Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Cells ; 12(23)2023 12 03.
Article in English | MEDLINE | ID: mdl-38067187

ABSTRACT

In mouse cardiomyocytes, the expression of two subfamilies of the calcium/calmodulin-regulated cyclic nucleotide phosphodiesterase 1 (PDE1)-PDE1A and PDE1C-has been reported. PDE1C was found to be the major subfamily in the human heart. It is a dual substrate PDE and can hydrolyze both 3',5'-cyclic adenosine monophosphate (cAMP) and 3',5'-cyclic guanosine monophosphate (cGMP). Previously, it has been reported that the PDE1 inhibitor ITI-214 shows positive inotropic effects in heart failure patients which were largely attributed to the cAMP-dependent protein kinase (PKA) signaling. However, the role of PDE1 in the regulation of cardiac cGMP has not been directly addressed. Here, we studied the effect of PDE1 inhibition on cGMP levels in adult mouse ventricular cardiomyocytes using a highly sensitive fluorescent biosensor based on Förster resonance energy transfer (FRET). Live-cell imaging in paced and resting cardiomyocytes showed an increase in cGMP after PDE1 inhibition with ITI-214. Furthermore, PDE1 inhibition and PDE1A knockdown amplified the cGMP-FRET responses to the nitric oxide (NO)-donor sodium nitroprusside (SNP) but not to the C-type natriuretic peptide (CNP), indicating a specific role of PDE1 in the regulation of the NO-sensitive guanylyl cyclase (NO-GC)-regulated cGMP microdomain. ITI-214, in combination with CNP or SNP, showed a positive lusitropic effect, improving the relaxation of isolated myocytes. Immunoblot analysis revealed increased phospholamban (PLN) phosphorylation at Ser-16 in cells treated with a combination of SNP and PDE1 inhibitor but not with SNP alone. Our findings reveal a previously unreported role of PDE1 in the regulation of the NO-GC/cGMP microdomain and mouse ventricular myocyte contractility. Since PDE1 serves as a cGMP degrading PDE in cardiomyocytes and has the highest hydrolytic activities, it can be expected that PDE1 inhibition might be beneficial in combination with cGMP-elevating drugs for the treatment of cardiac diseases.


Subject(s)
Myocytes, Cardiac , Phosphoric Diester Hydrolases , Adult , Mice , Humans , Animals , Phosphoric Diester Hydrolases/metabolism , Myocytes, Cardiac/metabolism , Cyclic GMP/metabolism , Cyclic AMP/metabolism , Phosphodiesterase Inhibitors/pharmacology , Calmodulin/metabolism
2.
Biochim Biophys Acta Biomembr ; 1860(2): 329-334, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29038022

ABSTRACT

The present study demonstrated for the first time the interaction between adenosine 3',5'-cyclic monophosphate (cAMP), one of the most important signaling compounds in living organisms, and the mitochondria-targeted antioxidant plastoquinonyl-decyltriphenylphosphonium (SkQ1). The data obtained on model liquid membranes and human platelets revealed the ability of SkQ1 to selectively transport cAMP, but not guanosine 3',5'-cyclic monophosphate (cGMP), across both artificial and natural membranes. In particular, SkQ1 elicited translocation of cAMP from the source to the receiving phase of a Pressman-type cell, while showing low activity with cGMP. Importantly, only conjugate with plastoquinone, but not dodecyl-triphenylphosphonium, was effective in carrying cAMP. In human platelets, SkQ1 also appeared to serve as a carrier of cAMP, but not cGMP, from outside to inside the cell, as measured by phosphorylation of the vasodilator stimulated phosphoprotein. The SkQ1-induced transfer of cAMP across the plasma membrane found here can be tentatively suggested to interfere with cAMP signaling pathways in living cells.


Subject(s)
Cell Membrane/metabolism , Cyclic AMP/metabolism , Membranes, Artificial , Onium Compounds/metabolism , Organophosphorus Compounds/metabolism , Plastoquinone/metabolism , Animals , Biological Transport , Blood Platelets/metabolism , Cyclic GMP/metabolism , Erythrocyte Membrane/metabolism , Humans , Liposomes/metabolism , Onium Compounds/chemistry , Organophosphorus Compounds/chemistry , Phosphorylation , Plastoquinone/chemistry , Rats
3.
Biol Reprod ; 94(5): 110, 2016 05.
Article in English | MEDLINE | ID: mdl-27009040

