Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
PLoS One ; 11(10): e0164114, 2016.
Article in English | MEDLINE | ID: mdl-27711243

ABSTRACT

While the mammalian brain functions within a very narrow range of oxygen concentrations and temperatures, the fruit fly, Drosophila melanogaster, has employed strategies to deal with a much wider range of acute environmental stressors. The foraging (for) gene encodes the cGMP-dependent protein kinase (PKG), has been shown to regulate thermotolerance in many stress-adapted species, including Drosophila, and could be a potential therapeutic target in the treatment of hyperthermia in mammals. Whereas previous thermotolerance studies have looked at the effects of PKG variation on Drosophila behavior or excitatory postsynaptic potentials at the neuromuscular junction (NMJ), little is known about PKG effects on presynaptic mechanisms. In this study, we characterize presynaptic calcium ([Ca2+]i) dynamics at the Drosophila larval NMJ to determine the effects of high temperature stress on synaptic transmission. We investigated the neuroprotective role of PKG modulation both genetically using RNA interference (RNAi), and pharmacologically, to determine if and how PKG affects presynaptic [Ca2+]i dynamics during hyperthermia. We found that PKG activity modulates presynaptic neuronal Ca2+ responses during acute hyperthermia, where PKG activation makes neurons more sensitive to temperature-induced failure of Ca2+ flux and PKG inhibition confers thermotolerance and maintains normal Ca2+ dynamics under the same conditions. Targeted motoneuronal knockdown of PKG using RNAi demonstrated that decreased PKG expression was sufficient to confer thermoprotection. These results demonstrate that the PKG pathway regulates presynaptic motoneuronal Ca2+ signaling to influence thermotolerance of presynaptic function during acute hyperthermia.


Subject(s)
Calcium/metabolism , Cyclic GMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic GMP-Dependent Protein Kinases/genetics , Drosophila melanogaster/physiology , Motor Neurons/drug effects , Temperature , Thermotolerance , Animals , Cyclic GMP-Dependent Protein Kinases/deficiency , Drosophila Proteins/deficiency , Drosophila Proteins/genetics , Drosophila melanogaster/drug effects , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Larva/drug effects , Larva/genetics , Larva/metabolism , Larva/physiology , Motor Neurons/cytology , Motor Neurons/metabolism , Presynaptic Terminals/drug effects , Presynaptic Terminals/metabolism , Protein Kinase Inhibitors/pharmacology , RNA Interference , Synaptic Transmission/drug effects , Synaptic Transmission/genetics , Thermotolerance/drug effects , Thermotolerance/genetics
2.
Am J Pathol ; 180(6): 2268-75, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22632818

ABSTRACT

Protein kinase G (PKG) plays an important role in the regulation of vascular smooth cell contractility and is a critical mediator of nitric oxide signaling, which regulates cardiovascular homeostasis. PKG-I-knockout (Prkg1(-/-)) mice exhibit impaired nitric oxide/cGMP-dependent vasorelaxation and systemic hypertension. However, it remains unknown whether PKG-I deficiency induces pulmonary hypertension. In this study, we characterized the hypertensive pulmonary phenotypes in Prkg1(-/-) mice and delineated the underlying molecular basis. We observed a significant increase in right ventricular systolic pressure in Prkg1(-/-) mice in the absence of systemic hypertension and left-sided heart dysfunction. In addition, we observed marked muscularization of distal pulmonary vessels in Prkg1(-/-) mice. Microangiography revealed impaired integrity of the pulmonary vasculature in Prkg1(-/-) mice. Mechanistically, PKG-I-mediated phosphorylation of Rho A Ser188 was markedly decreased, and the resultant Rho A activation was significantly increased in Prkg1(-/-) lung tissues, which resulted in Rho kinase activation. The i.t. administration of fasudil, a Rho kinase inhibitor, reversed the hypertensive pulmonary phenotype in Prkg1(-/-) mice. Taken together, these data show that PKG-I deficiency induces pulmonary hypertension through Rho A/Rho kinase activation-mediated vasoconstriction and pulmonary vascular remodeling.


Subject(s)
Cyclic GMP-Dependent Protein Kinases/physiology , Hypertension, Pulmonary/enzymology , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/therapeutic use , Animals , Antihypertensive Agents/therapeutic use , Cells, Cultured , Cyclic GMP-Dependent Protein Kinase Type I , Cyclic GMP-Dependent Protein Kinases/deficiency , Enzyme Activation/physiology , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/physiopathology , Mice , Mice, Knockout , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/physiopathology , Protein Kinase Inhibitors/therapeutic use , Pulmonary Artery/pathology , Signal Transduction/physiology , Vasoconstriction/physiology , rho-Associated Kinases/antagonists & inhibitors
3.
Diabetes ; 60(1): 148-56, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20978093

