Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters








Publication year range
1.
J Thromb Haemost ; 15(7): 1484-1486, 2017 07.
Article in English | MEDLINE | ID: mdl-28453902
2.
J Thromb Haemost ; 11(5): 941-50, 2013 May.
Article in English | MEDLINE | ID: mdl-23413961

ABSTRACT

BACKGROUND: Infective endocarditis (IE) is characterized by thrombus formation on a cardiac valve. The oral bacterium, Streptococcus oralis, is recognized for its ability to colonize damaged heart valves and is frequently isolated from patients with IE. Platelet interaction with S. oralis leads to the development of a thrombotic vegetation on heart valves, which results in valvular incompetence and congestive heart failure. OBJECTIVE: To investigate the mechanism through which platelets become activated upon binding S. oralis. PATIENTS AND METHODS: Platelet interactions with immobilized bacteria under shear conditions were assessed using a parallel flow chamber. S. oralis-inducible platelet reactivity was determined using light transmission aggregometry. Dense granule secretion was measured by luminometry using a luciferin/luciferase assay. RESULTS: Using shear rates that mimic physiological conditions, we demonstrated that S. oralis was able to support platelet adhesion under venous (50-200 s(-1) ) and arterial shear conditions (800 s(-1) ). Platelets rolled along immobilized S. oralis through an interaction with GPIbα. Following rolling, platelet microaggregate formation was observed on immobilized S. oralis. Aggregate formation was dependent on S. oralis binding IgG, which cross-links to platelet FcγRIIa. This interaction led to phosphorylation of the ITAM domain on FcγRIIa, resulting in dense granule secretion, amplification through the ADP receptor and activation of RAP1, culminating in platelet microaggregate formation. CONCLUSIONS: These results suggest a model of interaction between S. oralis and platelets that leads to the formation of a stable septic vegetation on damaged heart valves.


Subject(s)
Platelet Activation/physiology , Platelet Glycoprotein GPIb-IX Complex/physiology , Receptors, IgG/physiology , Streptococcus oralis/physiology , Cell Adhesion , Endocarditis/blood , Endocarditis/microbiology , Humans , Platelet Aggregation
3.
J Thromb Haemost ; 11(1): 161-8, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23140275

ABSTRACT

BACKGROUND: Platelets are highly specialized cells that regulate hemostasis and thrombosis in the vasculature. Upon activation, platelets release various granules that impact on platelets, the coagulation system, other blood cells and the vessel wall; however, the mechanisms controlling granule release are only partially known. We have shown previously that synaptotagmin-like protein (Slp)1 decreases dense granule release in platelets. OBJECTIVES: To determine the role of other Slps and their binding partners on platelet dense granule release. METHODS: RT-PCR and immunoblotting were used to identify Slps in human platelets. Interaction between Slp4 and Rab8 was investigated with pull-down assays, coimmunoprecipitation, and confocal microscopy. Secretion assays on permeabilized platelets were performed to investigate the effects of Slp4 and Rab8 on dense granule release. RESULTS: Slp4 mRNA and protein are expressed in human platelets. Slp4 interacts with Rab8 in transfected cells and at endogenous protein levels in platelets. We mapped the Rab interaction site to the Slp-homology domain of Slp4, and showed preferential binding of Slp4 to the GTP-bound form of Rab8. Live microscopy showed colocalization of green fluorescent protein-Slp4 and mCherry-Rab8 at the plasma membrane of transfected cells. Endogenous platelet Slp4 and Rab8 colocalized in the center of activated platelets, where granule secretion takes place. Secretion assays revealed that Slp4 and Rab8 enhance dense granule release and that the Slp4 effect is dependent on Rab8 binding. CONCLUSIONS: Slp4 and Rab8 are expressed and interact in human platelets, and might be involved in dense granule release.


Subject(s)
Blood Platelets/metabolism , Platelet Activation , Secretory Vesicles/metabolism , Vesicular Transport Proteins/metabolism , rab GTP-Binding Proteins/metabolism , Blotting, Western , HEK293 Cells , HeLa Cells , Humans , Immunoprecipitation , Microscopy, Confocal , Protein Binding , Protein Interaction Domains and Motifs , Protein Interaction Mapping , RNA, Messenger/metabolism , Recombinant Fusion Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Vesicular Transport Proteins/genetics , rab GTP-Binding Proteins/genetics
4.
J Thromb Haemost ; 10(2): 167-76, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22136590

ABSTRACT

Endothelial prostacyclin and nitric oxide potently inhibit platelet functions. Prostacyclin and nitric oxide actions are mediated by platelet adenylyl and guanylyl cyclases, which synthesize cyclic AMP (cAMP) and cyclic GMP (cGMP), respectively. Cyclic nucleotides stimulate cAMP-dependent protein kinase (protein kinase A [PKA]I and PKAII) and cGMP-dependent protein kinase (protein kinase G [PKG]I) to phosphorylate a broad panel of substrate proteins. Substrate phosphorylation results in the inactivation of small G-proteins of the Ras and Rho families, inhibition of the release of Ca(2+) from intracellular stores, and modulation of actin cytoskeleton dynamics. Thus, PKA/PKG substrates translate prostacyclin and nitric oxide signals into a block of platelet adhesion, granule release, and aggregation. cAMP and cGMP are degraded by phosphodiesterases, which might restrict signaling to specific subcellular compartments. An emerging principle of cyclic nucleotide signaling in platelets is the high degree of interconnection between activating and cAMP/cGMP-dependent inhibitory signaling pathways at all levels, including cAMP/cGMP synthesis and breakdown, and PKA/PKG-mediated substrate phosphorylation. Furthermore, defects in cAMP/cGMP pathways might contribute to platelet hyperreactivity in cardiovascular disease. This article focuses on recent insights into the regulation of the cAMP/cGMP signaling network and on new targets of PKA and PKG in platelets.