ABSTRACT

The meiotic cell cycle of mammalian oocytes in preovulatory follicles is held in prophase arrest by diffusion of cGMP from the surrounding granulosa cells into the oocyte. Luteinizing hormone (LH) then releases meiotic arrest by lowering cGMP in the granulosa cells. The LH-induced reduction of cGMP is caused in part by a decrease in guanylyl cyclase activity, but the observation that the cGMP phosphodiesterase PDE5 is phosphorylated during LH signaling suggests that an increase in PDE5 activity could also contribute. To investigate this idea, we measured cGMP-hydrolytic activity in rat ovarian follicles. Basal activity was due primarily to PDE1A and PDE5, and LH increased PDE5 activity. The increase in PDE5 activity was accompanied by phosphorylation of PDE5 at serine 92, a protein kinase A/G consensus site. Both the phosphorylation and the increase in activity were promoted by elevating cAMP and opposed by inhibiting protein kinase A, supporting the hypothesis that LH activates PDE5 by stimulating its phosphorylation by protein kinase A. Inhibition of PDE5 activity partially suppressed LH-induced meiotic resumption as indicated by nuclear envelope breakdown, but inhibition of both PDE5 and PDE1 activities was needed to completely inhibit this response. These results show that activities of both PDE5 and PDE1 contribute to the LH-induced resumption of meiosis in rat oocytes, and that phosphorylation and activation of PDE5 is a regulatory mechanism.


Subject(s)
Cyclic GMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 1/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Luteinizing Hormone/pharmacology , Meiosis/drug effects , Ovarian Follicle/drug effects , Animals , Cells, Cultured , Female , Mice , Mice, Inbred C57BL , Oocytes/drug effects , Oocytes/metabolism , Ovarian Follicle/metabolism , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley
4.
Proc Natl Acad Sci U S A ; 111(35): 12925-9, 2014 Sep 02.
Article in English | MEDLINE | ID: mdl-25139994

ABSTRACT

Conflicting results have been reported for the roles of cGMP and cGMP-dependent protein kinase I (cGKI) in various pathological conditions leading to cardiac hypertrophy and fibrosis. A cardioprotective effect of cGMP/cGKI has been reported in whole animals and isolated cardiomyocytes, but recent evidence from a mouse model expressing cGKIß only in smooth muscle (ßRM) but not in cardiomyocytes, endothelial cells, or fibroblasts has forced a reevaluation of the requirement for cGKI activity in the cardiomyocyte antihypertrophic effects of cGMP. In particular, ßRM mice developed the same hypertrophy as WT controls when subjected to thoracic aortic constriction or isoproterenol infusion. Here, we challenged ßRM and WT (Ctr) littermate control mice with angiotensin II (AII) infusion (7 d; 2 mg ⋅ kg(-1) ⋅ d(-1)) to induce hypertrophy. Both genotypes developed cardiac hypertrophy, which was more pronounced in Ctr animals. Cardiomyocyte size and interstitial fibrosis were increased equally in both genotypes. Addition of sildenafil, a phosphodiesterase 5 (PDE5) inhibitor, in the drinking water had a small effect in reducing myocyte hypertrophy in WT mice and no effect in ßRM mice. However, sildenafil substantially blocked the increase in collagen I, fibronectin 1, TGFß, and CTGF mRNA in Ctr but not in ßRM hearts. These data indicate that, for the initial phase of AII-induced cardiac hypertrophy, lack of cardiomyocyte cGKI activity does not worsen hypertrophic growth. However, expression of cGKI in one or more cell types other than smooth muscle is necessary to allow the antifibrotic effect of sildenafil.


Subject(s)
Angiotensin II/pharmacology , Cardiomegaly/metabolism , Cyclic GMP-Dependent Protein Kinase Type I/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Hypertension/metabolism , Animals , Cardiomegaly/chemically induced , Cyclic GMP/metabolism , Fibrosis/chemically induced , Fibrosis/metabolism , Genetic Markers , Hypertension/chemically induced , Mice , Muscle, Smooth/metabolism , Myocardial Contraction/drug effects , Myocardial Contraction/physiology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Nitric Oxide/metabolism , Phosphodiesterase 5 Inhibitors/pharmacology , Piperazines/pharmacology , Purines/pharmacology , Sildenafil Citrate , Sulfones/pharmacology , Vasoconstrictor Agents/pharmacology
5.
Trends Pharmacol Sci ; 35(8): 404-13, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24948380

ABSTRACT

cGMP inhibits hypertrophy, decreases fibrosis, and protects against cardiac ischemia-reperfusion (I/R) injury. Gene-targeting studies have not defined a clear role for its major downstream effector, cGMP-dependent protein kinase I (cGKI), in cardiac hypertrophy, but do implicate cGMP-cGKI signaling in fibrosis and I/R injury. No direct cGKI activators have advanced to clinical trials, whereas cardiac trials of agents that modulate cGMP via particulate or soluble guanylyl cyclases (GCs) and phosphodiesterase 5 (PDE5) are ongoing. Here we review concerns arising from preclinical and clinical studies that question whether targeting the cGMP pathway remains an encouraging concept for management of heart dysfunction. So far, trial results for GC modulators are inconclusive, and sildenafil, a PDE5 inhibitor, although cardioprotective in mouse models, has not shown positive clinical results. Preclinical cardioprotection observed for sildenafil may result from inhibition of PDE5 in non-cardiomyocytes or off-target effects, possibly on PDE1C. On the basis of such mechanistic considerations, re-evaluation of the cellular localization of drug target(s) and intervention protocols for cGMP-elevating agents may be needed.