ABSTRACT

OBJECTIVE: The physiologic significance of the nitric oxide (NO)/cGMP signaling pathway in islets is unclear. We hypothesized that cGMP-dependent protein kinase type I (cGKI) is directly involved in the secretion of islet hormones and glucose homeostasis. RESEARCH DESIGN AND METHODS: Gene-targeted mice that lack cGKI in islets (conventional cGKI mutants and cGKIα and Iß rescue mice [α/ßRM] that express cGKI only in smooth muscle) were studied in comparison to control (CTR) mice. cGKI expression was mapped in the endocrine pancreas by Western blot, immuno-histochemistry, and islet-specific recombination analysis. Insulin, glucagon secretion, and cytosolic Ca²(+) ([Ca²(+)](i)) were assayed by radioimmunoassay and FURA-2 measurements, respectively. Serum levels of islet hormones were analyzed at fasting and upon glucose challenge (2 g/kg) in vivo. RESULTS: Immunohistochemistry showed that cGKI is present in α- but not in ß-cells in islets of Langerhans. Mice that lack α-cell cGKI had significantly elevated fasting glucose and glucagon levels, whereas serum insulin levels were unchanged. High glucose concentrations strongly suppressed the glucagon release in CTR mice, but had only a moderate effect on islets that lacked cGKI. 8-Br-cGMP reduced stimulated [Ca²(+)](i) levels and glucagon release rates of CTR islets at 0.5 mmol/l glucose, but was without effect on [Ca²(+)](i) or hormone release in cGKI-deficient islets. CONCLUSIONS: We propose that cGKI modulates glucagon release by suppression of [Ca²(+)](i) in α-cells.


Subject(s)
Glucagon-Secreting Cells/metabolism , Glucagon/metabolism , Guanylate Kinases/metabolism , Animals , Blood Glucose/metabolism , Calcium/physiology , Cyclic GMP-Dependent Protein Kinase Type I , Cyclic GMP-Dependent Protein Kinases/deficiency , Cyclic GMP-Dependent Protein Kinases/genetics , DNA Primers , Gene Amplification , Genes, Reporter , Glucagon/blood , Glucagon-Secreting Cells/enzymology , Glucose Tolerance Test , Homeostasis , Hypoxanthine Phosphoribosyltransferase/genetics , Insulin/blood , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/cytology , Luminescent Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/physiology , Muscle, Smooth/enzymology , RNA/genetics , RNA/isolation & purification , Recombination, Genetic , Reverse Transcriptase Polymerase Chain Reaction
4.
Am J Physiol Lung Cell Mol Physiol ; 299(3): L323-33, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20453163

ABSTRACT

Increasing evidence suggests that endothelial cytotoxicity from reactive oxygen species (ROS) contributes to the pathogenesis of acute lung injury. Treatments designed to increase intracellular cGMP attenuate ROS-mediated apoptosis and necrosis in several cell types, but the mechanisms are not understood, and the effect of cGMP on pulmonary endothelial cell death remains controversial. In the current study, increasing intracellular cGMP by either 8pCPT-cGMP (50 microM) or atrial natriuretic peptide (10 nM) significantly attenuated cell death in H(2)O(2)-challenged mouse lung microvascular (MLMVEC) monolayers. 8pCPT-cGMP also decreased perfusate LDH release in isolated mouse lungs exposed to H(2)O(2) or ischemia-reperfusion. The protective effect of increasing cGMP in MLMVECs was accompanied by enhanced endothelial H(2)O(2) scavenging (measured by H(2)O(2) electrode) and decreased intracellular ROS concentration (measured by 2',7'-dichlorofluorescin fluorescence) as well as decreased phosphorylation of p38 MAPK and Akt. The cGMP-mediated cytoprotection and increased H(2)O(2) scavenging required >2 h of 8pCPT-cGMP incubation in wild-type MLMVEC and were absent in MLMVEC from protein kinase G (PKG(I))-/- mice suggesting a PKG(I)-mediated effect on gene regulation. Catalase and glutathione peroxidase 1 (Gpx-1) protein were increased by cGMP in wild-type but not PKG(I)-/- MLMVEC monolayers. Both the cGMP-mediated increases in antioxidant proteins and H(2)O(2) scavenging were prevented by inhibition of translation with cycloheximide. 8pCPT-cGMP had minimal effects on catalase and Gpx-1 mRNA. We conclude that cGMP, through PKG(I), attenuated H(2)O(2)-induced cytotoxicity in MLMVEC by increasing catalase and Gpx-1 expression through an unknown posttranscriptional effect.


Subject(s)
Antioxidants/metabolism , Cyclic GMP-Dependent Protein Kinases/metabolism , Cyclic GMP/metabolism , Endothelial Cells/metabolism , Hydrogen Peroxide/pharmacology , Lung/blood supply , Microvessels/physiopathology , Oxidants/pharmacology , Animals , Atrial Natriuretic Factor/pharmacology , Catalase/metabolism , Cell Death , Cells, Cultured , Cyclic GMP/analogs & derivatives , Cyclic GMP/pharmacology , Cyclic GMP-Dependent Protein Kinase Type I , Cyclic GMP-Dependent Protein Kinases/deficiency , Endothelial Cells/drug effects , Glutathione Peroxidase/metabolism , Intracellular Membranes/metabolism , Isoenzymes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidoreductases/metabolism , Protein Processing, Post-Translational , Reactive Oxygen Species/metabolism , Signal Transduction , Thionucleotides/pharmacology
6.
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
7.
Sci Signal ; 2(99): ra78, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19952371

ABSTRACT

Brown adipose tissue (BAT) is a primary site of energy expenditure through thermogenesis, which is mediated by the uncoupling protein-1 (UCP-1) in mitochondria. Here, we show that protein kinase G (PKG) is essential for brown fat cell differentiation. Induction of adipogenic markers and fat storage was impaired in the absence of PKGI. Furthermore, PKGI mediated the ability of nitric oxide (NO) and guanosine 3',5'-monophosphate (cGMP) to induce mitochondrial biogenesis and increase the abundance of UCP-1. Mechanistically, we found that PKGI controlled insulin signaling in BAT by inhibiting the activity of RhoA and Rho-associated kinase (ROCK), thereby relieving the inhibitory effects of ROCK on insulin receptor substrate-1 and activating the downstream phosphoinositide 3-kinase-Akt cascade. Thus, PKGI links NO and cGMP signaling with the RhoA-ROCK and the insulin pathways, thereby controlling induction of adipogenic and thermogenic programs during brown fat cell differentiation.