Subject(s)
Blood Platelets/metabolism , Cyclic AMP-Dependent Protein Kinases/blood , Cyclic AMP/blood , Cyclic GMP-Dependent Protein Kinases/blood , Cyclic GMP/blood , Signal Transduction , Animals , Blood Platelets/drug effects , Humans , Phosphoric Diester Hydrolases/metabolism , Phosphorylation , Platelet Aggregation Inhibitors/pharmacology , Signal Transduction/drug effects
5.
J Thromb Haemost ; 8(12): 2757-65, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20946179

ABSTRACT

BACKGROUND: Sepsis is the most common manifestation of invasive pneumococcal disease and is characterized by a severe systemic inflammatory state that leads to circulatory compromise or end organ malperfusion or dysfunction. Patients suffering from sepsis often display low platelet counts characterized by thrombocytopenia as a result of platelet activation. OBJECTIVE: To investigate the mechanism through which platelets become activated in sepsis upon binding to Streptococcus pneumoniae. PATIENTS AND METHODS: We determined S. pneumoniae inducible platelet reactivity using light transmission aggregometry. Dense granule secretion was measured by luminometry using a luciferin/luciferase assay. RESULTS: Streptococcus pneumoniae induced platelet aggregation in a strain-dependent manner. Induction of aggregation was not attributable to capsule serotype, as unencapsulated strains also induced platelet aggregation. Platelet aggregation was not associated with pneumolysin toxin, as a pneumolysin-deficient mutant of S. pneumoniae induced aggregation equally as well as the parent strain. Platelet aggregation also occurred in the absence of plasma proteins or antibody, and was GPIIbIIIa dependent but aspirin independent. Toll-like receptor 2 (TLR2) is present on platelets and acts as a receptor for gram-positive bacterial lipoteichoic acid and peptidoglycan. Inhibition of TLR2 but not TLR4 (also present on platelets) completely abolished platelet aggregation. S. pneumoniae-induced platelet aggregation resulted in activation of the PI3kinase/RAP1 pathway, leading to integrin GPIIbIIIa activation and dense granule release. CONCLUSIONS: Our results demonstrate a novel interaction between S. pneumoniae and TLR2, which results in platelet activation that is likely to contribute to the thrombotic complications of sepsis.


Subject(s)
Platelet Activation/physiology , Streptococcus pneumoniae/physiology , Toll-Like Receptor 2/physiology , Blood Platelets/microbiology , Blood Proteins/physiology , Enzyme-Linked Immunosorbent Assay , Humans , Platelet Aggregation/physiology , Signal Transduction
6.
Circulation ; 103(17): 2188-94, 2001 May 01.
Article in English | MEDLINE | ID: mdl-11331261

ABSTRACT

BACKGROUND: Chronic in vivo treatment with nitroglycerin (NTG) induces tolerance to nitrates and cross-tolerance to nitrovasodilators and endothelium-derived nitric oxide (NO). We previously identified increased vascular superoxide formation and reduced NO bioavailability as one causal mechanism. It is still controversial whether intracellular downstream signaling to nitrovasodilator-derived NO is affected as well. METHODS AND RESULTS: We therefore studied the effects of 3-day NTG treatment of rats and rabbits on activity and expression of the immediate NO target soluble guanylyl cyclase (sGC) and on the cGMP-activated protein kinase I (cGK-I). Tolerance was induced either by chronic NTG infusion via osmotic minipumps (rats) or by NTG patches (rabbits). Western blot analysis, semiquantitative reverse transcription-polymerase chain reaction, and Northern blot analysis revealed significant and comparable increases in the expression of sGC alpha(1) and beta(1) subunit protein and mRNA. Studies with the oxidative fluorescent dye hydroethidine revealed an increase in superoxide in the endothelium and smooth muscle. Stimulation with NADH increased superoxide signals in both layers. Although cGK-I expression in response to low-dose NTG was not changed, a strong reduction in vasodilator-stimulated phosphoprotein (VASP) serine239 phosphorylation (specific substrate of cGK-I) was observed in tolerant tissue from rats and rabbits. Concomitant in vivo and in vitro treatment with vitamin C improved tolerance, reduced oxidative stress, and improved P-VASP. CONCLUSIONS: We therefore conclude that increased expression of sGC in the setting of tolerance reflects a chronic inhibition rather than an induction of the sGC-cGK-I pathway and may be mediated at least in part by increased vascular superoxide.