Subject(s)
Cyclic GMP/metabolism , Heart Diseases/drug therapy , Heart Diseases/metabolism , Animals , Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Heart Diseases/enzymology , Humans , Phosphodiesterase 5 Inhibitors/pharmacology
6.
Methods Mol Biol ; 1020: 51-62, 2013.
Article in English | MEDLINE | ID: mdl-23709025

ABSTRACT

Cyclic nucleotides (cAMP and cGMP) as second messengers regulate a wide variety of biological processes such as cellular growth, secretary signaling, and neuroplasticity. These processes can be regulated by increasing the synthesis of cyclic nucleotides (cyclases), by regulation of cAMP and cGMP effector proteins such as cAMP- and cGMP-dependent protein kinases, or by regulation of cyclic nucleotide degradation via cyclic nucleotide phosphodiestases (PDEs). At present PDEs are classified into 11 gene families, each containing several different isoforms and splice variants. All PDEs share considerable homology in their catalytic domains but substantially differ in their N-terminal regions, that contain different types of regulatory. The different PDEs show complex substrate specificity. PDE5, PDE6, and PDE9 are considered to be cGMP specific, while PDE1, PDE2, PDE3, PDE10, and PDE11 can hydrolyze both cGMP and cAMP. PDE4, PDE7, and PDE8 use mainly cAMP as their substrates at physiological substrate levels. Here we describe two methods designed for measuring cGMP (cAMP) hydrolytic activities. The first one is a traditional method using radioactive substrates and the second one is a recently developed nonradioactive method based on Isothermal Titration Calorimetry.


Subject(s)
Cyclic GMP/metabolism , Enzyme Assays/methods , Phosphoric Diester Hydrolases/metabolism , Calorimetry/methods , Cyclic AMP/metabolism , Hydrolysis , Kinetics , Substrate Specificity , Temperature
7.
Proc Natl Acad Sci U S A ; 107(12): 5646-51, 2010 Mar 23.
Article in English | MEDLINE | ID: mdl-20212138

ABSTRACT

It has been suggested that cGMP kinase I (cGKI) dampens cardiac hypertrophy. We have compared the effect of isoproterenol (ISO) and transverse aortic constriction (TAC) on hypertrophy in WT [control (CTR)] mice, total cGKI-KO mice, and cGKIbeta rescue mice (betaRM) lacking cGKI specifically in cardiomyocytes (CMs). Infusion of ISO did not change the expression of cGKI in the hearts of CTR mice or betaRM but raised the heart weight by approximately 20% in both. An identical hypertrophic growth response was measured in CMs from CTR mice and betaRM and in isolated adult CMs cultured with or without 1 muM ISO. In both genotypes, ISO infusion induced similar changes in the expression of hypertrophy-associated cardiac genes and significant elevation of serum atrial natriuretic peptide and total cardiac cGMP. No differences in cardiac hypertrophy were obtained by 7-day ISO infusion in 4- to 6-week-old conventional cGKI-KO and CTR mice. Furthermore, TAC-induced hypertrophy of CTR mice and betaRM was not different and did not result in changes of the cGMP-hydrolyzing phosphodiesterase activities in hypertropic hearts or CMs. These results strongly suggest that cardiac myocyte cGKI does not affect the development of heart hypertrophy induced by pressure overload or chronic ISO infusion.


Subject(s)
Cardiomegaly/enzymology , Cardiomegaly/etiology , Cyclic GMP-Dependent Protein Kinases/deficiency , Myocytes, Cardiac/enzymology , Animals , Base Sequence , Cardiomegaly/genetics , Cardiomegaly/pathology , Cells, Cultured , Cyclic GMP-Dependent Protein Kinase Type I , Cyclic GMP-Dependent Protein Kinases/genetics , DNA Primers/genetics , Gene Expression , Isoproterenol/pharmacology , Mice , Mice, Knockout , Models, Cardiovascular , Myocytes, Cardiac/pathology , Myocytes, Smooth Muscle/enzymology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction
8.
Mol Pharmacol ; 77(4): 670-7, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20086037

ABSTRACT

cGMP-specific phosphodiesterase (PDE5) has become a target for drug development for the treatment of a number of physiological dysfunctions, affected by changes in the cGMP/cGMP-dependent protein kinase (PKG) signaling pathway. PDE5 has two highly homologous regulatory domains, GAF-A and GAF-B. We showed previously that PDE5 could be converted from a low-activity (nonactivated) state to a high-activity state upon cGMP binding to the GAF-A domain with higher sensitivities toward sildenafil (EMBO J 22:469-478, 2003). Here we investigated whether sildenafil sensitivity of PDE5 could be modified by cGMP-independent mechanisms. Individually expressed recombinant GAF-A and GAF-B proteins were tested for their ability to modulate full-length recombinant PDE5 affinity to sildenafil. The GAF-A domain protein had the most dramatic effect on the affinity of the nonactivated recombinant PDE5 for sildenafil, revealing much higher sensitivity to sildenafil inhibition. The apparent affinity for sildenafil increased from the nanomolar range to the picomolar range, providing evidence for the presence of a "super-high" sensitivity state of PDE5 for sildenafil inhibition. In human platelet, higher sensitivity of PDE5 for sildenafil inhibition has been detected after blocking cGMP-binding sites of the GAF-A domain. Thus, our data demonstrate that high sensitivity of PDE5 for sildenafil can be obtained not only through cGMP-induced activation of PDE, but also through cGMP-independent modulation of PDE5 in the nonactivated state, possibly through protein-protein interaction. Furthermore, data suggest that nonactivated PDE5 with "super-high" affinities for sildenafil inhibition may be responsible for therapeutic effects of long-term treatments with low doses of PDE5 inhibitors.


Subject(s)
Cyclic GMP/physiology , Phosphodiesterase 5 Inhibitors , Phosphodiesterase Inhibitors/pharmacology , Piperazines/pharmacology , Sulfones/pharmacology , Animals , Blood Platelets/enzymology , Cells, Cultured , Cyclic Nucleotide Phosphodiesterases, Type 5/chemistry , Humans , Mice , Protein Conformation , Protein Structure, Tertiary , Purines/pharmacology , Sildenafil Citrate
9.
Circ Res ; 105(10): 956-64, 2009 Nov 06.
Article in English | MEDLINE | ID: mdl-19797176

ABSTRACT

RATIONALE: Cyclic nucleotide phosphodiesterases (PDEs) through the degradation of cGMP play critical roles in maintaining cardiomyocyte homeostasis. Ca(2+)/calmodulin (CaM)-activated cGMP-hydrolyzing PDE1 family may play a pivotal role in balancing intracellular Ca(2+)/CaM and cGMP signaling; however, its function in cardiomyocytes is unknown. OBJECTIVE: Herein, we investigate the role of Ca(2+)/CaM-stimulated PDE1 in regulating pathological cardiomyocyte hypertrophy in neonatal and adult rat ventricular myocytes and in the heart in vivo. METHODS AND RESULTS: Inhibition of PDE1 activity using a PDE1-selective inhibitor, IC86340, or downregulation of PDE1A using siRNA prevented phenylephrine induced pathological myocyte hypertrophy and hypertrophic marker expression in neonatal and adult rat ventricular myocytes. Importantly, administration of the PDE1 inhibitor IC86340 attenuated cardiac hypertrophy induced by chronic isoproterenol infusion in vivo. Both PDE1A and PDE1C mRNA and protein were detected in human hearts; however, PDE1A expression was conserved in rodent hearts. Moreover, PDE1A expression was significantly upregulated in vivo in the heart and myocytes from various pathological hypertrophy animal models and in vitro in isolated neonatal and adult rat ventricular myocytes treated with neurohumoral stimuli such as angiotensin II (Ang II) and isoproterenol. Furthermore, PDE1A plays a critical role in phenylephrine-induced reduction of intracellular cGMP- and cGMP-dependent protein kinase (PKG) activity and thereby cardiomyocyte hypertrophy in vitro. CONCLUSIONS: These results elucidate a novel role for Ca(2+)/CaM-stimulated PDE1, particularly PDE1A, in regulating pathological cardiomyocyte hypertrophy via a cGMP/PKG-dependent mechanism, thereby demonstrating Ca(2+) and cGMP signaling cross-talk during cardiac hypertrophy.


Subject(s)
Calcium Signaling/radiation effects , Calcium/metabolism , Calmodulin/metabolism , Cardiomegaly/enzymology , Cyclic Nucleotide Phosphodiesterases, Type 1/biosynthesis , Myocytes, Cardiac/enzymology , Second Messenger Systems/physiology , Angiotensin II/metabolism , Animals , Calcium Signaling/drug effects , Cardiomegaly/chemically induced , Cardiotonic Agents/adverse effects , Cardiotonic Agents/pharmacology , Cells, Cultured , Cyclic GMP/metabolism , Enzyme Inhibitors/adverse effects , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/physiology , Heart Ventricles/enzymology , Humans , Isoproterenol/adverse effects , Isoproterenol/pharmacology , Male , Mice , Rats , Rats, Sprague-Dawley , Second Messenger Systems/drug effects
11.
Eur J Pharmacol ; 521(1-3): 105-14, 2005 Oct 03.
Article in English | MEDLINE | ID: mdl-16182282

ABSTRACT

Sildenafil (Viagra), a selective inhibitor of phosphodiesterase 5 (PDE5), induces headache and migraine. Although previously supposed to be a "vascular" headache, no significant cerebral artery dilatation was found in vivo. Thus, we hypothesised that PDE5 may not be present or that sildenafil is less effective on the cGMP hydrolysis in cerebral arteries, and that sildenafil may not be an effective dilator of cerebral arteries under baseline conditions. We evaluated the presence of PDE5 mRNA and protein in human arteries. Furthermore, the effects of two selective PDE5 inhibitors, sildenafil and UK-114,542, and a PDE1 inhibitor UK-90,234 on cGMP hydrolysis were investigated in human and guinea pig cerebral arteries. The vasoactive responses of the compounds were evaluated in guinea pig basilar arteries in vitro, with concomitant measurements of cAMP and cGMP. PDE5 was found in human middle cerebral arteries. Sildenafil and UK-114,542 inhibited cGMP hydrolysis concentration-dependently in both species. In guinea pig arteries, sildenafil induced an endothelium-dependent vasodilatation only at concentrations above 10 nM, which was augmented by sodium nitroprusside and attenuated by reduction of cGMP, but was cGMP independent at high concentrations. UK-114,542 was more and UK-90,234 was less potent than sildenafil. In conclusion, PDE5 is present in human and guinea pig cerebral arteries, and is inhibited by sildenafil at micromolar levels. Sildenafil in vitro is a poor dilator of guinea pig cerebral arteries unless a nitric oxide donor is co-administered, corresponding to the previous findings in vivo.


Subject(s)
3',5'-Cyclic-GMP Phosphodiesterases/antagonists & inhibitors , Cerebral Arteries/drug effects , Piperazines/pharmacology , 3',5'-Cyclic-GMP Phosphodiesterases/genetics , 3',5'-Cyclic-GMP Phosphodiesterases/metabolism , Aged , Aged, 80 and over , Animals , Basilar Artery/drug effects , Basilar Artery/enzymology , Basilar Artery/physiology , Blotting, Western , Cerebral Arteries/enzymology , Cerebral Arteries/physiology , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 5 , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Female , Guanylate Cyclase/antagonists & inhibitors , Guinea Pigs , Humans , Hydrolysis/drug effects , In Vitro Techniques , Infant , Male , Morpholines/pharmacology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/enzymology , Nitroprusside/pharmacology , Oxadiazoles/pharmacology , Purines , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Pyrimidinones/pharmacology , Quinoxalines/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sildenafil Citrate , Sulfones , Vasodilation/drug effects , Vasodilator Agents/pharmacology
12.
J Mol Cell Cardiol ; 38(1): 175-84, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15623434

ABSTRACT

Angiotensin II (Ang II) and nitric oxide (NO)/natriuretic peptide (NP) signaling pathways mutually regulate each other. Imbalance of Ang II and NO/NP has been implicated in the pathophysiology of many vascular diseases. cGMP functions as a key mediator in the interaction between Ang II and NO/NP. Cyclic nucleotide phosphodiesterase 5A (PDE5A) is important in modulating cGMP signaling by hydrolyzing cGMP in vascular smooth muscle cells (VSMC). Therefore, we examined whether Ang II negatively modulates intracellular cGMP signaling in VSMC by regulating PDE5A. Ang II rapidly and transiently increased PDE5A mRNA levels in rat aortic VSMC. Upregulation of PDE5A mRNA was associated with a time-dependent increase of both PDE5 protein expression and activity. Increased PDE5A mRNA level was transcription-dependent and mediated by the Ang II type 1 receptor. Ang II-mediated activation of extracellular signal-regulated kinases 1/2 (ERK1/2) was essential for Ang II-induced PDE5A upregulation. Pretreatment of VSMC with Ang II inhibited C-type NP (CNP) stimulated cGMP signaling, such as cGMP dependent protein kinase (PKG)-mediated phosphorylation of vasodilator-stimulated-phosphoprotein (VASP). Ang II-mediated inhibition of PKG was blocked when PDE5 activity was decreased by selective PDE5 inhibitors, suggesting that upregulation of PDE5A expression is an important mechanism for Ang II to attenuate cGMP signaling. PDE5A may also play a critical role in the growth promoting effects of Ang II because inhibition of PDE5A activity significantly decreased Ang II-stimulated VSMC growth. These observations establish a new mechanism by which Ang II antagonizes cGMP signaling and stimulates VSMC growth.


Subject(s)
3',5'-Cyclic-GMP Phosphodiesterases/metabolism , Angiotensin II/pharmacology , Cyclic GMP/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Signal Transduction/drug effects , 3',5'-Cyclic-GMP Phosphodiesterases/antagonists & inhibitors , 3',5'-Cyclic-GMP Phosphodiesterases/genetics , Animals , Cell Proliferation/drug effects , Cells, Cultured , Cyclic Nucleotide Phosphodiesterases, Type 5 , Extracellular Signal-Regulated MAP Kinases/metabolism , Male , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/enzymology , Protein Kinase Inhibitors/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Transcription, Genetic/drug effects , Transcription, Genetic/genetics , Up-Regulation/drug effects
13.
Cell ; 118(3): 375-87, 2004 Aug 06.
Article in English | MEDLINE | ID: mdl-15294162

ABSTRACT

The G protein-coupled, receptor-activated phosphoinositide 3-kinase gamma (PI3Kgamma) mediates inflammatory responses and negatively controls cardiac contractility by reducing cAMP concentration. Here, we report that mice carrying a targeted mutation in the PI3Kgamma gene causing loss of kinase activity (PI3KgammaKD/KD) display reduced inflammatory reactions but no alterations in cardiac contractility. We show that, in PI3KgammaKD/KD hearts, cAMP levels are normal and that PI3Kgamma-deficient mice but not PI3KgammaKD/KD mice develop dramatic myocardial damage after chronic pressure overload induced by transverse aortic constriction (TAC). Finally, our data indicate that PI3Kgamma is an essential component of a complex controlling PDE3B phosphodiesterase-mediated cAMP destruction. Thus, cardiac PI3Kgamma participates in two distinct signaling pathways: a kinase-dependent activity that controls PKB/Akt as well as MAPK phosphorylation and contributes to TAC-induced cardiac remodeling, and a kinase-independent activity that relies on protein interactions to regulate PDE3B activity and negatively modulates cardiac contractility.


Subject(s)
Isoenzymes/metabolism , Myocardium/metabolism , Phosphatidylinositol 3-Kinases/metabolism , 3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Animals , Cell Movement/physiology , Class Ib Phosphatidylinositol 3-Kinase , Cyclic AMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 3 , Hypertension/metabolism , Isoenzymes/genetics , Leukocytes/physiology , Mice , Mitogen-Activated Protein Kinases/metabolism , Myocardium/enzymology , Phosphatidylinositol 3-Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Signal Transduction/physiology
14.
Circ Res ; 93(4): 280-91, 2003 Aug 22.
Article in English | MEDLINE | ID: mdl-12933699

ABSTRACT

Cyclic GMP (cGMP) made in response to atrial natriuretic peptide (ANP) or nitric oxide (NO) is an important regulator of short-term changes in smooth muscle tone and longer-term responses to chronic drug treatment or proliferative signals. The ability of smooth muscle cells (SMCs) to utilize different combinations of phosphodiesterase (PDE) isozymes allows cGMP to mediate these multiple processes. For example, PDE5 as a major cGMP-hydrolyzing PDE effectively controls the development of smooth muscle relaxation. In order for contraction to occur, PDE5 is activated and cGMP falls. Conversely, blockade of PDE5 activity allows the relaxation cycle to be prolonged and enhanced. A recently shown direct activation of PDE5 by cGMP binding to the GAF A domain suggests that this regulatory site might be a target for new drug development. The calcium surge associated with vasoconstrictor initiated contraction also activates a calcium/calmodulin-dependent PDE (PDE1A). Together, PDE5 and PDE1A lower cGMP sufficiently to allow contraction. Longer term, both PDE5 and PDE1A mRNA are induced by chronic stimulation of guanylyl cyclase. This induction is a major cause of the tolerance that develops to NO-releasing drugs. Finally, high levels of cGMP or cAMP also act as a brake to attenuate the proliferative response of SMCs to many mitogens. After vessel damage, in order for SMC proliferation to occur, the levels of cGMP and cAMP must be decreased. In humans, this decrease is caused in large part by induction of another Ca2+/calmodulin-dependent PDE (PDE1C) that allows the brake to be released and proliferation to start.


Subject(s)
3',5'-Cyclic-GMP Phosphodiesterases/metabolism , Muscle, Smooth, Vascular/physiology , 3',5'-Cyclic-GMP Phosphodiesterases/genetics , Animals , Cell Division/physiology , Cyclic GMP/metabolism , Enzyme Activation , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Multigene Family/genetics , Muscle, Smooth, Vascular/cytology
15.
J Neurosci ; 23(16): 6452-9, 2003 Jul 23.
Article in English | MEDLINE | ID: mdl-12878685

ABSTRACT

The nitric oxide (NO)-cGMP pathway has been implicated as playing a crucial role in the induction of cerebellar long-term depression (LTD). The amplitude and duration of the cGMP signal is controlled by cyclic nucleotide phosphodiesterases (PDEs). Here we identify PDE5 and PDE1B as the two major cGMP-hydrolyzing PDEs specifically and differentially expressed in the Purkinje neurons of mouse cerebellum. PDE5 was found in all Purkinje neurons, whereas PDE1B was detected only in a subset of these cells, suggesting that individual Purkinje cells may differentially regulate cGMP, depending on the PDE isozymes expressed. Although expression of guanylate cyclase and/or cGMP-dependent protein kinase (PKG) in Purkinje cells have been reported, neither cGMP accumulation nor PKG activation in these cells in vivo has been demonstrated. To determine if changes in PKG activation and PDE5 regulation occur in vivo we have examined the phosphorylation of PDE5 in mouse cerebellar Purkinje cells by immunocytochemistry and Western blot analyses using a phosphospecific PDE5 antibody. Injection of sodium nitroprusside or selective PKG activators into the lateral ventricle of mouse brain induced PDE5 phosphorylation in vivo, but was completely missing in Purkinje cell-specific PKG I knock-out mice. In cerebellar slices, treatment with sildenafil or IBMX led to different levels of phospho-PDE5 accumulation and activation of PDE5. These results suggest that phosphorylation of PDE5 in Purkinje neurons after cGMP-PKG activation performs a critical role in the termination of the cGMP signal during LTD progression; moreover, PDE5 phosphorylation may be used as an in vivo indicator for PKG activation.


Subject(s)
3',5'-Cyclic-GMP Phosphodiesterases/metabolism , Cerebellum/metabolism , Cyclic GMP-Dependent Protein Kinases/metabolism , Cyclic GMP/analogs & derivatives , Purkinje Cells/metabolism , 3',5'-Cyclic-GMP Phosphodiesterases/antagonists & inhibitors , 3',5'-Cyclic-GMP Phosphodiesterases/chemistry , Animals , Antibody Specificity , Cerebellum/chemistry , Cerebellum/cytology , Cyclic GMP/metabolism , Cyclic GMP/pharmacology , Cyclic GMP-Dependent Protein Kinases/drug effects , Cyclic Nucleotide Phosphodiesterases, Type 1 , Cyclic Nucleotide Phosphodiesterases, Type 5 , Cytosol/chemistry , Cytosol/enzymology , Enzyme Activators/pharmacology , Enzyme Inhibitors , Immunohistochemistry , Injections, Intraventricular , Long-Term Synaptic Depression/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitroprusside/pharmacology , Phosphoric Diester Hydrolases/metabolism , Phosphorylation/drug effects , Purkinje Cells/enzymology
16.
EMBO J ; 22(3): 469-78, 2003 Feb 03.
Article in English | MEDLINE | ID: mdl-12554648

ABSTRACT

cGMP-specific, cGMP-binding phosphodiesterase (PDE5) regulates such physiological processes as smooth muscle relaxation and neuronal survival. PDE5 contains two N-terminal domains (GAF A and GAF B), but the functional roles of these domains have not been determined. Here we show that recombinant PDE5 is activated directly upon cGMP binding to the GAF A domain, and this effect does not require PDE5 phosphorylation. PDE5 exhibited time- and concentration-dependent reversible activation in response to cGMP, with the highest activation (9- to 11-fold) observed at low substrate concentrations (0.1 micro M cGMP). A monoclonal antibody directed against GAF A blocked cGMP binding, prevented PDE5 activation and decreased basal activity, revealing that PDE5 in its non-activated state has low intrinsic catalytic activity. Activated PDE5 showed higher sensitivity towards sildenafil than non-activated PDE5. The stimulatory effect of cGMP binding on the catalytic activity of PDE5 suggests that this mechanism of enzyme activation may be common among other GAF domain-containing proteins. The data also suggest that development of agonists and antagonists of PDE5 activity based on binding to this site might be possible.


Subject(s)
3',5'-Cyclic-GMP Phosphodiesterases/metabolism , Cyclic GMP/metabolism , 3',5'-Cyclic-GMP Phosphodiesterases/chemistry , 3',5'-Cyclic-GMP Phosphodiesterases/genetics , Animals , Antibodies, Monoclonal/metabolism , Cell Line , Cyclic Nucleotide Phosphodiesterases, Type 5 , Enzyme Activation , Humans , Mice , Mutagenesis, Site-Directed , Phosphodiesterase Inhibitors/metabolism , Phosphorylation , Piperazines/metabolism , Protein Binding , Protein Structure, Tertiary , Purines , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sildenafil Citrate , Sulfones
17.
Circ Res ; 90(2): 151-7, 2002 Feb 08.
Article in English | MEDLINE | ID: mdl-11834707

ABSTRACT

Proliferation of arterial smooth muscle cells (SMCs) is a key event in the formation of advanced atherosclerotic lesions and restenosis after angioplasty. Cyclic nucleotides (cAMP and cGMP) inhibit arterial SMC proliferation, and elevation of cyclic nucleotides reduces neointimal formation after angioplasty in animal models. Degradation of cAMP and cGMP is catalyzed by cyclic nucleotide phosphodiesterases (PDEs). One of these, PDE1C, hydrolyzes cAMP and cGMP and is expressed in proliferating human SMCs but is absent in quiescent human aorta. Thus, PDE1C expression is low in cultured human SMCs made quiescent by attaching to fibrillar collagen type I. After release from the fibrillar collagen, PDE1C expression is induced and associated with traverse through S-phase of the cell cycle. Further, PDE1C is expressed in vivo in human fetal aorta containing proliferating SMCs, but not in newborn aorta in which SMC proliferation has ceased. Inhibition of PDE1C in SMCs isolated from normal aorta or from lesions of atherosclerosis using antisense oligonucleotides or a PDE1 inhibitor results in suppression of SMC proliferation. In conclusion, PDE1C expression is a marker of human SMC proliferation ex vivo and in vivo. Inhibition of PDE1C leads to inhibition of human SMC proliferation. Because PDE1C is absent in quiescent SMCs, PDE1C inhibitors may target proliferating SMCs in lesions of atherosclerosis or restenosis.


Subject(s)
Muscle, Smooth, Vascular/enzymology , Phosphoric Diester Hydrolases/metabolism , Aorta/cytology , Aorta/embryology , Aorta/enzymology , Cell Division/drug effects , Cell Division/physiology , Cells, Cultured , Collagen Type I/pharmacology , Cyclic Nucleotide Phosphodiesterases, Type 1 , Enzyme Induction/drug effects , Enzyme Induction/physiology , Enzyme Inhibitors/pharmacology , Humans , Infant, Newborn , Isoenzymes/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects
18.
J Biol Chem ; 277(5): 3310-7, 2002 Feb 01.
Article in English | MEDLINE | ID: mdl-11723116

ABSTRACT

Nitric oxide and endogenous nitrovasodilators regulate smooth muscle tone by elevation of cGMP and activation of cyclic GMP-dependent protein kinase (PKG). The amplitude and duration of the cGMP signal in smooth muscle is regulated in large part by cGMP-specific cyclic nucleotide phosphodiesterase (PDE5). Previous in vitro data have suggested that both cAMP-dependent protein kinase and PKG can regulate the activity of PDE5. To test if this type of regulation is important in the intact cell, we have generated phospho-PDE5-specific antisera and have utilized isolated smooth muscle cells from mice having a disruption in the PKG I gene as well as cells from normal human smooth muscle. The data show that in human smooth muscle cells, activation of PKG by 8-Br-cGMP led to phosphorylation and activation of PDE5. In the same cells, 8-Br-cAMP had no significant effect on PDE5 phosphorylation. Treatment of wild-type mouse aortic smooth muscle cells with 8-Br-cGMP also induced the phosphorylation of PDE5, whereas no phosphorylation was seen in smooth muscle cells isolated from mice in which the gene for PKG I had been disrupted. As with the human cells, no phosphorylation was seen in the mouse cells in response to 8-Br-cAMP. These results strongly suggest that a major regulatory pathway for control of PDE5 phosphorylation and activity in intact smooth muscle is via PKG-dependent phosphorylation of PDE5. Finally, experiments with calyculin A and okadaic acid suggest that PP1 phosphatase, the catalytic subunit of myosin phosphatase, can regulate PDE5 dephosphorylation. Together, the data suggest that phosphorylation and activation of PDE5 by PKG I and its subsequent dephosphorylation by myosin phosphatase may be key steps in the regulation of relaxation/contraction cycles of smooth muscle.


Subject(s)
3',5'-Cyclic-GMP Phosphodiesterases/metabolism , Muscle, Smooth/enzymology , Uterus/enzymology , 3',5'-Cyclic-GMP Phosphodiesterases/chemistry , Antibodies , Cells, Cultured , Chromatography, High Pressure Liquid , Cyclic Nucleotide Phosphodiesterases, Type 5 , Female , Homeostasis , Humans , Kinetics , Muscle, Smooth/cytology , Muscle, Smooth/physiology , Phosphopeptides/chemistry , Phosphopeptides/immunology , Phosphoric Diester Hydrolases/metabolism , Phosphorylation , Uterine Contraction , Uterus/cytology , Uterus/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...