Subject(s)
Adipose Tissue, Brown/cytology , Cell Differentiation , Cyclic GMP-Dependent Protein Kinases/physiology , Mitochondria/metabolism , Adipogenesis , Animals , Cyclic GMP , Cyclic GMP-Dependent Protein Kinases/deficiency , Insulin/metabolism , Ion Channels , Mice , Mice, Knockout , Mitochondrial Proteins , Nitric Oxide , Uncoupling Protein 1 , rho-Associated Kinases , rhoA GTP-Binding Protein
8.
J Neurosci ; 29(48): 15155-60, 2009 Dec 02.
Article in English | MEDLINE | ID: mdl-19955367

ABSTRACT

The cGMP-dependent protein kinase type I (cGKI) has multiple functions including a role in axonal growth and pathfinding of sensory neurons, and counteracts Semaphorin 3A (Sema3A)-induced growth cone collapse. Within the nervous system, however, the transcriptional regulation of cGKI is still obscure. Recently, the transcription factor and tumor suppressor p53 has been reported to promote neurite outgrowth by regulating the gene expression of factors that promote growth cone extension, but specific p53 targets genes that may counteract growth cone collapse have not been identified so far. Here, we show that p53 promotes cGKI expression in neuronal-like PC-12 cells and primary neurons by occupying specific regulatory elements in a chromatin environment during neuronal maturation. Importantly, we demonstrate that p53-dependent expression of cGKI is required for the ability of cGMP to counteract growth cone collapse. Growth cone retraction mediated by Sema3A is overcome by cGMP only in wild-type, but not in p53-null dorsal root ganglia. Reconstitution of p53 levels is sufficient to recover both cGKI expression and the ability of cGMP to counteract growth cone collapse, while cGKI overexpression rescues growth cone collapse in p53-null primary neurons. In conclusion, this study identifies p53 as a transcription factor that regulates the expression of cGKI during neuronal maturation and cGMP-dependent inhibition of growth cone collapse.


Subject(s)
Cyclic GMP-Dependent Protein Kinases/metabolism , Gene Expression Regulation, Developmental/physiology , Growth Cones/physiology , Neurons/physiology , Tumor Suppressor Protein p53/physiology , Animals , Animals, Newborn , Cells, Cultured , Cerebral Cortex/cytology , Chlorocebus aethiops , Chromatin Immunoprecipitation/methods , Cyclic GMP/analogs & derivatives , Cyclic GMP/pharmacology , Cyclic GMP-Dependent Protein Kinase Type I , Cyclic GMP-Dependent Protein Kinases/deficiency , Embryo, Mammalian , Ganglia, Spinal/cytology , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/genetics , Green Fluorescent Proteins/genetics , Growth Cones/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics , Neurons/cytology , Neurons/drug effects , Organ Culture Techniques , RNA, Messenger/metabolism , Rats , Semaphorin-3A/genetics , Transfection/methods , Tumor Suppressor Protein p53/deficiency
9.
Mol Cell Biol ; 29(22): 6018-32, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19737918

ABSTRACT

Vascular fibrosis is a major complication of hypertension and atherosclerosis, yet it is largely untreatable. Natriuretic peptides (NPs) repress fibrogenic activation of vascular smooth muscle cells (VSMCs), but the intracellular mechanism mediating this effect remains undetermined. Here we show that inhibition of RhoA through phosphorylation at Ser188, the site targeted by the NP effector cyclic GMP (cGMP)-dependent protein kinase I (cGK I), is critical to fully exert antifibrotic potential. cGK I(+/-) mouse blood vessels exhibited an attenuated P-RhoA level and concurrently increased RhoA/ROCK signaling. Importantly, cGK I insufficiency caused dynamic recruitment of ROCK into the fibrogenic programs, thereby eliciting exaggerated vascular hypertrophy and fibrosis. Transgenic expression of cGK I-unphosphorylatable RhoA(A188) in VSMCs augmented ROCK activity, vascular hypertrophy, and fibrosis more prominently than did that of wild-type RhoA, consistent with the notion that RhoA(A188) escapes the intrinsic inhibition by cGK I. Additionally, VSMCs expressing RhoA(A188) became refractory to the antifibrotic effects of NPs. Our results identify cGK I-mediated Ser188 phosphorylation of RhoA as a converging node for pro- and antifibrotic signals and may explain how diminished cGMP signaling, commonly associated with vascular malfunction, predisposes individuals to vascular fibrosis.