Subject(s)
Aorta/drug effects , Cell Adhesion Molecules/metabolism , Cyclic GMP-Dependent Protein Kinases/metabolism , Guanylate Cyclase/metabolism , Nitroglycerin/pharmacology , Phosphoproteins/metabolism , Vasodilator Agents/pharmacology , Administration, Cutaneous , Animals , Antioxidants/pharmacology , Aorta/enzymology , Ascorbic Acid/pharmacology , Cyclic GMP/physiology , Drug Tolerance , Enzyme Activation/drug effects , Enzyme Induction/drug effects , Infusion Pumps, Implantable , Infusions, Intravenous , Male , Microfilament Proteins , Nitric Oxide/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type III , Nitroglycerin/administration & dosage , Rabbits , Rats , Rats, Wistar , Second Messenger Systems , Superoxides/metabolism , Vasodilator Agents/administration & dosage
7.
Am J Physiol Renal Physiol ; 280(5): F851-9, 2001 May.
Article in English | MEDLINE | ID: mdl-11292628

ABSTRACT

The cGMP-cGMP-dependent protein kinase (protein kinase G) system plays an important role in the pathogenesis of mesangial proliferative glomerulonephritis. However, the molecular mechanisms of the inhibitory effects of the cGMP-protein kinase G system in the cell cycle progression of mesangial cells are not well known. To determine the inhibitory pathway of cGMP-protein kinase G in cultured mesangial cells, we investigated the effects of cGMP- and adenovirus-mediated overexpression of protein kinase G on the promoter activities of cyclin E, cyclin D1, and cyclin A. 8-Bromo-cGMP (8-BrcGMP) and overexpression of protein kinase G reduced [(3)H]thymidine uptake, reduced the numbers of cells in S and G(2)/M phases, and decreased the phosphorylation of retinoblastoma (Rb) protein. 8-BrcGMP (10(-3) M), protein kinase G adenovirus (Ad-cGKIbeta; 10(10) plaque-forming units/ml), atrial natriuretic peptide (ANP), and C-type natriuretic peptide (CNP) inhibited the promoter activity of cyclin E to 49, 57, 77, and 78%, respectively. On the other hand, the promoter activities of cyclin D1 and cyclin A were not changed significantly. In Western blot analysis, 8-BrcGMP, Ad-cGKIbeta, ANP, and CNP also inhibited cyclin E protein expression dose and time dependently. The p44/p42 mitogen-activated protein kinase (MAPK) kinase 1-p44/p42 MAPK had no effect on cyclin E promoter activities, and the cGMP-protein kinase G pathway did not change MAPK activity. In conclusion, our findings suggest that the reduction of the cyclin E promoter activity that downregulates G(1)/S transition plays a dominant role in the cGMP- and protein kinase G-induced inhibition of mesangial cell proliferation.


Subject(s)
Adenoviridae/genetics , Cyclic GMP-Dependent Protein Kinases/biosynthesis , Cyclin E/biosynthesis , Glomerular Mesangium/cytology , Glomerular Mesangium/metabolism , Animals , Cell Count , Cell Cycle/physiology , Cyclic GMP/biosynthesis , Cyclin E/genetics , Flow Cytometry , Genes, Reporter , Genetic Vectors , Glomerular Mesangium/enzymology , Luciferases , Male , Mitogen-Activated Protein Kinases/metabolism , Plasmids , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Thymidine/metabolism , Transcription, Genetic/genetics
8.
Circ Res ; 87(11): 999-1005, 2000 Nov 24.
Article in English | MEDLINE | ID: mdl-11090544

ABSTRACT

Studies with cGMP-dependent protein kinase I (cGK-I)-deficient human cells and mice demonstrated that cGK-I ablation completely disrupts the NO/cGMP pathway in vascular tissue, which indicates a key role of this protein kinase as a mediator of the NO/cGMP action. Analysis of the vasodilator-stimulated phosphoprotein phosphorylated at serine 239 (P-VASP) is a useful tool to monitor cGK-I activation in platelets and cultured endothelial and smooth muscle cells. Therefore, we investigated whether endothelial dysfunction and/or vascular NO bioavailability is reflected by decreased vessel wall P-VASP and whether improvement of endothelial dysfunction restores this P-VASP. Incubation of aortic tissue from New Zealand White Rabbits with the NOS inhibitor N:(G)-nitro-Ld-arginine and endothelial removal strikingly reduced P-VASP. Oxidative stress induced by inhibition of CuZn superoxide dismutase increased superoxide and decreased P-VASP. Endothelial dysfunction in hyperlipidemic Watanabe rabbits (WHHL) was associated with increased vascular superoxide and with decreased P-VASP. Treatment of WHHL with AT(1) receptor blockade improved endothelial dysfunction, reduced vascular superoxide, increased vascular NO bioavailability, and increased P-VASP. Therefore, the level of vessel P-VASP closely follows changes in endothelial function and vascular oxidative stress. P-VASP is suggested to represent a novel biochemical marker for monitoring the NO-stimulated sGC/cGK-I pathway and endothelial integrity in vascular tissue.


