 1/genetics , Caspase 1/metabolism , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 3/deficiency , Cyclic Nucleotide Phosphodiesterases, Type 3/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Down-Regulation/drug effects , Insulin/metabolism , Interleukin-1beta/blood , Interleukin-1beta/metabolism , Macrophages/cytology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Receptors, CCR2/genetics , Receptors, CCR2/metabolism , Receptors, LDL/deficiency , Receptors, LDL/genetics , Signal Transduction , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/metabolism
4.
Proc Natl Acad Sci U S A ; 112(17): E2253-62, 2015 Apr 28.
Article in English | MEDLINE | ID: mdl-25877153

ABSTRACT

Although inhibition of cyclic nucleotide phosphodiesterase type 3 (PDE3) has been reported to protect rodent heart against ischemia/reperfusion (I/R) injury, neither the specific PDE3 isoform involved nor the underlying mechanisms have been identified. Targeted disruption of PDE3 subfamily B (PDE3B), but not of PDE3 subfamily A (PDE3A), protected mouse heart from I/R injury in vivo and in vitro, with reduced infarct size and improved cardiac function. The cardioprotective effect in PDE3B(-/-) heart was reversed by blocking cAMP-dependent PKA and by paxilline, an inhibitor of mitochondrial calcium-activated K channels, the opening of which is potentiated by cAMP/PKA signaling. Compared with WT mitochondria, PDE3B(-/-) mitochondria were enriched in antiapoptotic Bcl-2, produced less reactive oxygen species, and more frequently contacted transverse tubules where PDE3B was localized with caveolin-3. Moreover, a PDE3B(-/-) mitochondrial fraction containing connexin-43 and caveolin-3 was more resistant to Ca(2+)-induced opening of the mitochondrial permeability transition pore. Proteomics analyses indicated that PDE3B(-/-) heart mitochondria fractions were enriched in buoyant ischemia-induced caveolin-3-enriched fractions (ICEFs) containing cardioprotective proteins. Accumulation of proteins into ICEFs was PKA dependent and was achieved by ischemic preconditioning or treatment of WT heart with the PDE3 inhibitor cilostamide. Taken together, these findings indicate that PDE3B deletion confers cardioprotective effects because of cAMP/PKA-induced preconditioning, which is associated with the accumulation of proteins with cardioprotective function in ICEFs. To our knowledge, our study is the first to define a role for PDE3B in cardioprotection against I/R injury and suggests PDE3B as a target for cardiovascular therapies.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 3/deficiency , Myocardial Reperfusion Injury , Myocardium/enzymology , Animals , Caveolin 3/genetics , Caveolin 3/metabolism , Connexin 43/genetics , Connexin 43/metabolism , Cyclic AMP/genetics , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism , Mice , Mice, Knockout , Mitochondria, Heart/genetics , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Membrane Transport Proteins/pharmacology , Mitochondrial Permeability Transition Pore , Myocardial Infarction/enzymology , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/enzymology , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/prevention & control , Myocardium/pathology , Phosphodiesterase Inhibitors/pharmacology , Quinolones/pharmacology
5.
J Biol Chem ; 286(29): 26238-49, 2011 Jul 22.
Article in English | MEDLINE | ID: mdl-21632535

ABSTRACT

Cyclic nucleotide phosphodiesterase 3 (PDE3) is an important regulator of cyclic adenosine monophosphate (cAMP) signaling within the cardiovascular system. In this study, we examined the role of PDE3A and PDE3B isoforms in regulation of growth of cultured vascular smooth muscle cells (VSMCs) and the mechanisms by which they may affect signaling pathways that mediate mitogen-induced VSMC proliferation. Serum- and PDGF-induced DNA synthesis in VSMCs grown from aortas of PDE3A-deficient (3A-KO) mice was markedly less than that in VSMCs from PDE3A wild type (3A-WT) and PDE3B-deficient (3B-KO) mice. The reduced growth response was accompanied by significantly less phosphorylation of extracellular signal-regulated kinase (ERK) in 3A-KO VSMCs, most likely due to a combination of greater site-specific inhibitory phosphorylation of Raf-1(Ser-²59) by protein kinase A (PKA) and enhanced dephosphorylation of ERKs due to elevated mitogen-activated protein kinase phosphatase 1 (MKP-1). Furthermore, 3A-KO VSMCs, compared with 3A-WT, exhibited higher basal PKA activity and cAMP response element-binding protein (CREB) phosphorylation, higher levels of p53 and p53 phosphorylation, and elevated p21 protein together with lower levels of Cyclin-D1 and retinoblastoma (Rb) protein and Rb phosphorylation. Adenoviral overexpression of inactive CREB partially restored growth effects of serum in 3A-KO VSMCs. In contrast, exposure of 3A-WT VSMCs to VP16 CREB (active CREB) was associated with inhibition of serum-induced DNA synthesis similar to that in untreated 3A-KO VSMCs. Transfection of 3A-KO VSMCs with p53 siRNA reduced p21 and MKP-1 levels and completely restored growth without affecting amounts of Cyclin-D1 and Rb phosphorylation. We conclude that PDE3A regulates VSMC growth via two complementary pathways, i.e. PKA-catalyzed inhibitory phosphorylation of Raf-1 with resulting inhibition of MAPK signaling and PKA/CREB-mediated induction of p21, leading to G0/G1 cell cycle arrest, as well as by increased accumulation of p53, which induces MKP-1, p21, and WIP1, leading to inhibition of G1 to S cell cycle progression.


Subject(s)
Cell Cycle Proteins/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 3/deficiency , Cyclic Nucleotide Phosphodiesterases, Type 3/genetics , Gene Deletion , MAP Kinase Signaling System/genetics , Mitogen-Activated Protein Kinases/metabolism , Muscle, Smooth, Vascular/cytology , Animals , Biocatalysis/drug effects , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Cycle Proteins/genetics , Cell Proliferation/drug effects , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , DNA/biosynthesis , Dual Specificity Phosphatase 1/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/genetics , Gene Knockout Techniques , MAP Kinase Signaling System/drug effects , Male , Mice , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Phosphorylation/genetics , Platelet-Derived Growth Factor/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-raf/metabolism , RNA, Small Interfering/genetics
6.
J Biol Chem ; 283(33): 22430-42, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18508768

ABSTRACT

One of the defining properties of beta2-adrenergic receptor (beta(2)AR) signaling is the transient and rapidly reversed accumulation of cAMP. Here we have investigated the contribution of different PDE4 proteins to the generation of this transient response. To this aim, mouse embryonic fibroblasts deficient in PDE4A, PDE4B, or PDE4D were generated, and the regulation of PDE activity, the accumulation of cAMP, and CREB phosphorylation in response to isoproterenol were monitored. Ablation of PDE4D, but not PDE4A or PDE4B, had a major effect on the beta-agonist-induced PDE activation, with only a minimal increase in PDE activity being retained in PDE4D knock-out (KO) cells. Accumulation of cAMP was markedly enhanced, and the kinetics of cAMP accumulation were altered in their properties in PDE4DKO but not PDE4BKO cells. Modest effects were observed in PDE4AKO mouse embryonic fibroblasts. The return to basal levels of both cAMP accumulation and CREB phosphorylation was greatly delayed in the PDE4DKO cells, suggesting that PDE4D is critical for dissipation of the beta2AR stimulus. This effect of PDE4D ablation was in large part due to inactivation of a negative feedback mechanism consisting of the PKA-mediated activation of PDE4D in response to elevated cAMP levels, as indicated by experiments using the cAMP-dependent protein kinase inhibitors H89 and PKI. Finally, PDE4D ablation affected the kinetics of beta2AR desensitization as well as the interaction of the receptor with Galphai. These findings demonstrate that PDE4D plays a major role in shaping the beta2AR signal.


Subject(s)
Cyclic AMP/physiology , Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Embryo, Mammalian/physiology , Animals , Cell Division/drug effects , Cell Line , Cyclic Nucleotide Phosphodiesterases, Type 3/deficiency , Cyclic Nucleotide Phosphodiesterases, Type 3/genetics , Cyclic Nucleotide Phosphodiesterases, Type 4/deficiency , Female , Isoproterenol/pharmacology , Mice , Pregnancy , Rolipram/pharmacology , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...