Subject(s)
Blood Vessels/enzymology , Blood Vessels/pathology , Cyclic GMP-Dependent Protein Kinases/metabolism , Phosphoserine/metabolism , Signal Transduction , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Angiotensin II/pharmacology , Animals , Blood Vessels/drug effects , Cyclic GMP/pharmacology , Cyclic GMP-Dependent Protein Kinases/deficiency , Enzyme Activation/drug effects , Fibrosis , Gene Expression Regulation/drug effects , Humans , Hypertrophy , Mice , Mice, Transgenic , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/pathology , Mutant Proteins/metabolism , Organ Specificity/drug effects , Phosphorylation/drug effects , Protein Biosynthesis/drug effects , Serum Response Element/genetics , Signal Transduction/drug effects , Transcription, Genetic/drug effects
10.
Am J Physiol Gastrointest Liver Physiol ; 297(4): G834-9, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19628652

ABSTRACT

cGMP-dependent protein kinase I (cGKI) induces relaxation of smooth muscle via several pathways that include inhibition of intracellular Ca(2+) signaling and/or involve activation of myosin phosphatase. In the present study, we investigated these mechanisms comparatively in colon and jejunum longitudinal smooth muscle from mice. In simultaneous recordings from colon muscle, 8-bromo-cGMP (8-Br-cGMP) reduced both carbachol-induced tension and carbachol-induced increase in intracellular Ca(2+) concentration ([Ca(2+)](i)). These effects of 8-Br-cGMP were absent in colon from mice carrying a mutated inositol-1,4,5 trisphosphate receptor I-associated G kinase substrate (IRAG) gene or lacking cGKI. However, in jejunum, 8-Br-cGMP reduced carbachol-induced tension but did not change corresponding [Ca(2+)](i) signals. This setting was also observed in jejunum from mice carrying a mutated IRAG gene, whereas no response to 8-Br-cGMP was observed in jejunum from mice lacking cGKI. After inhibition of phosphatase activity by calyculin A, 8-Br-cGMP did not relax jejunum but still relaxed colon muscle. In Western blot analysis, 8-Br-cGMP reduced the signal for phosphorylated MYPT-1 in carbachol-stimulated jejunum but not in colon. These results suggest that cGMP/cGKI signaling differentially inhibits contraction in the muscles investigated: in jejunum, inhibition is performed without changing [Ca(2+)](i) and is dependent on phosphatase activity, whereas in colon, inhibition is mediated by inhibition of [Ca(2+)](i) signals.


Subject(s)
Calcium Signaling , Colon/enzymology , Cyclic GMP-Dependent Protein Kinases/metabolism , Cyclic GMP/metabolism , Jejunum/enzymology , Muscle Relaxation , Muscle, Smooth/enzymology , Amides/pharmacology , Animals , Calcium Signaling/drug effects , Carbachol/pharmacology , Cholinergic Agonists/pharmacology , Colon/drug effects , Cyclic GMP/analogs & derivatives , Cyclic GMP/pharmacology , Cyclic GMP-Dependent Protein Kinases/deficiency , Cyclic GMP-Dependent Protein Kinases/genetics , Enzyme Inhibitors/pharmacology , In Vitro Techniques , Jejunum/drug effects , Marine Toxins , Membrane Proteins , Mice , Mice, Knockout , Muscle Relaxation/drug effects , Muscle, Smooth/drug effects , Mutation , Myosin-Light-Chain Kinase/metabolism , Myosin-Light-Chain Phosphatase , Oxazoles/pharmacology , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphoprotein Phosphatases/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation , Pyridines/pharmacology , Time Factors , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism
11.
J Neurosci ; 28(52): 14202-12, 2008 Dec 24.
Article in English | MEDLINE | ID: mdl-19109502

ABSTRACT

Long-term potentiation (LTP) of inputs relaying sensory information from cortical and thalamic neurons to principal neurons in the lateral amygdala (LA) is thought to serve as a cellular mechanism for associative fear learning. Nitric oxide (NO), a messenger molecule widely implicated in synaptic plasticity and behavior, has been shown to enhance LTP in the LA as well as consolidation of associative fear memory. Additional evidence suggests that NO-induced enhancement of LTP and amygdala-dependent learning requires signaling through soluble guanylyl cyclase (sGC) and cGMP-dependent protein kinase (cGK). Mammals possess two genes for cGK: the prkg1 gene gives rise to the cGK type I isoforms, cGKIalpha and cGKIbeta, and the prkg2 gene encodes the cGK type II. Reportedly, both cGKI and cGKII are expressed in the amygdala, and cGKII is involved in controlling anxiety-like behavior. Because selective pharmacological tools for individual cGK isoforms are lacking, we used different knock-out mouse models to examine the function of cGKI and cGKII for LTP in the LA and pavlovian fear conditioning. We found robust expression of the cGKI specifically in the LA with cGKIbeta as the prevailing isoform. We further show a marked reduction of LTP at both thalamic and cortical inputs to the LA and a selective impairment of auditory-cued fear memory in cGKI-deficient mutants. In contrast, cGKII null mutants lack these phenotypes. Our data suggest a function of cGKI, likely the beta isoform, in the LA, supporting synaptic plasticity and consolidation of fear memory.


Subject(s)
Amygdala/physiology , Cues , Cyclic GMP-Dependent Protein Kinases/metabolism , Fear , Long-Term Potentiation/physiology , Memory/physiology , Acoustic Stimulation/adverse effects , Amygdala/metabolism , Analysis of Variance , Animals , Conditioning, Psychological/physiology , Cyclic GMP-Dependent Protein Kinase Type I , Cyclic GMP-Dependent Protein Kinase Type II , Cyclic GMP-Dependent Protein Kinases/deficiency , Cyclic GMP-Dependent Protein Kinases/genetics , Electric Stimulation/methods , Exploratory Behavior/physiology , In Vitro Techniques , Long-Term Potentiation/drug effects , Long-Term Potentiation/genetics , Male , Maze Learning/physiology , Mice , Mice, Transgenic , Motor Activity/genetics , Neural Pathways/physiology , Pain Threshold/physiology , Patch-Clamp Techniques , Signal Transduction , Synaptic Transmission/genetics , Time Factors
12.
Proc Natl Acad Sci U S A ; 105(18): 6771-6, 2008 May 06.
Article in English | MEDLINE | ID: mdl-18443297

ABSTRACT

To explore the functional significance of cGMP-dependent protein kinase type I (cGKI) in the regulation of erythrocyte survival, gene-targeted mice lacking cGKI were compared with their control littermates. By the age of 10 weeks, cGKI-deficient mice exhibited pronounced anemia and splenomegaly. Compared with control mice, the cGKI mutants had significantly lower red blood cell count, packed cell volume, and hemoglobin concentration. Anemia was associated with a higher reticulocyte number and an increase of plasma erythropoietin concentration. The spleens of cGKI mutant mice were massively enlarged and contained a higher fraction of Ter119(+) erythroid cells, whereas the relative proportion of leukocyte subpopulations was not changed. The Ter119(+) cGKI-deficient splenocytes showed a marked increase in annexin V binding, pointing to phosphatidylserine (PS) exposure at the outer membrane leaflet, a hallmark of suicidal erythrocyte death or eryptosis. Compared with control erythrocytes, cGKI-deficient erythrocytes exhibited in vitro a higher cytosolic Ca(2+) concentration, a known trigger of eryptosis, and showed increased PS exposure, which was paralleled by a faster clearance in vivo. Together, these results identify a role of cGKI as mediator of erythrocyte survival and extend the emerging concept that cGMP/cGKI signaling has an antiapoptotic/prosurvival function in a number of cell types in vivo.


Subject(s)
Anemia/complications , Anemia/enzymology , Cyclic GMP-Dependent Protein Kinases/deficiency , Splenomegaly/complications , Splenomegaly/enzymology , Animals , Calcium/metabolism , Cell Size , Cyclic GMP-Dependent Protein Kinase Type I , Erythrocytes/enzymology , Erythrocytes/pathology , Fluoresceins/metabolism , Intracellular Space/metabolism , Mice , Succinimides/metabolism
13.
Arterioscler Thromb Vasc Biol ; 28(7): 1244-50, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18420996

ABSTRACT

BACKGROUND: Nitric oxide (NO) is of crucial importance for smooth muscle cell (SMC) function and exerts numerous, and sometimes opposing, effects on vascular restenosis. Although cGMP-dependent protein kinase type I (cGKI) is a principal effector of NO, the molecular pathway of vascular NO signaling in restenosis is unclear. The purpose of this study was to examine the functional role of the smooth muscle cGMP/cGKI signaling cascade in restenosis of vessels. METHODS AND RESULTS: Tissue-specific mouse mutants were generated in which the cGKI protein was ablated in SMCs. We investigated whether the absence of cGKI in SMCs would affect vascular remodeling after carotid ligation or removal of the endothelium. No differences were detected between the tissue-specific cGKI mutants and control mice at different time points after vascular injury on a normolipidemic or apoE-deficient background. In line with these results, chronic drug treatment of injured control mice with the phosphodiesterase-5 inhibitor sildenafil elevated cGMP levels but had no influence on the ligation-induced remodeling. CONCLUSIONS: The genetic and pharmacological manipulation of the cGMP/cGKI signaling indicates that this pathway is not involved in the protective effects of NO, suggesting that NO affects vascular remodeling during restenosis via alternative mechanisms.


Subject(s)
Carotid Artery Injuries/metabolism , Carotid Stenosis/metabolism , Cyclic GMP-Dependent Protein Kinases/metabolism , Cyclic GMP/metabolism , Muscle, Smooth, Vascular/metabolism , Nitric Oxide/metabolism , Signal Transduction , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Carotid Artery Injuries/enzymology , Carotid Artery Injuries/pathology , Carotid Artery, Common/metabolism , Carotid Artery, Common/surgery , Carotid Stenosis/enzymology , Carotid Stenosis/pathology , Cyclic GMP-Dependent Protein Kinase Type I , Cyclic GMP-Dependent Protein Kinases/deficiency , Cyclic GMP-Dependent Protein Kinases/genetics , Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Disease Models, Animal , Ligation , Mice , Mice, Knockout , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/pathology , Phosphodiesterase 5 Inhibitors , Phosphodiesterase Inhibitors/pharmacology , Piperazines/pharmacology , Purines/pharmacology , Recurrence , Signal Transduction/drug effects , Sildenafil Citrate , Sulfones/pharmacology , Time Factors
14.
Learn Mem ; 15(3): 133-42, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18310460

ABSTRACT

The role of the foraging (for) gene, which encodes a cyclic guanosine-3',5'-monophosphate (cGMP)-dependent protein kinase (PKG), in food-search behavior in Drosophila has been intensively studied. However, its functions in other complex behaviors have not been well-characterized. Here, we show experimentally in Drosophila that the for gene is required in the operant visual learning paradigm. Visual pattern memory was normal in a natural variant rover (for(R)) but was impaired in another natural variant sitter (for(S)), which has a lower PKG level. Memory defects in for(S) flies could be rescued by either constitutive or adult-limited expression of for in the fan-shaped body. Interestingly, we showed that such rescue also occurred when for was expressed in the ellipsoid body. Additionally, expression of for in the fifth layer of the fan-shaped body restored sufficient memory for the pattern parameter "elevation" but not for "contour orientation," whereas expression of for in the ellipsoid body restored sufficient memory for both parameters. Our study defines a Drosophila model for further understanding the role of cGMP-PKG signaling in associative learning/memory and the neural circuit underlying this for-dependent visual pattern memory.


Subject(s)
Conditioning, Operant/physiology , Cyclic GMP-Dependent Protein Kinases/physiology , Drosophila Proteins/physiology , Drosophila melanogaster/enzymology , Memory, Short-Term/physiology , Nerve Tissue Proteins/physiology , Pattern Recognition, Visual/physiology , Alleles , Animals , Animals, Genetically Modified , Brain/enzymology , Brain/ultrastructure , Cyclic GMP-Dependent Protein Kinases/deficiency , Cyclic GMP-Dependent Protein Kinases/genetics , Drosophila Proteins/deficiency , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/physiology , Gene Library , Isoenzymes/genetics , Isoenzymes/physiology , Mutagenesis, Insertional , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Recombinant Fusion Proteins/physiology
15.
J Cell Biol ; 179(2): 331-40, 2007 Oct 22.
Article in English | MEDLINE | ID: mdl-17954614

ABSTRACT

Sensory axonal projections into the spinal cord display a highly stereotyped pattern of T- or Y-shaped axon bifurcation at the dorsal root entry zone (DREZ). Here, we provide evidence that embryonic mice with an inactive receptor guanylyl cyclase Npr2 or deficient for cyclic guanosine monophosphate-dependent protein kinase I (cGKI) lack the bifurcation of sensory axons at the DREZ, i.e., the ingrowing axon either turns rostrally or caudally. This bifurcation error is maintained to mature stages. In contrast, interstitial branching of collaterals from primary stem axons remains unaffected, indicating that bifurcation and interstitial branching are processes regulated by a distinct molecular mechanism. At a functional level, the distorted axonal branching at the DREZ is accompanied by reduced synaptic input, as revealed by patch clamp recordings of neurons in the superficial layers of the spinal cord. Hence, our data demonstrate that Npr2 and cGKI are essential constituents of the signaling pathway underlying axonal bifurcation at the DREZ and neuronal connectivity in the dorsal spinal cord.


Subject(s)
Axons/enzymology , Guanylate Cyclase/metabolism , Receptors, Atrial Natriuretic Factor/metabolism , Spinal Cord/enzymology , Animals , Cyclic GMP-Dependent Protein Kinases/deficiency , Cyclic GMP-Dependent Protein Kinases/metabolism , Electrophysiology , Enzyme Activation , Ganglia, Spinal/cytology , Ganglia, Spinal/embryology , Ganglia, Spinal/enzymology , Mice , Mice, Mutant Strains , Models, Biological , Mutation/genetics , Nociceptors/metabolism , Proprioception , Spinal Cord/cytology , Spinal Nerve Roots/cytology , Spinal Nerve Roots/enzymology
16.
Blood ; 107(3): 965-72, 2006 Feb 01.
Article in English | MEDLINE | ID: mdl-16210341

ABSTRACT

Integrin activation (inside-out signaling) in platelets can be initiated by agonists such as von Willebrand factor (VWF) and thrombin. Here we show that a mitogen-activated protein kinase (MAPK), p38, plays an important role in the activation of integrin alphaIIb beta3 induced by VWF and thrombin. A dominant-negative mutant of p38, p38AF, inhibits alphaIIb beta3 activation induced by VWF binding to its receptor, the platelet glycoprotein Ib-IX (GPIb-IX), and p38 inhibitors diminish platelet aggregation induced by VWF or low-dose thrombin. The inhibitory effect of p38 inhibitor is unlikely to be caused by the previous suggested effect on cyclo-oxygenase, as inhibition also was observed in the presence of high concentrations of cyclo-oxygenase inhibitor, aspirin. VWF or thrombin induces p38 activation, which is inhibited in cGMP-dependent protein kinase (PKG)-knockout mouse platelets and PKG inhibitor-treated human platelets, indicating that activation of p38 is downstream from PKG in the signaling pathway. p38AF or p38 inhibitors diminish PKG-induced phosphorylation of extracellular stimuli-responsive kinase (ERK), which also is important in integrin activation. Thus, p38 plays an important role in mediating PKG-dependent activation of ERK. These data delineate a novel signaling pathway in which platelet agonists sequentially activate PKG, p38, and ERK pathways leading to integrin activation.


Subject(s)
Hemostatics/pharmacology , MAP Kinase Signaling System/drug effects , Platelet Activation/drug effects , Platelet Glycoprotein GPIIb-IIIa Complex/agonists , Thrombin/pharmacology , von Willebrand Factor/pharmacology , Amino Acid Substitution , Animals , Aspirin/pharmacology , Blood Platelets/metabolism , Cell Line , Cyclic GMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic GMP-Dependent Protein Kinases/deficiency , Cyclic GMP-Dependent Protein Kinases/metabolism , Cyclooxygenase Inhibitors/pharmacology , Dose-Response Relationship, Drug , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , MAP Kinase Signaling System/physiology , Mice , Mice, Knockout , Phosphorylation/drug effects , Platelet Activation/physiology , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Platelet Glycoprotein GPIb-IX Complex/metabolism , Point Mutation , Protein Processing, Post-Translational/drug effects , Protein Processing, Post-Translational/physiology , Thrombin/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism , von Willebrand Factor/metabolism
17.
J Immunol ; 175(3): 1919-29, 2005 Aug 01.
Article in English | MEDLINE | ID: mdl-16034136

ABSTRACT

The regulation of neutrophil functions by Type I cGMP-dependent protein kinase (cGKI) was investigated in wild-type (WT) and cGKI-deficient (cGKI-/-) mice. We demonstrate that murine neutrophils expressed cGKIalpha. Similar to the regulation of Ca2+ by cGKI in other cells, there was a cGMP-dependent decrease in Ca2+ transients in response to C5a in WT, but not cGKI-/- bone marrow neutrophils. In vitro chemotaxis of bone marrow neutrophils to C5a or IL-8 was significantly greater in cGKI-/- than in WT. Enhanced chemotaxis was also observed with cGKI-/- peritoneal exudate neutrophils (PE-N). In vivo chemotaxis with an arachidonic acid-induced inflammatory ear model revealed an increase in both ear weight and myeloperoxidase (MPO) activity in ear punches of cGKI-/- vs WT mice. These changes were attributable to enhanced vascular permeability and increased neutrophil infiltration. The total extractable content of MPO, but not lysozyme, was significantly greater in cGKI-/- than in WT PE-N. Furthermore, the percentage of MPO released in response to fMLP from cGKI-/- (69%) was greater than that from WT PE-N (36%). PMA failed to induce MPO release from PE-N of either genotype. In contrast, fMLP and PMA released equivalent amounts of lysozyme from PE-N. However, the percentage released was less in cGKI-/- (approximately 60%) than in WT (approximately 90%) PE-N. Superoxide release (maximum velocity) revealed no genotype differences in responses to PMA or fMLP stimulation. In summary, these results show that cGKIalpha down-regulates Ca2+ transients and chemotaxis in murine neutrophils. The regulatory influences of cGKIalpha on the secretagogue responses are complex, depending on the granule subtype.


Subject(s)
Cyclic GMP-Dependent Protein Kinases/deficiency , Cyclic GMP-Dependent Protein Kinases/genetics , Neutrophils/enzymology , Neutrophils/pathology , Animals , Ascitic Fluid/enzymology , Ascitic Fluid/metabolism , Ascitic Fluid/pathology , Bone Marrow Cells/enzymology , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Calcium/antagonists & inhibitors , Calcium/metabolism , Cell Lineage/genetics , Cell Migration Inhibition , Chemotaxis, Leukocyte/genetics , Cyclic GMP-Dependent Protein Kinases/biosynthesis , Cyclic GMP-Dependent Protein Kinases/physiology , Cytoplasmic Granules/enzymology , Cytoplasmic Granules/metabolism , Cytosol/metabolism , Leukocyte Count , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Activation/genetics , Neutrophils/metabolism , Respiratory Burst/genetics , Superoxides/metabolism
18.
Eur J Neurosci ; 20(12): 3498-506, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15610182

ABSTRACT

Cyclic GMP is a second messenger for nitric oxide (NO) that acts as a mediator for many different physiological functions. The cGMP-dependent protein kinases (cGKs) mediate cellular signalling induced by NO and cGMP. Here, we explored the localization of cGMP-dependent protein kinase type II (cGKII) in the mouse brain. In situ hybridization revealed high levels of cGKII mRNA in cerebral cortex, thalamic nuclei, hypothalamic nuclei, and in several basal forebrain regions including medial septum, striatum and amygdala. The close link to NO and the distribution pattern of cGKII suggested that this enzyme might be involved in emotional reactions and responses to drugs of abuse. Therefore, cGKII knockout animals (cGKII-/-) were compared with littermate controls in behavioural tests (i) for emotion-linked and (ii) for acute and chronic ethanol responses. Deletion of cGKII did not influence aggressive behaviour but led to enhanced anxiety-like behaviour. In terms of acute responses to ethanol, cGKII-/- mice were hyposensitive to hypnotic doses of ethanol as measured by the loss of righting reflex, without an alteration in their blood alcohol elimination. In a two-bottle free choice test, cGKII-/- mice showed elevated alcohol consumption. No taste differences to sweet solutions were observed compared to control animals. In summary, our data show that cGKII activity modulates anxiety-like behaviour and neurobehavioural effects of alcohol.


Subject(s)
Alcohol Drinking/metabolism , Cyclic GMP-Dependent Protein Kinases/physiology , Cyclic GMP/physiology , Emotions/physiology , Nitric Oxide/physiology , Signal Transduction , Alcohol Drinking/genetics , Alcohol Drinking/psychology , Animals , Anxiety/enzymology , Brain/metabolism , Cyclic GMP-Dependent Protein Kinase Type II , Cyclic GMP-Dependent Protein Kinases/biosynthesis , Cyclic GMP-Dependent Protein Kinases/deficiency , Cyclic GMP-Dependent Protein Kinases/genetics , Ethanol/administration & dosage , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide/biosynthesis , Nitric Oxide/genetics , Signal Transduction/genetics , Signal Transduction/physiology
19.
Proc Natl Acad Sci U S A ; 101(9): 3253-7, 2004 Mar 02.
Article in English | MEDLINE | ID: mdl-14973199

ABSTRACT

cGMP-dependent protein kinase I (PKG-I) has been suggested to contribute to the facilitation of nociceptive transmission in the spinal cord presumably by acting as a downstream target of nitric oxide. However, PKG-I activators caused conflicting effects on nociceptive behavior. In the present study we used PKG-I(-/-) mice to further assess the role of PKG-I in nociception. PKG-I deficiency was associated with reduced nociceptive behavior in the formalin assay and zymosan-induced paw inflammation. However, acute thermal nociception in the hot-plate test was unaltered. After spinal delivery of the PKG inhibitor, Rp-8-Br-cGMPS, nociceptive behavior of PKG-I(+/+) mice was indistinguishable from that of PKG-I(-/-) mice. On the other hand, the PKG activator, 8-Br-cGMP (250 nmol intrathecally) caused mechanical allodynia only in PKG-I(+/+) mice, indicating that the presence of PKG-I was essential for this effect. Immunofluorescence studies of the spinal cord revealed additional morphological differences. In the dorsal horn of 3- to 4-week-old PKG-I(-/-) mice laminae I-III were smaller and contained fewer neurons than controls. Furthermore, the density of substance P-positive neurons and fibers was significantly reduced. The paucity of substance P in laminae I-III may contribute to the reduction of nociception in PKG-I(-/-) mice and suggests a role of PKG-I in substance P synthesis.


Subject(s)
Cyclic GMP-Dependent Protein Kinases/deficiency , Cyclic GMP/analogs & derivatives , Hyperalgesia/prevention & control , Pain/genetics , Pain/physiopathology , Animals , Cyclic GMP/pharmacology , Cyclic GMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic GMP-Dependent Protein Kinases/genetics , Disease Models, Animal , Formaldehyde , Hot Temperature , Hyperalgesia/genetics , Inflammation/genetics , Inflammation/prevention & control , Mice , Mice, Knockout , Neurons/physiology , Pain/chemically induced , Reaction Time , Substance P/physiology , Thionucleotides/pharmacology
20.
Blood ; 103(7): 2601-9, 2004 Apr 01.
Article in English | MEDLINE | ID: mdl-14684423

ABSTRACT

Glycoprotein Ib-IX-V (GPIb-IX-V) mediates platelet tethering to von Willebrand factor (VWF), recruiting platelets into the thrombus, and activates integrin alphaIIbbeta3 through a pathway that is dependent on Src kinases. In addition, recent reports indicate that activation of alphaIIbbeta3 by VWF is dependent on protein kinase G (PKG) and mitogen-activated protein (MAP) kinases. The present study compares the importance of these signaling pathways in the activation of alphaIIbbeta3 by GPIb-IX-V. In contrast to a recent report, VWF did not promote an increase in cyclic guanosine monophosphate (cGMP), while agents that elevate cGMP, such as the nitrous oxide (NO) donor glyco-SNAP-1 (N-(beta-D-glucopyranosyl)-N2-acetyl-S-nitroso-D,L-penicillaminamide) or the type 5 phosphosdiesterase inhibitor, sildenafil, inhibited rather than promoted activation of alphaIIbbeta3 by GPIb-IX-V and blocked aggregate formation on collagen at an intermediate rate of shear (800 s(-1)). Additionally, sildenafil increased blood flow in a rabbit model of thrombus formation in vivo. A novel inhibitor of the MAP kinase pathway, which is active in plasma, PD184161, had no effect on aggregate formation on collagen under flow conditions, whereas a novel inhibitor of Src kinases, which is also active in plasma, PD173952, blocked this response. These results demonstrate a critical role for Src kinases but not MAP kinases in VWF-dependent platelet activation and demonstrate an inhibitory role for cGMP-elevating agents in regulating this process.


Subject(s)
Blood Platelets/physiology , Cyclic GMP-Dependent Protein Kinases/blood , Mitogen-Activated Protein Kinases/blood , Platelet Activation/physiology , Platelet Glycoprotein GPIb-IX Complex/physiology , Platelet Membrane Glycoproteins , Animals , Blood Platelets/drug effects , Cyclic GMP/blood , Cyclic GMP-Dependent Protein Kinases/deficiency , Cyclic GMP-Dependent Protein Kinases/genetics , Humans , Kinetics , Mice , Mice, Knockout , Nitric Oxide Donors/pharmacology , Receptors, Antigen, B-Cell/blood , von Willebrand Factor/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...