Subject(s)
Cell Adhesion Molecules/metabolism , Cyclic GMP/metabolism , Endothelium, Vascular/metabolism , Nitric Oxide/metabolism , Phosphoproteins/metabolism , Vasodilator Agents/pharmacology , Angiotensin Receptor Antagonists , Animals , Antihypertensive Agents/pharmacology , Aorta/drug effects , Aorta/metabolism , Biphenyl Compounds/pharmacology , Endothelium, Vascular/drug effects , Enzyme Inhibitors/pharmacology , Hyperlipidemias/genetics , Hyperlipidemias/metabolism , In Vitro Techniques , Irbesartan , Microfilament Proteins , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type III , Nitroarginine/pharmacology , Oxidative Stress/drug effects , Phosphorylation/drug effects , Rabbits , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Serine/metabolism , Signal Transduction/drug effects , Superoxide Dismutase/metabolism , Tetrazoles/pharmacology , Vasodilation/drug effects , Vasodilation/genetics
10.
J Biol Chem ; 275(33): 25723-32, 2000 Aug 18.
Article in English | MEDLINE | ID: mdl-10851246

ABSTRACT

cGMP-dependent protein kinase type I (cGK I), a major constituent of the atrial natriuretic peptide (ANP)/nitric oxide/cGMP signal transduction pathway, phosphorylates the vasodilator-stimulated phosphoprotein (VASP), a member of the Ena/VASP family of proteins involved in regulation of the actin cytoskeleton. Here we demonstrate that stimulation of human umbilical vein endothelial cells (HUVECs) by both ANP and 8-(4-chlorophenylthio)guanosine 3':5'-monophosphate (8-pCPT-cGMP) activates transfected cGK I and causes detachment of VASP and its known binding partner (zyxin) from focal adhesions in >60% of cells after 30 min. The ANP effects, but not the 8-pCPT-cGMP effects, reversed after 3 h of treatment. In contrast, a catalytically inactive cGK Ibeta mutant (cGK Ibeta-K405A) was incapable of mediating these effects. VASP mutated (Ser/Thr to Ala) at all three of its established phosphorylation sites (vesicular stomatitis virus-tagged VASP-AAA mutant) was not phosphorylated by cGK I and was resistant to detaching from HUVEC focal adhesions in response to 8-pCPT-cGMP. Furthermore, activation of cGK I, but not of mutant cGK Ibeta-K405A, caused a 1.5-2-fold inhibition of HUVEC migration, a dynamic process highly dependent on focal adhesion formation and disassembly. These results indicate that cGK I phosphorylation of VASP results in loss of VASP and zyxin from focal adhesions, a response that could contribute to cGK alteration of cytoskeleton-regulated processes such as cell migration.


Subject(s)
Cyclic GMP-Dependent Protein Kinases/metabolism , Endothelium, Vascular/metabolism , Adenoviridae/genetics , Atrial Natriuretic Factor/metabolism , Binding Sites , Blotting, Western , Catalysis , Cell Adhesion , Cell Adhesion Molecules/metabolism , Cell Line , Cell Movement , Cells, Cultured , Collagen/metabolism , Cyclic GMP/analogs & derivatives , Cyclic GMP/pharmacology , Cyclic GMP-Dependent Protein Kinases/genetics , Cytoskeletal Proteins , Endothelium, Vascular/cytology , Fibrinogen/metabolism , Fibronectins/metabolism , Fluorescent Antibody Technique, Indirect , Glycoproteins , Humans , Metalloproteins/metabolism , Microfilament Proteins , Mutagenesis , Phosphoproteins/metabolism , Phosphorylation , Plasmids/metabolism , Platelet Aggregation Inhibitors/pharmacology , Precipitin Tests , Thionucleotides/pharmacology , Time Factors , Umbilical Veins/metabolism , Vinculin/metabolism , Zyxin
11.
J Biol Chem ; 275(28): 21722-9, 2000 Jul 14.
Article in English | MEDLINE | ID: mdl-10783386

ABSTRACT

The potent vasodilator action of cyclic GMP-dependent protein kinase (cGK) involves decreasing the Ca(2+) sensitivity of contraction of smooth muscle via stimulation of myosin light chain phosphatase through unknown mechanisms (Wu, X., Somlyo, A. V., and Somlyo, A. P. (1996) Biochem. Biophys. Res. Commun. 220, 658-663). Myosin light chain phosphatase activity is controlled by the small GTPase RhoA and its target Rho kinase. Here we demonstrate cGMP effects mediated by cGK that inhibit RhoA-dependent Ca(2+) sensitization of contraction of blood vessels and actin cytoskeleton organization in cultured vascular myocytes. Ca(2+) sensitization and actin organization were inhibited by both 8-bromo-cGMP and sodium nitroprusside (SNP). SNP also caused translocation of activated RhoA from the membrane to the cytosol. SNP-induced actin disassembly was lost in vascular myocytes in culture after successive passages but was restored by transfection of cells with cGK I. Furthermore, cGK phosphorylated RhoA in vitro, and addition of cGK I inhibited RhoA-induced Ca(2+) sensitization in permeabilized smooth muscle. 8-Bromo-cGMP-induced actin disassembly was inhibited in vascular myocytes expressing RhoA(Ala-188), a mutant that could not be phosphorylated. Collectively, these results indicate that cGK phosphorylates and inhibits RhoA and suggest that the consequent inhibition of RhoA-induced Ca(2+) sensitization and actin cytoskeleton organization contributes to the vasodilator action of nitric oxide.


Subject(s)
Calcium Signaling/physiology , Calcium/physiology , Cyclic GMP-Dependent Protein Kinases/metabolism , Isometric Contraction/physiology , Muscle, Smooth, Vascular/physiology , rhoA GTP-Binding Protein/metabolism , Actins/drug effects , Actins/metabolism , Animals , Aorta/physiology , Cells, Cultured , Cyclic GMP/analogs & derivatives , Cyclic GMP/pharmacology , Cytoskeleton/drug effects , Cytoskeleton/physiology , Gallopamil/pharmacology , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacology , Guinea Pigs , In Vitro Techniques , Muscle, Smooth, Vascular/drug effects , Nitroprusside/pharmacology , Phenylephrine/pharmacology , Phosphorylation , Portal Vein/physiology , Pulmonary Artery/physiology , Rabbits , Rats , Rats, Wistar , Signal Transduction , Thapsigargin/pharmacology
12.
J Biol Chem ; 275(7): 5179-87, 2000 Feb 18.
Article in English | MEDLINE | ID: mdl-10671564

ABSTRACT

Endothelial nitric-oxide synthase (NOS-III) is defined as being strictly dependent on Ca(2+)/calmodulin (CaM) for activity, although NO release from endothelial cells has been reported to also occur at intracellular free Ca(2+) levels that are substimulatory for the purified enzyme. We demonstrate here that NOS-III, but neither NOS-I nor -II, is rapidly and strongly activated and phosphorylated on both Ser and Thr in the presence of cGMP-dependent protein kinase II (cGK II) and the catalytic subunit of cAMP-dependent protein kinase (cAK) in vitro. Phosphopeptide analysis by mass spectrometry identified Ser(1177), as well as Ser(633) which is situated in a recently defined CaM autoinhibitory domain within the flavin-binding region of human NOS-III. Phosphoamino acid analysis identified a putative phosphorylation site at Thr(495) in the CaM-binding domain. Importantly, both cAK and cGK phosphorylation of NOS-III in vitro caused a highly reproducible partial (10-20%) NOS-III activation which was independent of Ca(2+)/CaM, and as much as a 4-fold increase in V(max) in the presence of Ca(2+)/CaM. cAK stimulation in intact endothelial cells also increased both Ca(2+/)CaM-independent and -dependent activation of NOS-III. These data collectively provide new evidence for cAK and cGK stimulation of both Ca(2+)/CaM-independent and -dependent NOS-III activity, and suggest possible cross-talk between the NO and prostaglandin I(2) pathways and a positive feedback mechanism for NO/cGMP signaling.


Subject(s)
Calcium/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic GMP-Dependent Protein Kinases/metabolism , Nitric Oxide Synthase/metabolism , Amino Acid Sequence , Animals , Calmodulin/metabolism , Enzyme Activation , Flavins/metabolism , Humans , Molecular Sequence Data , Nitric Oxide Synthase/chemistry , Nitric Oxide Synthase Type III , Phosphorylation , Protein Conformation , Sequence Homology, Amino Acid , Substrate Specificity
13.
Proc Natl Acad Sci U S A ; 96(11): 6084-9, 1999 May 25.
Article in English | MEDLINE | ID: mdl-10339545

ABSTRACT

Atrial natriuretic peptide (ANP) and nitric oxide (NO) are key regulators of ion and water transport in the kidney. Here, we report that these cGMP-elevating hormones stimulate Ca2+ reabsorption via a novel mechanism specifically involving type II cGMP-dependent protein kinase (cGK II). ANP and the NO donor, sodium nitroprusside (SNP), markedly increased Ca2+ uptake in freshly immunodissected rabbit connecting tubules (CNT) and cortical collecting ducts (CCD). Although readily increasing cGMP, ANP and SNP did not affect Ca2+ and Na+ reabsorption in primary cultures of these segments. Immunoblot analysis demonstrated that cGK II, and not cGK I, was present in freshly isolated CNT and CCD but underwent a complete down-regulation during the primary cell culture. However, upon adenoviral reexpression of cGK II in primary cultures, ANP, SNP, and 8-Br-cGMP readily increased Ca2+ reabsorption. In contrast, no cGMP-dependent effect on electrogenic Na+ transport was observed. The membrane localization of cGK II proved to be crucial for its action, because a nonmyristoylated cGK II mutant that was shown to be localized in the cytosol failed to mediate ANP-stimulated Ca2+ transport. The Ca2+-regulatory function of cGK II appeared isotype-specific because no cGMP-mediated increase in Ca2+ transport was observed after expression of the cytosolic cGK Ibeta or a membrane-bound cGK II/Ibeta chimer. These results demonstrate that ANP- and NO-stimulated Ca2+ reabsorption requires membrane-targeted cGK II.


Subject(s)
Atrial Natriuretic Factor/pharmacology , Calcium/metabolism , Cell Membrane/enzymology , Cyclic GMP-Dependent Protein Kinases/genetics , Cyclic GMP-Dependent Protein Kinases/metabolism , Kidney Cortex/metabolism , Kidney Tubules/metabolism , Animals , Biological Transport/drug effects , Cells, Cultured , Cyclic GMP-Dependent Protein Kinase Type II , Humans , Kidney Cortex/drug effects , Kidney Tubules/drug effects , Kidney Tubules, Collecting/drug effects , Kidney Tubules, Collecting/metabolism , Kinetics , Rabbits , Rats , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Deletion
14.
Brain Res Mol Brain Res ; 67(2): 258-66, 1999 Apr 20.
Article in English | MEDLINE | ID: mdl-10216224

ABSTRACT

There is contradictory information on the relevance of nitric oxide (NO) and cGMP for the function of brain capillary endothelial cells (BCEC) forming the blood-brain barrier (BBB). Therefore, NO/cGMP-mediated signal transduction was investigated in cell cultures of BCEC and of astrocytes (AC) inducing BBB properties in BCEC. Constitutive, Ca2+-activated isoforms of NO synthase (NOS) were found in BCEC (endothelial NOS: eNOS) and in AC (neuronal NOS: nNOS), leading to increased NO release after incubation with the Ca2+-ionophore A23187. Both cell types expressed inducible NOS (iNOS) after incubation with cytokines. Soluble guanylate cyclase (sGC) was detected in both cell types. NO-dependent cGMP formation were observed in BCEC and, less pronounced, in AC. Furthermore, both cell types formed cGMP independently of NO via stimulation of particulate guanylate cyclase (pGC). cGMP-dependent protein kinase (PKG) type Ibeta, but not type II, was expressed in BCEC and AC. In BCEC, vasodilator-stimulated phosphoprotein (VASP) was detected, an established substrate of PKG and associated with microfilaments and cell-cell contacts. Phosphorylation of VASP was intensified by increased intracellular cGMP concentrations. The results indicate that BCEC and, to a smaller degree, AC can form NO and cGMP in response to different stimuli. In BCEC, NO/cGMP-dependent phosphorylation of VASP is demonstrated, thus providing a possibility of influencing cell-cell contacts.


Subject(s)
Blood-Brain Barrier/physiology , Cell Adhesion Molecules/metabolism , Cyclic GMP/metabolism , Endothelium, Vascular/enzymology , Nitric Oxide/metabolism , Phosphoproteins/metabolism , Animals , Astrocytes/chemistry , Astrocytes/cytology , Astrocytes/enzymology , Blood Proteins/metabolism , Capillaries/chemistry , Capillaries/cytology , Capillaries/enzymology , Cell Adhesion Molecules/analysis , Cell Communication/physiology , Cells, Cultured , Cyclic GMP/analysis , Cyclic GMP-Dependent Protein Kinase Type I , Cyclic GMP-Dependent Protein Kinases/genetics , Cyclic GMP-Dependent Protein Kinases/metabolism , Endothelium, Vascular/chemistry , Endothelium, Vascular/cytology , Gene Expression Regulation, Enzymologic , Guanylate Cyclase/genetics , Guanylate Cyclase/metabolism , Microfilament Proteins , Nitrates/analysis , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Nitrites/analysis , Phosphoproteins/analysis , Phosphorylation , RNA, Messenger/analysis , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology
15.
Mol Pharmacol ; 54(6): 962-7, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9855623

ABSTRACT

Nitric oxide (NO), the physiological activator of soluble guanylyl cyclase (sGC), induces inhibitory effects on platelet activation via elevation of cGMP levels and stimulation of the cGMP-dependent protein kinase. YC-1, a benzylindazole derivative, was shown to activate sGC in intact platelets, resulting in inhibition of platelet aggregation. In a previous study, we demonstrated that YC-1 not only stimulates purified sGC but also potentiates the stimulatory action of submaximally effective NO and carbon monoxide (CO) concentrations. Here, we investigated the potentiating effect of YC-1 in intact platelets. YC-1 together with NO or CO led to complete inhibition of platelet aggregation at concentrations that were ineffective by themselves. Maximally effective 2, 2-diethyl-1-nitroso-oxyhydrazine (3 microM) and YC-1 (100 microM) concentrations each elevated the cGMP levels in intact platelets approximately 13-fold, and administration of the two drugs together resulted in enormous potentiation of cGMP formation, which greatly exceeded the effect on the purified enzyme and yielded a >1300-fold increase in cGMP levels. Similar results were obtained using CO instead of NO. Furthermore, YC-1 not only stimulated sGC but also inhibited cGMP-hydrolyzing phosphodiesterases in platelets. The enormous elevation of cGMP levels led to enhanced phosphorylation of the cGMP-dependent protein kinase substrate vasodilator-stimulated phosphoprotein. Thus, by the combination of two effects (i.e., potentiation of NO-induced sGC stimulation and phosphodiesterase inhibition), YC-1-like substances are potent activators of the sGC/cGMP pathways and are therefore interesting candidates to act as modulators of cGMP-mediated effects, especially within the cardiovascular system.


Subject(s)
Blood Platelets/drug effects , Carbon Monoxide/pharmacology , Cyclic GMP/biosynthesis , Indazoles/pharmacology , Nitric Oxide/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Vasodilator Agents/pharmacology , 1-Methyl-3-isobutylxanthine/pharmacology , Blood Platelets/metabolism , Cyclic AMP/biosynthesis , Drug Synergism , Humans , Phosphodiesterase Inhibitors/pharmacology , Phosphorylation
16.
Naunyn Schmiedebergs Arch Pharmacol ; 358(1): 134-9, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9721015

ABSTRACT

NO and cGMP have emerged as important signal transduction mediators of the effects of certain hormones, inter-/intracellular signals, toxins and drugs. However, a major challenge is to define relevant criteria for determining which of the many NO and/or cGMP effects are dependent on cGMP-dependent protein kinases (cGKs). Important criteria include that: (1) the cell types/tissues investigated contain at least one form of cGK which is activated by the cGMP-elevating agent in the intact cell system; (2) specific activators/inhibitors of cGKs mimic/block the effects of cGMP-elevating agents in the intact cell system; and (3) the cGMP effect is absent or blunted in cGK-deficient systems, or can be reconstituted by the introduction of active cGKs. Previously, analysis of cGK activity in intact cells has been very difficult. However, the analysis of vasodilator-stimulated phosphoprotein (VASP) phosphorylation by polyclonal antibodies and newly developed monoclonal antibodies, each of which specifically recognize different phosphorylation sites, allows the quantitative measurement of cGK activity in intact cells. With the use of these methods, the properties of certain cGK mutants, cGK activators (cGMP, 8-Br-cGMP, 8-pCPT-cGMP) as well as various "specific cGK inhibitors" (KT 5823, Rp-8Br-PET-cGMPS, Rp-8-pCPT-cGMPS, H8 and H89) were investigated. Although these "specific cGK inhibitors" have been widely used to establish or rule out functional roles of cGKs, very few studies have actually addressed the efficiency/specificity of such compounds in intact cells. Our results demonstrate that these inhibitors are useful tools only when used in combination with other experimental approaches and biochemical evidence.


Subject(s)
Cyclic GMP-Dependent Protein Kinases/physiology , Cyclic GMP/physiology , Isoenzymes/physiology , Nitric Oxide/physiology , Animals , Cyclic GMP-Dependent Protein Kinases/drug effects , Enzyme Activation/drug effects , Humans , Transfection
17.
J Biol Chem ; 273(32): 20029-35, 1998 Aug 07.
Article in English | MEDLINE | ID: mdl-9685341

ABSTRACT

The development and functional analysis of a monoclonal antibody (16C2) are reported; the antibody recognizes vasodilator-stimulated phosphoprotein (VASP; an established substrate of both cAMP- and cGMP-dependent protein kinase) only when serine 239 is phosphorylated. VASP serine 239 represents one of the best characterized cGMP-dependent protein kinase phosphorylation sites in vitro and in intact cells. Experiments with purified, recombinant human VASP and various VASP constructs with mutated phosphorylation sites (S157A, S239A, T278A) and experiments with intact cells (human/rat platelets and other cells) treated with cyclic nucleotide-elevating agents demonstrated the specificity of the monoclonal antibody 16C2. Quantitative analysis of the VASP shift from 46 to 50 kDa (indicating VASP serine 157 phosphorylation) and the appearance of VASP detected by the 16C2 monoclonal antibody (VASP serine 239 phosphorylation) in human platelets stimulated by selective protein kinase activators confirmed that serine 239 is the VASP phosphorylation site preferred by cGMP-dependent protein kinase in intact cells. Immunofluorescence experiments with human platelets treated with cGMP analogs showed that the 16C2 monoclonal antibody also detects VASP serine 239 phosphorylation in situ at established intracellular localization sites. Analysis of VASP serine 239 phosphorylation by the 16C2 antibody appears to be the best method presently available to measure cGMP-dependent protein kinase activation in intact cells. Also, the 16C2 antibody promises to be an excellent tool for the evaluation of VASP function in intact cells.


Subject(s)
Antibodies, Monoclonal/immunology , Cell Adhesion Molecules/physiology , Phosphoproteins/immunology , Phosphoproteins/physiology , Animals , Binding Sites/physiology , Blood Platelets/drug effects , Blood Platelets/metabolism , Cell Line , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic GMP/analogs & derivatives , Cyclic GMP/pharmacology , Cyclic GMP-Dependent Protein Kinases/physiology , Dichlororibofuranosylbenzimidazole/analogs & derivatives , Dichlororibofuranosylbenzimidazole/pharmacology , Epoprostenol/pharmacology , Fluorescent Antibody Technique , Humans , Microfilament Proteins , Nitroprusside/pharmacology , Phosphorylation , Rats , Recombinant Proteins/metabolism , Serine/metabolism , Thionucleotides/pharmacology , Transfection
18.
Proc Natl Acad Sci U S A ; 95(15): 9003-8, 1998 Jul 21.
Article in English | MEDLINE | ID: mdl-9671794

ABSTRACT

An overactive renin-angiotensin-aldosterone system (RAAS) has a central role in the pathogenesis of hypertension and cardiac hypertrophy, precursors of cardiac failure. Natriuretic peptides and NO acting through their second messenger, cGMP, increase natriuresis and diuresis, and inhibit renin release; however the mechanism by which this inhibition of the RAAS system functions is obscure. We recently reported cloning of the cDNA for type II cGMP-dependent protein kinase (cGK II), elucidated its first known function of inhibiting the cystic fibrosis transmembrane conductance regulator in rat intestine, and initially described its location in rat kidney juxtaglomerular (JG) cells, the ascending thin limb, and the brush border of proximal tubules. Here, we demonstrate inhibition of isoproterenol- or forskolin-stimulated renin release by 8-para-chlorophenylthio-cGMP (8-pCPT-cGMP), a selective activator of cGK, and prevention of this inhibition by a selective inhibitor of cGK, Rp-8-pCPT-cGMPS. In systems of differing complexity, inhibition by 8-pCPT-cGMP was nearly complete in isolated perfused kidney and microdissected afferent arterioles but only approximately 25% in isolated JG cells. Expression of either cGK II or cGK I in JG cells by using adenoviral vectors enhanced the inhibition of forskolin-stimulated renin release by 8-pCPT-cGMP to 50%. Our results indicate that cGK II, and possibly cGK I, can mediate cGMP inhibitory effects on renin release and are physiological components of the cGMP signal transduction system which opposes the RAAS.


Subject(s)
Cyclic AMP/metabolism , Cyclic GMP-Dependent Protein Kinases/metabolism , Juxtaglomerular Apparatus/metabolism , Kidney Glomerulus/metabolism , Kidney/metabolism , Renin/metabolism , Animals , Cells, Cultured , Colforsin/antagonists & inhibitors , Colforsin/pharmacology , Cyclic GMP-Dependent Protein Kinases/genetics , In Vitro Techniques , Isoproterenol/antagonists & inhibitors , Isoproterenol/pharmacology , Juxtaglomerular Apparatus/cytology , Juxtaglomerular Apparatus/ultrastructure , Kidney/drug effects , Kidney/ultrastructure , Kidney Glomerulus/ultrastructure , Mice , Microscopy, Immunoelectron , Rats , Rats, Sprague-Dawley
19.
Proc Natl Acad Sci U S A ; 95(4): 1466-71, 1998 Feb 17.
Article in English | MEDLINE | ID: mdl-9465038

ABSTRACT

A recently cloned isoform of cGMP-dependent protein kinase (cGK), designated type II, was implicated as the mediator of cGMP-provoked intestinal Cl- secretion based on its localization in the apical membrane of enterocytes and on its capacity to activate cystic fibrosis transmembrane conductance regulator (CFTR) Cl- channels. In contrast, the soluble type I cGK was unable to activate CFTR in intact cells, although both cGK I and cGK II could phosphorylate CFTR in vitro. To investigate the molecular basis for the cGK II isotype specificity of CFTR channel gating, we expressed cGK II or cGK I mutants possessing different membrane binding properties by using adenoviral vectors in a CFTR-transfected intestinal cell line, and we examined the ability of cGMP to phosphorylate and activate the Cl- channel. Mutation of the cGK II N-terminal myristoylation site (Gly2 --> Ala) reduced cGK II membrane binding and severely impaired cGK II activation of CFTR. Conversely, a chimeric protein, in which the N-terminal membrane-anchoring domain of cGK II was fused to the N terminus of cGK Ibeta, acquired the ability to associate with the membrane and activate the CFTR Cl- channel. The potency order of cGK constructs for activation of CFTR (cGK II > membrane-bound cGK I chimer >> nonmyristoylated cGK II > cGK Ibeta) correlated with the extent of 32P incorporation into CFTR observed in parallel measurements. These results strongly support the concept that membrane targeting of cGK is a major determinant of CFTR Cl- channel activation in intact cells.


Subject(s)
Chloride Channels/physiology , Cyclic GMP-Dependent Protein Kinases/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Animals , Cell Compartmentation , Cell Membrane/enzymology , Humans , Ion Channel Gating , Myristates , Phosphorylation , Protein Processing, Post-Translational , Rats , Recombinant Proteins , Transfection
20.
Trends Biochem Sci ; 22(8): 307-12, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9270304

ABSTRACT

cGMP-dependent protein kinases I and II conduct signals from widespread signaling systems. Whereas the type I kinase mediates numerous effects of natriuretic peptides and nitric oxide in cardiovascular cells, the type II kinase transduces signals from the Escherichia coli heat-stable enterotoxin, STa, and from the endogenous intestinal peptide, guanylin, stimulating Cl- conductance of the cystic fibrosis transmembrane conductance regulator (CFTR). Although the two kinases may be interchangeable for several functions, CFTR regulation specifically requires the type II kinase.


Subject(s)
Cyclic GMP-Dependent Protein Kinases/physiology , Signal Transduction , Amino Acid Sequence , Animals , Cyclic GMP/metabolism , Cyclic GMP-Dependent Protein Kinases/chemistry , Cyclic GMP-Dependent Protein Kinases/genetics , Gene Expression Regulation, Enzymologic , Humans , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL