Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Sci Rep ; 9(1): 6993, 2019 05 06.
Article in English | MEDLINE | ID: mdl-31061522

ABSTRACT

Acute-on-chronic liver disease is a clinical syndrome characterized by decompensated liver fibrosis, portal hypertension and splanchnic hyperdynamic circulation. We aimed to determine whether the alpha-1 agonist phenylephrine (Phe) facilitates endothelial nitric oxide (NO) release by mesenteric resistance arteries (MRA) in rats subjected to an experimental microsurgical obstructive liver cholestasis model (LC). Sham-operated (SO) and LC rats were maintained for eight postoperative weeks. Phe-induced vasoconstriction (in the presence/absence of the NO synthase -NOS- inhibitor L-NAME) and vasodilator response to NO donor DEA-NO were analysed. Phe-induced NO release was determined in the presence/absence of either H89 (protein kinase -PK- A inhibitor) or LY 294002 (PI3K inhibitor). PKA and PKG activities, alpha-1 adrenoceptor, endothelial NOS (eNOS), PI3K, AKT and soluble guanylate cyclase (sGC) subunit expressions, as well as eNOS and AKT phosphorylation, were determined. The results show that LC blunted Phe-induced vasoconstriction, and enhanced DEA-NO-induced vasodilation. L-NAME increased the Phe-induced contraction largely in LC animals. The Phe-induced NO release was greater in MRA from LC animals. Both H89 and LY 294002 reduced NO release in LC. Alpha-1 adrenoceptor, eNOS, PI3K and AKT expressions were unchanged, but sGC subunit expression, eNOS and AKT phosphorylation and the activities of PKA and PKG were higher in MRA from LC animals. In summary, these mechanisms may help maintaining splanchnic vasodilation and hypotension observed in decompensated LC.


Subject(s)
Acute-On-Chronic Liver Failure/drug therapy , Adrenergic alpha-1 Receptor Antagonists/pharmacology , Cyclic AMP-Dependent Protein Kinases/genetics , Liver Cirrhosis/drug therapy , Mesenteric Arteries/drug effects , Phenylephrine/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/genetics , Acute-On-Chronic Liver Failure/genetics , Acute-On-Chronic Liver Failure/metabolism , Acute-On-Chronic Liver Failure/pathology , Animals , Cholestasis/genetics , Cholestasis/metabolism , Cholestasis/pathology , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic GMP/metabolism , Cyclic GMP-Dependent Protein Kinase Type I/antagonists & inhibitors , Cyclic GMP-Dependent Protein Kinase Type I/genetics , Cyclic GMP-Dependent Protein Kinase Type I/metabolism , Hypertension, Portal/metabolism , Hypertension, Portal/pathology , Isoquinolines/pharmacology , Liver/drug effects , Liver/metabolism , Liver/pathology , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Male , Mesenteric Arteries/metabolism , Mesenteric Arteries/pathology , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/antagonists & inhibitors , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Receptors, Adrenergic, alpha-1/genetics , Receptors, Adrenergic, alpha-1/metabolism , Sulfonamides/pharmacology , Vasoconstriction/drug effects
2.
Nature ; 566(7745): 548-552, 2019 02.
Article in English | MEDLINE | ID: mdl-30760924

ABSTRACT

Singlet molecular oxygen (1O2) has well-established roles in photosynthetic plants, bacteria and fungi1-3, but not in mammals. Chemically generated 1O2 oxidizes the amino acid tryptophan to precursors of a key metabolite called N-formylkynurenine4, whereas enzymatic oxidation of tryptophan to N-formylkynurenine is catalysed by a family of dioxygenases, including indoleamine 2,3-dioxygenase 15. Under inflammatory conditions, this haem-containing enzyme is expressed in arterial endothelial cells, where it contributes to the regulation of blood pressure6. However, whether indoleamine 2,3-dioxygenase 1 forms 1O2 and whether this contributes to blood pressure control have remained unknown. Here we show that arterial indoleamine 2,3-dioxygenase 1 regulates blood pressure via formation of 1O2. We observed that in the presence of hydrogen peroxide, the enzyme generates 1O2 and that this is associated with the stereoselective oxidation of L-tryptophan to a tricyclic hydroperoxide via a previously unrecognized oxidative activation of the dioxygenase activity. The tryptophan-derived hydroperoxide acts in vivo as a signalling molecule, inducing arterial relaxation and decreasing blood pressure; this activity is dependent on Cys42 of protein kinase G1α. Our findings demonstrate a pathophysiological role for 1O2 in mammals through formation of an amino acid-derived hydroperoxide that regulates vascular tone and blood pressure under inflammatory conditions.


Subject(s)
Blood Pressure/physiology , Inflammation/blood , Inflammation/physiopathology , Singlet Oxygen/metabolism , Vasodilator Agents/metabolism , Animals , Cell Line , Cyclic GMP-Dependent Protein Kinase Type I/antagonists & inhibitors , Cyclic GMP-Dependent Protein Kinase Type I/chemistry , Cyclic GMP-Dependent Protein Kinase Type I/metabolism , Cysteine/metabolism , Enzyme Activation/drug effects , Female , Humans , Hydrogen Peroxide/chemistry , Hydrogen Peroxide/metabolism , Hydrogen Peroxide/pharmacology , Indoleamine-Pyrrole 2,3,-Dioxygenase/chemistry , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Inflammation/enzymology , Male , Oxidation-Reduction/drug effects , Rats , Signal Transduction , Singlet Oxygen/chemistry , Tryptophan/chemistry , Tryptophan/metabolism
3.
J Biol Chem ; 293(28): 10985-10992, 2018 07 13.
Article in English | MEDLINE | ID: mdl-29769318

ABSTRACT

Activation of protein kinase G (PKG) Iα in nociceptive neurons induces long-term hyperexcitability that causes chronic pain. Recently, a derivative of the fungal metabolite balanol, N46, has been reported to inhibit PKG Iα with high potency and selectivity and attenuate thermal hyperalgesia and osteoarthritic pain. Here we determined co-crystal structures of the PKG Iα C-domain and cAMP-dependent protein kinase (PKA) Cα, each bound with N46, at 1.98 Å and 2.65 Å, respectively. N46 binds the active site with its external phenyl ring, specifically interacting with the glycine-rich loop and the αC helix. Phe-371 at the PKG Iα glycine-rich loop is oriented parallel to the phenyl ring of N46, forming a strong π-stacking interaction, whereas the analogous Phe-54 in PKA Cα rotates 30° and forms a weaker interaction. Structural comparison revealed that steric hindrance between the preceding Ser-53 and the propoxy group of the phenyl ring may explain the weaker interaction with PKA Cα. The analogous Gly-370 in PKG Iα, however, causes little steric hindrance with Phe-371. Moreover, Ile-406 on the αC helix forms a hydrophobic interaction with N46 whereas its counterpart in PKA, Thr-88, does not. Substituting these residues in PKG Iα with those in PKA Cα increases the IC50 values for N46, whereas replacing these residues in PKA Cα with those in PKG Iα reduces the IC50, consistent with our structural findings. In conclusion, our results explain the structural basis for N46-mediated selective inhibition of human PKG Iα and provide a starting point for structure-guided design of selective PKG Iα inhibitors.


Subject(s)
Azepines/chemistry , Azepines/pharmacology , Cyclic GMP-Dependent Protein Kinase Type I/antagonists & inhibitors , Cyclic GMP-Dependent Protein Kinase Type I/metabolism , Hydroxybenzoates/chemistry , Hydroxybenzoates/pharmacology , Catalytic Domain , Crystallography, X-Ray , Cyclic GMP-Dependent Protein Kinase Type I/chemistry , Humans , Models, Molecular , Phosphorylation , Protein Conformation
4.
J Biol Chem ; 293(21): 7916-7929, 2018 05 25.
Article in English | MEDLINE | ID: mdl-29602907

ABSTRACT

The type I cGMP-dependent protein kinases (PKG I) serve essential physiological functions, including smooth muscle relaxation, cardiac remodeling, and platelet aggregation. These enzymes form homodimers through their N-terminal dimerization domains, a feature implicated in regulating their cooperative activation. Previous investigations into the activation mechanisms of PKG I isoforms have been largely influenced by structures of the cAMP-dependent protein kinase (PKA). Here, we examined PKG Iα activation by cGMP and cAMP by engineering a monomeric form that lacks N-terminal residues 1-53 (Δ53). We found that the construct exists as a monomer as assessed by whole-protein MS, size-exclusion chromatography, and small-angle X-ray scattering (SAXS). Reconstruction of the SAXS 3D envelope indicates that Δ53 has a similar shape to the heterodimeric RIα-C complex of PKA. Moreover, we found that the Δ53 construct is autoinhibited in its cGMP-free state and can bind to and be activated by cGMP in a manner similar to full-length PKG Iα as assessed by surface plasmon resonance (SPR) spectroscopy. However, we found that the Δ53 variant does not exhibit cooperative activation, and its cyclic nucleotide selectivity is diminished. These findings support a model in which, despite structural similarities, PKG Iα activation is distinct from that of PKA, and its cooperativity is driven by in trans interactions between protomers.


Subject(s)
Cyclic GMP-Dependent Protein Kinase Type I/antagonists & inhibitors , Cyclic GMP-Dependent Protein Kinase Type I/metabolism , Cyclic GMP/metabolism , Protein Multimerization , Amino Acid Sequence , Animals , Cattle , Crystallography, X-Ray , Enzyme Activation , Humans , Mice , Models, Molecular , Phosphorylation , Protein Binding , Rats , Scattering, Small Angle , Sequence Homology
5.
Diab Vasc Dis Res ; 14(5): 434-449, 2017 09.
Article in English | MEDLINE | ID: mdl-28631500

ABSTRACT

Early studies showed nitric oxide as a pro-inflammatory-cytokine-induced toxin involved in pancreatic ß-cell destruction during pathogenesis of type-1 diabetes. However, nitric oxide has both cytotoxic and cytoprotective effects on mammalian cells, depending on concentration and micro-environmental surroundings. Our studies have shown that low/physiological-level nitric oxide selectively activates protein kinase G type Iα isoform, promoting cytoprotective/pro-cell-survival effects in many cell types. In bone marrow-derived stromal/mesenchymal stem cells, protein kinase G type Iα mediates autocrine effects of nitric oxide and atrial natriuretic peptide, promoting DNA-synthesis/proliferation and cell survival. In this study, endothelial nitric oxide synthase/neuronal nitric oxide synthase inhibitor L-NIO (L-N(5)-(1-iminoethyl)ornithine), soluble guanylyl cyclase inhibitor ODQ (1H-[1,2,4]oxadiazolo[4,3,-a] quinoxalin-1-one), atrial natriuretic peptide-receptor inhibitor A71915 and protein kinase G type Iα kinase activity inhibitor DT-2 all increased apoptosis and decreased insulin secretion in RINm5F pancreatic ß-cells, suggesting autocrine regulatory role for endogenous nitric oxide- and atrial natriuretic peptide-induced activation of protein kinase G type Iα. In four pancreatic ß-cell lines, Beta-TC-6, RINm5F, INS-1 and 1.1B4, protein kinase G type Iα small-interfering RNA decreased phospho-serine-239-VASP (indicator of endogenous protein kinase G type Iα kinase activity), increased apoptosis and decreased proliferation. In protein kinase G type Iα-knockdown ß-cell lines, expressions of phospho-protein kinase B (PKB/AKT) (AKT), phospho-Forkhead box protein O1 (FOXO1) (transcriptional repressor of pancreas duodenum homobox-1) and pancreas duodenum homobox-1 were decreased, suppressing proliferation and survival in pancreatic ß-cells. The data suggest autocrine nitric oxide/atrial natriuretic peptide-induced activation of protein kinase G type Iα/p-AKT/p-FOXO1 promotes survival and proliferation in pancreatic ß-cells, providing therapeutic implications for development of new therapeutic agents for diabetes.


Subject(s)
Cell Proliferation , Cyclic GMP-Dependent Protein Kinase Type I/metabolism , Forkhead Box Protein O1/metabolism , Insulin-Secreting Cells/enzymology , Proto-Oncogene Proteins c-akt/metabolism , Animals , Apoptosis , Atrial Natriuretic Factor/metabolism , Autocrine Communication , Cell Line , Cell Proliferation/drug effects , Cell Survival , Coculture Techniques , Cyclic GMP/metabolism , Cyclic GMP-Dependent Protein Kinase Type I/antagonists & inhibitors , Cyclic GMP-Dependent Protein Kinase Type I/genetics , Enzyme Activation , Homeodomain Proteins/metabolism , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Male , Mesenchymal Stem Cells/metabolism , Mice, Inbred C57BL , Nitric Oxide/metabolism , Phosphorylation , Protein Kinase Inhibitors/pharmacology , RNA Interference , Signal Transduction , Tissue Culture Techniques , Trans-Activators/metabolism , Transfection
6.
Redox Biol ; 11: 176-191, 2017 04.
Article in English | MEDLINE | ID: mdl-27978504

ABSTRACT

Cyclic GMP-dependent protein kinase 1 (PKG1) mediates presynaptic nociceptive long-term potentiation (LTP) in the spinal cord and contributes to inflammatory pain in rodents but the present study revealed opposite effects in the context of neuropathic pain. We used a set of loss-of-function models for in vivo and in vitro studies to address this controversy: peripheral neuron specific deletion (SNS-PKG1-/-), inducible deletion in subsets of neurons (SLICK-PKG1-/-) and redox-dead PKG1 mutants. In contrast to inflammatory pain, SNS-PKG1-/- mice developed stronger neuropathic hyperalgesia associated with an impairment of nerve regeneration, suggesting specific repair functions of PKG1. Although PKG1 accumulated at the site of injury, its activity was lost in the proximal nerve due to a reduction of oxidation-dependent dimerization, which was a consequence of mitochondrial damage in injured axons. In vitro, PKG1 deficiency or its redox-insensitivity resulted in enhanced outgrowth and reduction of growth cone collapse in response to redox signals, which presented as oxidative hotspots in growing cones. At the molecular level, PKG1 deficiency caused a depletion of phosphorylated cofilin, which is essential for growth cone collapse and guidance. Hence, redox-mediated guidance required PKG1 and consequently, its deficiency in vivo resulted in defective repair and enhanced neuropathic pain after nerve injury. PKG1-dependent repair functions will outweigh its signaling functions in spinal nociceptive LTP, so that inhibition of PKG1 is no option for neuropathic pain.


Subject(s)
Axons/metabolism , Cyclic GMP-Dependent Protein Kinase Type I/genetics , Hyperalgesia/genetics , Neuralgia/genetics , Animals , Axons/pathology , Cyclic GMP/genetics , Cyclic GMP-Dependent Protein Kinase Type I/antagonists & inhibitors , Humans , Hyperalgesia/metabolism , Hyperalgesia/pathology , Long-Term Potentiation/genetics , Mice , Neuralgia/metabolism , Neuralgia/pathology , Neurons/metabolism , Neurons/pathology , Oxidation-Reduction , Signal Transduction
7.
Biol Pharm Bull ; 40(3): 365-374, 2017 Mar 01.
Article in English | MEDLINE | ID: mdl-27980245

ABSTRACT

Guanosine 3',5'-cyclic monophosphate (cGMP)-dependent protein kinases (PKG) are kinases regulating diverse physiological functions including vascular smooth muscle relaxation, neuronal synaptic plasticity, and platelet activities. Certain PKG inhibitors, such as Rp-diastereomers of derivatives of guanosine 3',5'-cyclic monophosphorothioate (Rp-cGMPS), have been designed and used to study PKG-regulated cell signaling. 8-Nitroguanosine 3',5'-cyclic monophosphate (8-nitro-cGMP) is an endogenous cGMP derivative formed as a result of excess production of reactive oxygen species and nitric oxide. 8-Nitro-cGMP causes persistent activation of PKG1α through covalent attachment of cGMP moieties to cysteine residues of the enzyme (i.e., the process called protein S-guanylation). In this study, we synthesized a nitrated analogue of Rp-cGMPS, 8-nitroguanosine 3',5'-cyclic monophosphorothioate Rp-isomer (Rp-8-nitro-cGMPS), and investigated its effects on PKG1α activity. We synthesized Rp-8-nitro-cGMPS by reacting Rp-8-bromoguanosine 3',5'-cyclic monophosphorothioate (Rp-8-bromo-cGMPS) with sodium nitrite. Rp-8-Nitro-cGMPS reacted with the thiol compounds cysteine and glutathione to form Rp-8-thioalkoxy-cGMPS adducts to a similar extent as did 8-nitro-cGMP. As an important finding, a protein S-guanylation-like modification was clearly observed, by using Western blotting, in the reaction between recombinant PKG1α and Rp-8-nitro-cGMPS. Rp-8-Nitro-cGMPS inhibited PKG1α activity with an inhibitory constant of 22 µM in a competitive manner. An organ bath assay with mouse aorta demonstrated that Rp-8-nitro-cGMPS inhibited vascular relaxation induced by acetylcholine or 8-bromo-cGMP more than Rp-8-bromo-cGMPS did. These findings suggest that Rp-8-nitro-cGMPS inhibits PKG through induction of an S-guanylation-like modification by attaching the Rp-cGMPS moiety to the enzyme. Additional study is warranted to explore the potential application of Rp-8-nitro-cGMPS to biochemical and therapeutic research involving PKG1α activation.


Subject(s)
Cyclic GMP-Dependent Protein Kinase Type I/antagonists & inhibitors , Cyclic GMP/analogs & derivatives , Guanosine/analogs & derivatives , Nitro Compounds/pharmacology , Thionucleotides/pharmacology , Vasodilation/drug effects , Acetylcholine , Animals , Aorta , Cyclic GMP/chemical synthesis , Cyclic GMP/metabolism , Cyclic GMP/pharmacology , Cyclic GMP-Dependent Protein Kinase Type I/metabolism , Guanosine/metabolism , Guanosine/pharmacology , Isomerism , Male , Mice, Inbred C57BL , Nitro Compounds/metabolism , Protein Processing, Post-Translational , Signal Transduction , Thionucleotides/chemical synthesis , Thionucleotides/metabolism
8.
FEBS Lett ; 591(1): 221-230, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27914169

ABSTRACT

The R-diastereomer of phosphorothioate analogs of cGMP, Rp-cGMPS, is one of few known inhibitors of cGMP-dependent protein kinase I (PKG I); however, its mechanism of inhibition is currently not fully understood. Here, we determined the crystal structure of the PKG Iß cyclic nucleotide-binding domain (PKG Iß CNB-B), considered a 'gatekeeper' for cGMP activation, bound to Rp-cGMPS at 1.3 Å. Our structural and NMR data show that PKG Iß CNB-B bound to Rp-cGMPS displays an apo-like structure with its helical domain in an open conformation. Comparison with the cAMP-dependent protein kinase regulatory subunit (PKA RIα) showed that this conformation resembles the catalytic subunit-bound inhibited state of PKA RIα more closely than the apo or Rp-cAMPS-bound conformations. These results suggest that Rp-cGMPS inhibits PKG I by stabilizing the inactive conformation of CNB-B.


Subject(s)
Apoenzymes/chemistry , Apoenzymes/metabolism , Cyclic GMP-Dependent Protein Kinase Type I/chemistry , Cyclic GMP-Dependent Protein Kinase Type I/metabolism , Cyclic GMP/analogs & derivatives , Cyclic GMP/metabolism , Thionucleotides/metabolism , Amino Acid Sequence , Crystallography, X-Ray , Cyclic GMP/chemistry , Cyclic GMP-Dependent Protein Kinase Type I/antagonists & inhibitors , Enzyme Stability , Kinetics , Magnetic Resonance Spectroscopy , Protein Conformation , Protein Domains , Stereoisomerism , Thionucleotides/chemistry
9.
BMB Rep ; 50(4): 208-213, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27802840

ABSTRACT

Vascular endothelial growth factor (VEGF) is an essential cytokine that has functions in the formation of new blood vessels and regression of cardiac hypertrophy. VEGF/VEGF-receptor-1 (VEGFR1) signaling plays a key role in the regression of cardiac hypertrophy, whereas VEGF/VEGFR2 signaling leads to cardiac hypertrophy. In this study, we identified the prohypertrophic role of miR-374 using neonatal rat ventricular myocytes (NRVMs). Our results showed that overexpression of miR-374 activated G protein-coupled receptor-mediated prohypertrophic pathways by the inhibition of VEGFR1-dependent regression pathways. Luciferase assays revealed that miR-374 could directly target the 3'-untranslated regions of VEGFR1 and cGMP-dependent protein kinase-1. Collectively, these findings demonstrated that miR-374 was a novel pro-hypertrophic microRNA functioning to suppress the VEGFR1-mediated regression pathway. [BMB Reports 2017; 50(4): 208-213].


Subject(s)
MicroRNAs/metabolism , Signal Transduction , Vascular Endothelial Growth Factor Receptor-1/metabolism , 3' Untranslated Regions , Animals , Antagomirs/metabolism , Base Sequence , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cardiomegaly/genetics , Cardiomegaly/metabolism , Cardiomegaly/pathology , Cells, Cultured , Cyclic GMP-Dependent Protein Kinase Type I/antagonists & inhibitors , Cyclic GMP-Dependent Protein Kinase Type I/genetics , Cyclic GMP-Dependent Protein Kinase Type I/metabolism , Genes, Reporter , MEF2 Transcription Factors/metabolism , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , NFATC Transcription Factors/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Sequence Alignment , Vascular Endothelial Growth Factor Receptor-1/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-1/genetics
10.
Am J Physiol Heart Circ Physiol ; 311(4): H904-H912, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27521420

ABSTRACT

Homeostatic control of vascular smooth muscle cell (VSMC) differentiation is critical for contractile activity and regulation of blood flow. Recently, we reported that precontracted blood vessels are relaxed and the phenotype of VSMC is regulated from a synthetic to contractile state by glucose-6-phosphate dehydrogenase (G6PD) inhibition. In the current study, we investigated whether the increase in the expression of VSMC contractile proteins by inhibition and knockdown of G6PD is mediated through a protein kinase G (PKG)-dependent pathway and whether it regulates blood pressure. We found that the expression of VSMC-restricted contractile proteins, myocardin (MYOCD), and miR-1 and miR-143 are increased by G6PD inhibition or knockdown. Importantly, RNA-sequence analysis of aortic tissue from G6PD-deficient mice revealed uniform increases in VSMC-restricted genes, particularly those regulated by the MYOCD-serum response factor (SRF) switch. Conversely, expression of Krüppel-like factor 4 (KLF4) is decreased by G6PD inhibition. Interestingly, the G6PD inhibition-induced expression of miR-1 and contractile proteins was blocked by Rp-ß-phenyl-1,N2-etheno-8-bromo-guanosine-3',5'-cyclic monophosphorothioate, a PKG inhibitor. On the other hand, MYOCD and miR-143 levels are increased by G6PD inhibition through a PKG-independent manner. Furthermore, blood pressure was lower in the G6PD-deficient compared with wild-type mice. Therefore, our results suggest that the expression of VSMC contractile proteins induced by G6PD inhibition occurs via PKG1α-dependent and -independent pathways.


Subject(s)
Aorta/metabolism , Contractile Proteins/genetics , Cyclic GMP-Dependent Protein Kinase Type I/metabolism , Glucosephosphate Dehydrogenase/antagonists & inhibitors , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Animals , Aorta/drug effects , Blotting, Western , Cattle , Chromatography, Liquid , Contractile Proteins/drug effects , Contractile Proteins/metabolism , Cyclic GMP-Dependent Protein Kinase Type I/antagonists & inhibitors , Cyclic GMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic GMP-Dependent Protein Kinases/metabolism , Gene Knockdown Techniques , Glucosephosphate Dehydrogenase/genetics , Immunoprecipitation , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/drug effects , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Mice , MicroRNAs/drug effects , MicroRNAs/genetics , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Nuclear Proteins/drug effects , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Polymerase Chain Reaction , Rats , Serum Response Factor/drug effects , Serum Response Factor/genetics , Serum Response Factor/metabolism , Tandem Mass Spectrometry , Trans-Activators/drug effects , Trans-Activators/genetics , Trans-Activators/metabolism
11.
J Biol Chem ; 291(33): 17427-36, 2016 08 12.
Article in English | MEDLINE | ID: mdl-27342776

ABSTRACT

Phosphodiesterase 5 (PDE5) inhibitors limit myocardial injury caused by stresses, including doxorubicin chemotherapy. cGMP binding to PKG Iα attenuates oxidant-induced disulfide formation. Because PDE5 inhibition elevates cGMP and protects from doxorubicin-induced injury, we reasoned that this may be because it limits PKG Iα disulfide formation. To investigate the role of PKG Iα disulfide dimerization in the development of apoptosis, doxorubicin-induced cardiomyopathy was compared in male wild type (WT) or disulfide-resistant C42S PKG Iα knock-in (KI) mice. Echocardiography showed that doxorubicin treatment caused loss of myocardial tissue and depressed left ventricular function in WT mice. Doxorubicin also reduced pro-survival signaling and increased apoptosis in WT hearts. In contrast, KI mice were markedly resistant to the dysfunction induced by doxorubicin in WTs. In follow-on experiments the influence of the PDE5 inhibitor tadalafil on the development of doxorubicin-induced cardiomyopathy in WT and KI mice was investigated. In WT mice, co-administration of tadalafil with doxorubicin reduced PKG Iα oxidation caused by doxorubicin and also protected against cardiac injury and loss of function. KI mice were again innately resistant to doxorubicin-induced cardiotoxicity, and therefore tadalafil afforded no additional protection. Doxorubicin decreased phosphorylation of RhoA (Ser-188), stimulating its GTPase activity to activate Rho-associated protein kinase (ROCK) in WTs. These pro-apoptotic events were absent in KI mice and were attenuated in WTs co-administered tadalafil. PKG Iα disulfide formation triggers cardiac injury, and this initiation of maladaptive signaling can be blocked by pharmacological therapies that elevate cGMP, which binds kinase to limit its oxidation.


Subject(s)
Cardiomegaly , Cyclic GMP-Dependent Protein Kinase Type I , Disulfides/metabolism , Doxorubicin , Heart Failure , Phosphodiesterase 5 Inhibitors/pharmacology , Second Messenger Systems , Tadalafil/pharmacology , Animals , Cardiomegaly/chemically induced , Cardiomegaly/enzymology , Cardiomegaly/genetics , Cardiomegaly/prevention & control , Cyclic GMP/genetics , Cyclic GMP/metabolism , Cyclic GMP-Dependent Protein Kinase Type I/antagonists & inhibitors , Cyclic GMP-Dependent Protein Kinase Type I/genetics , Cyclic GMP-Dependent Protein Kinase Type I/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 5/genetics , Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Doxorubicin/adverse effects , Doxorubicin/pharmacology , Heart Failure/chemically induced , Heart Failure/enzymology , Heart Failure/genetics , Heart Failure/prevention & control , Mice , Mice, Mutant Strains , Oxidation-Reduction , Second Messenger Systems/drug effects , Second Messenger Systems/genetics , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism
12.
Am J Physiol Heart Circ Physiol ; 308(4): H281-90, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25485902

ABSTRACT

Diabetes mellitus increases morbidity/mortality of ischemic heart disease. Although atrial natriuretic peptide and C-type natriuretic peptide reduce the myocardial ischemia-reperfusion damage in nondiabetic rats, whether vasonatrin peptide (VNP), the artificial synthetic chimera of atrial natriuretic peptide and C-type natriuretic peptide, confers cardioprotective effects against ischemia-reperfusion injury, especially in diabetic patients, is still unclear. This study was designed to investigate the effects of VNP on ischemia-reperfusion injury in diabetic rats and to further elucidate its mechanisms. The high-fat diet-fed streptozotocin-induced diabetic Sprague-Dawley rats were subjected to ischemia-reperfusion operation. VNP treatment (100 µg/kg iv, 10 min before reperfusion) significantly improved the instantaneous first derivation of left ventricle pressure (±LV dP/dtmax) and LV systolic pressure and reduced LV end-diastolic pressure, apoptosis index, caspase-3 activity, plasma creatine kinase (CK), and lactate dehydrogenase (LDH) activities. Moreover, VNP inhibited endoplasmic reticulum (ER) stress by suppressing glucose-regulated protein 78 (GRP78) and C/EBP homologous protein (CHOP). These effects were mimicked by 8-bromine-cyclic guanosinemonophosphate (8-Br-cGMP), a cGMP analog, whereas they were inhibited by KT-5823, the selective inhibitor of PKG. In addition, pretreatment with tauroursodeoxycholic acid (TUDCA), a specific inhibitor of ER stress, could not further promote the VNP's cardioprotective effect in diabetic rats. In vitro H9c2 cardiomyocytes were subjected to hypoxia/reoxygenation and incubated with or without VNP (10(-8) mol/l). Gene knockdown of PKG1α with siRNA blunted VNP inhibition of ER stress and apoptosis, while overexpression of PKG1α resulted in significant decreased ER stress and apoptosis. VNP protects the diabetic heart against ischemia-reperfusion injury by inhibiting ER stress via the cGMP-PKG signaling pathway. These results suggest that VNP may have potential therapeutic value for the diabetic patients with ischemic heart disease.


Subject(s)
Atrial Natriuretic Factor/pharmacology , Diabetes Mellitus, Experimental/metabolism , Heart Ventricles/drug effects , Myocardial Reperfusion Injury/metabolism , Animals , Apoptosis , Atrial Natriuretic Factor/therapeutic use , Carbazoles/pharmacology , Caspase 3/metabolism , Cell Hypoxia , Cell Line , Creatine Kinase/blood , Cyclic GMP/analogs & derivatives , Cyclic GMP/pharmacology , Cyclic GMP-Dependent Protein Kinase Type I/antagonists & inhibitors , Cyclic GMP-Dependent Protein Kinase Type I/genetics , Cyclic GMP-Dependent Protein Kinase Type I/metabolism , Diabetes Mellitus, Experimental/complications , Endoplasmic Reticulum Stress , Heart Ventricles/metabolism , Heart Ventricles/pathology , Male , Myocardial Reperfusion Injury/complications , Myocardial Reperfusion Injury/drug therapy , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Rats , Rats, Sprague-Dawley , Taurochenodeoxycholic Acid/pharmacology , Transcription Factor CHOP/metabolism , Ventricular Function/drug effects
13.
Spine (Phila Pa 1976) ; 39(19): 1533-41, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-24921837

ABSTRACT

STUDY DESIGN: A prospective, randomized experimental research. OBJECTIVE: To demonstrate the role of cGMP (cyclic guanosine monophosphate)-cGKI (cGMP-dependent protein kinase I) pathway in dorsal root ganglia (DRG) in bone cancer pain. SUMMARY OF BACKGROUND DATA: Treating bone cancer pain continues to possess a major clinical challenge because the specific cellular and molecular mechanisms underlying bone cancer pain remain elusive. cGMP and cGMP-dependent protein kinases pathway in DRG plays important role in nerve injury-induced hyperexcitability of DRG neurons, as well as neuropathic pain, however, whether this pathway participates in bone cancer pain is unknown. METHODS: The rat model of bone cancer pain was produced by intramedullary injection of rat breast cancer cells (Walker 256) into right tibia. Thermal hyperalgesia and mechanical allodynia were measured before and after administration of inhibitor of cGMP-cGKs pathway (Rp-8-pCPT-cGMPS). Immunofluorescence and reverse transcription-polymerase chain reaction were used to reflect expression of cGKI in DRG neurons, whereas the concentration of cGMP in DRG was tested using enzyme-linked immunosorbent assay method. Whole-cell patch clamp was used to record the hyperexcitability of small neurons in DRG with or without cGKs inhibitor after tumor cell implantation (TCI). RESULTS: TCI treatment significantly increased the concentration of cGMP in DRG and activity of cGKs in DRG and the spinal cord. TCI treatment also induced upregulation of cGKI messenger ribonucleic acid and protein in DRG, as well as enhanced hyperexcitability in DRG neurons. Spinal administration of Rp-8-pCPT-cGMPS, cGMP-cGKs inhibitor, significantly suppressed TCI-induced activation of cGMP-cGKI signaling, and hyperexcitability of DRG neurons. Meanwhile, in vivo intrathecal delivery of the Rp-8-pCPT-cGMPS significantly prevented and suppressed TCI-induced hyperalgesia and allodynia. CONCLUSION: From these results, we confirm that TCI treatment activates cGMP-cGKI signaling pathway and continuing activation of this pathway in DRG is required for hyperalgesia and/or hyperalgesia and allodynia after TCI treatment. LEVEL OF EVIDENCE: N/A.


Subject(s)
Bone Neoplasms/secondary , Carcinoma 256, Walker/secondary , Cyclic GMP-Dependent Protein Kinase Type I/physiology , Cyclic GMP/physiology , Ganglia, Spinal/physiopathology , Hyperalgesia/physiopathology , Neoplasm Proteins/physiology , Sensory Receptor Cells/physiology , Tibia , Animals , Bone Neoplasms/physiopathology , Carcinoma 256, Walker/physiopathology , Cyclic GMP/analogs & derivatives , Cyclic GMP/pharmacology , Cyclic GMP-Dependent Protein Kinase Type I/antagonists & inhibitors , Cyclic GMP-Dependent Protein Kinase Type I/biosynthesis , Cyclic GMP-Dependent Protein Kinase Type I/genetics , Enzyme Induction , Female , Hot Temperature , Hyperalgesia/etiology , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Pain Threshold , Patch-Clamp Techniques , Protein Kinase Inhibitors/pharmacology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , Random Allocation , Rats , Rats, Sprague-Dawley , Thionucleotides/pharmacology , Tibia/innervation , Touch
14.
Biochem Biophys Res Commun ; 446(1): 328-34, 2014 Mar 28.
Article in English | MEDLINE | ID: mdl-24602613

ABSTRACT

Podocyte resistance to the actions of insulin on glucose transport could contribute to the pathogenesis of diabetic podocytopathy (DP) via disturbances in cyclic-dependent protein kinase signaling. To determine whether cGMP-dependent protein kinase (PKG) is involved in the insulin regulation of glucose transport, we measured insulin-dependent glucose uptake into cultured rat podocytes under conditions of modified PKG activity using pharmacological (PKG activator or inhibitor) and biochemical (siRNA PKGIα, siRNA insulin receptor ß) means. Our findings indicate the participation of PKG in insulin-stimulated transport and provide new insights into how PKG may trigger the resistance of glucose transport to insulin in DP.


Subject(s)
Cyclic GMP-Dependent Protein Kinase Type I/metabolism , Glucose/metabolism , Insulin/metabolism , Podocytes/metabolism , Animals , Biological Transport, Active/drug effects , Cells, Cultured , Cyclic GMP-Dependent Protein Kinase Type I/antagonists & inhibitors , Cyclic GMP-Dependent Protein Kinase Type I/genetics , Diabetic Nephropathies/etiology , Diabetic Nephropathies/metabolism , Enzyme Activation , Insulin/pharmacology , Insulin Resistance , Podocytes/drug effects , RNA, Small Interfering/genetics , Rats , Signal Transduction/drug effects
15.
J Biol Chem ; 289(11): 7948-61, 2014 Mar 14.
Article in English | MEDLINE | ID: mdl-24469460

ABSTRACT

The cGMP-dependent protein kinase G-1α (PKG-1α) is a downstream mediator of nitric oxide and natriuretic peptide signaling. Alterations in this pathway play a key role in the pathogenesis and progression of vascular diseases associated with increased vascular tone and thickness, such as pulmonary hypertension. Previous studies have shown that tyrosine nitration attenuates PKG-1α activity. However, little is known about the mechanisms involved in this event. Utilizing mass spectrometry, we found that PKG-1α is susceptible to nitration at tyrosine 247 and 425. Tyrosine to phenylalanine mutants, Y247F- and Y425F-PKG-1α, were both less susceptible to nitration than WT PKG-1α, but only Y247F-PKG-1α exhibited preserved activity, suggesting that the nitration of Tyr(247) is critical in attenuating PKG-1α activity. The overexpression of WT- or Y247F-PKG-1α decreased the proliferation of pulmonary artery smooth muscle cells (SMC), increased the expression of SMC contractile markers, and decreased the expression of proliferative markers. Nitrosative stress induced a switch from a contractile to a synthetic phenotype in cells expressing WT- but not Y247F-PKG-1α. An antibody generated against 3-NT-Y247 identified increased levels of nitrated PKG-1α in humans with pulmonary hypertension. Finally, to gain a more mechanistic understanding of how nitration attenuates PKG activity, we developed a homology model of PKG-1α. This model predicted that the nitration of Tyr(247) would decrease the affinity of PKG-1α for cGMP, which we confirmed using a [(3)H]cGMP binding assay. Our study shows that the nitration of Tyr(247) and the attenuation of cGMP binding is an important mechanism regulating in PKG-1α activity and SMC proliferation/differentiation.


Subject(s)
Cyclic GMP-Dependent Protein Kinase Type I/antagonists & inhibitors , Cyclic GMP/chemistry , Myocytes, Smooth Muscle/metabolism , Nitrogen/chemistry , Tyrosine/chemistry , Adult , Animals , Aorta/cytology , Cardiovascular Diseases/metabolism , Catalytic Domain , Cell Differentiation , Cell Proliferation , Cells, Cultured , Cyclic GMP-Dependent Protein Kinase Type I/metabolism , Female , HEK293 Cells , Humans , Male , Mass Spectrometry , Middle Aged , Models, Molecular , Peroxynitrous Acid/chemistry , Protein Binding , Sheep , Young Adult
16.
Am J Physiol Cell Physiol ; 306(4): C343-53, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24259420

ABSTRACT

The in vitro perfused rectal gland of the dogfish shark (Squalus acanthias) and filter-grown monolayers of primary cultures of shark rectal gland (SRG) epithelial cells were used to analyze the signal transduction pathway by which C-type natriuretic peptide (CNP) stimulates chloride secretion. CNP binds to natriuretic receptors in the basolateral membrane, elevates cellular cGMP, and opens cystic fibrosis transmembrane conductance regulator (CFTR) chloride channels in the apical membrane. CNP-provoked chloride secretion was completely inhibitable by the nonspecific protein kinase inhibitor staurosporine and the PKA inhibitor H89 but insensitive to H8, an inhibitor of type I and II isoforms of cGMP-dependent protein kinase (cGKI and cGKII). CNP-induced secretion could not be mimicked by nonhydrolyzable cGMP analogs added alone or in combination with the protein kinase C activator phorbolester, arguing against a role for cGK or for cGMP-induced PKC signaling. We failed to detect a dogfish ortholog of cGKII by molecular cloning and affinity chromatography. However, inhibitors of the cGMP-inhibitable isoform of phosphodiesterase (PDE3) including milrinone, amrinone, and cilostamide but not inhibitors of other PDE isoenzymes mimicked the effect of CNP on chloride secretion in perfused glands and monolayers. CNP raised cGMP and cAMP levels in the SRG epithelial cells. This rise in cAMP as well as the CNP and amrinone-provoked chloride secretion, but not the rise in cGMP, was almost completely blocked by the Gαi-coupled adenylyl cyclase inhibitor somatostatin, arguing against a role for cGMP cross-activation of PKA in CNP action. These data provide molecular, functional, and pharmacological evidence for a CNP/cGMP/PDE3/cAMP/PKA signaling cascade coupled to CFTR in the SRG.


Subject(s)
Chlorides/metabolism , Cyclic GMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Dogfish/metabolism , Fish Proteins/metabolism , Natriuretic Peptide, C-Type/metabolism , Salt Gland/enzymology , Adenylyl Cyclase Inhibitors , Adenylyl Cyclases/metabolism , Animals , Cells, Cultured , Cloning, Molecular , Cyclic GMP-Dependent Protein Kinase Type I/antagonists & inhibitors , Cyclic GMP-Dependent Protein Kinase Type I/metabolism , Cyclic GMP-Dependent Protein Kinase Type II/antagonists & inhibitors , Cyclic GMP-Dependent Protein Kinase Type II/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/drug effects , Epithelial Cells/drug effects , Epithelial Cells/enzymology , Female , GTP-Binding Protein alpha Subunits, Gi-Go/antagonists & inhibitors , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Ion Channel Gating , Male , Phosphodiesterase 3 Inhibitors/pharmacology , Protein Binding , Protein Kinase Inhibitors/pharmacology , Receptors, Atrial Natriuretic Factor/metabolism , Salt Gland/drug effects , Second Messenger Systems , Time Factors
17.
J Proteome Res ; 12(8): 3792-800, 2013 Aug 02.
Article in English | MEDLINE | ID: mdl-23795919

ABSTRACT

Protein kinases are key regulators of cellular processes, and aberrant function is often associated with human disease. Consequently, kinases represent an important class of therapeutic targets and about 20 kinase inhibitors (KIs) are in clinical use today. Detailed knowledge about the selectivity of KIs is important for the correct interpretation of their pharmacological and systems biological effects. Chemical proteomic approaches for systematic kinase inhibitor selectivity profiling have emerged as important molecular tools in this regard, but the coverage of the human kinome is still incomplete. Here, we describe a new affinity probe targeting Akt and many other members of the AGC kinase family that considerably extends the scope of KI profiling by chemical proteomics. In combination with the previously published kinobeads, the synthesized probe was applied to selectivity profiling of the Akt inhibitors GSK690693 and GSK2141795 in human cancer cells. The results confirmed the inhibition of all Akt isoforms and of a number of known as well as CDC42BPB as a novel putative target for GSK690693. This work also established, for the first time, the kinase selectivity profile of the clinical phase I drug GSK2141795 and identified PRKG1 as a low nanomolar kinase target as well as the ATP-dependent 5'-3' DNA helicase ERCC2 as a potential new non-kinase off-target.


Subject(s)
Antineoplastic Agents/chemistry , Cyclic GMP-Dependent Protein Kinase Type I/genetics , Molecular Probes/chemistry , Protein Kinase Inhibitors/chemistry , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cyclic GMP-Dependent Protein Kinase Type I/antagonists & inhibitors , Cyclic GMP-Dependent Protein Kinase Type I/metabolism , Gene Expression/drug effects , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Isoenzymes/metabolism , Molecular Probes/chemical synthesis , Myotonin-Protein Kinase , Oxadiazoles/chemistry , Oxadiazoles/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Proteomics/methods , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Sepharose/chemistry , Xeroderma Pigmentosum Group D Protein/antagonists & inhibitors , Xeroderma Pigmentosum Group D Protein/genetics , Xeroderma Pigmentosum Group D Protein/metabolism
18.
Am J Physiol Heart Circ Physiol ; 305(3): H365-77, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23729209

ABSTRACT

Nitroxyl (HNO) is a redox congener of NO. We now directly compare the antihypertrophic efficacy of HNO and NO donors in neonatal rat cardiomyocytes and compare their contributing mechanisms of actions in this setting. Isopropylamine-NONOate (IPA-NO) elicited concentration-dependent inhibition of endothelin-1 (ET1)-induced increases in cardiomyocyte size, with similar suppression of hypertrophic genes. Antihypertrophic IPA-NO actions were significantly attenuated by l-cysteine (HNO scavenger), Rp-8-pCTP-cGMPS (cGMP-dependent protein kinase inhibitor), and 1-H-(1,2,4)-oxodiazolo-quinxaline-1-one [ODQ; to target soluble guanylyl cyclase (sGC)] but were unaffected by carboxy-PTIO (NO scavenger) or CGRP8-37 (calcitonin gene-related peptide antagonist). Furthermore, IPA-NO significantly increased cardiomyocyte cGMP 3.5-fold (an l-cysteine-sensitive effect) and stimulated sGC activity threefold, without detectable NO release. IPA-NO also suppressed ET1-induced cardiomyocyte superoxide generation. The pure NO donor diethylamine-NONOate (DEA-NO) reproduced these IPA-NO actions but was sensitive to carboxy-PTIO rather than l-cysteine. Although IPA-NO stimulation of purified sGC was preserved under pyrogallol oxidant stress (in direct contrast to DEA-NO), cardiomyocyte sGC activity after either donor was attenuated by this stress. Excitingly IPA-NO also exhibited acute antihypertrophic actions in response to pressure overload in the intact heart. Together these data strongly suggest that IPA-NO protection against cardiomyocyte hypertrophy is independent of both NO and CGRP but rather utilizes novel HNO activation of cGMP signaling. Thus HNO acutely limits hypertrophy independently of NO, even under conditions of elevated superoxide. Development of longer-acting HNO donors may thus represent an attractive new strategy for the treatment of cardiac hypertrophy, as stand-alone and/or add-on therapy to standard care.


Subject(s)
Cardiomegaly/drug therapy , Cardiovascular Agents/therapeutic use , Cyclic GMP/metabolism , Hydrazines/pharmacology , Myocytes, Cardiac/drug effects , Nitrogen Oxides/metabolism , Second Messenger Systems/drug effects , Animals , Animals, Newborn , Antioxidants/pharmacology , Cardiomegaly/genetics , Cardiomegaly/metabolism , Cardiomegaly/pathology , Cells, Cultured , Cyclic GMP-Dependent Protein Kinase Type I/antagonists & inhibitors , Cyclic GMP-Dependent Protein Kinase Type I/metabolism , Dose-Response Relationship, Drug , Endothelin-1/pharmacology , Enzyme Inhibitors/pharmacology , Gene Expression Regulation , Guanylate Cyclase/antagonists & inhibitors , Guanylate Cyclase/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Nitric Oxide Donors/pharmacology , Pyrogallol/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/metabolism , Soluble Guanylyl Cyclase , Time Factors
19.
Neurosci Lett ; 541: 120-5, 2013 Apr 29.
Article in English | MEDLINE | ID: mdl-23428510

ABSTRACT

Protein kinase G (PKG) has been implicated in a variety of physiological functions including synaptic plasticity in the brain. This study investigated the involvement of dopamine D3 (D3) receptors in PKG-regulated dopamine release, long-term changes in gene expression and behavioral sensitization after repeated cocaine administration. Repeated systemic injections of cocaine (20mg/kg), once a day for seven consecutive days, increased extracellular dopamine concentrations in the dorsal striatum. Inhibition of neuronal nitric oxide synthase, cGMP or PKG, stimulation of D3 receptors, and simultaneous inhibition of each of them with D3 receptor stimulation decreased the repeated cocaine-induced increase in dopamine concentrations and locomotor activity. Similarly, inhibition of PKG and simultaneous inhibition of PKG with D3 receptor stimulation decreased ΔFosB immunoreactivity elevated by repeated cocaine administration, however stimulation of D3 receptors alone did not. These findings suggest that activation of PKG after repeated cocaine administration is more sensitive to interact with D3 receptors in the dopamine terminals than those in medium spiny neurons. This interaction may result in the development of behavioral sensitization by the upregulation of dopamine releases in the dorsal striatum.


Subject(s)
Central Nervous System Stimulants/pharmacology , Cocaine/pharmacology , Corpus Striatum/drug effects , Dopamine/metabolism , Motor Activity/drug effects , Protein Kinase C/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Receptors, Dopamine D3/metabolism , Animals , Biosensing Techniques , Corpus Striatum/metabolism , Cyclic GMP/antagonists & inhibitors , Cyclic GMP-Dependent Protein Kinase Type I/antagonists & inhibitors , Cyclic GMP-Dependent Protein Kinase Type I/metabolism , Enzyme Activation , Male , Neuronal Plasticity/drug effects , Nitric Oxide Synthase Type I/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Receptors, Dopamine D3/agonists , Receptors, Presynaptic/agonists , Receptors, Presynaptic/metabolism , Synapses/drug effects , Synapses/physiology
20.
Int J Cardiol ; 167(5): 2114-9, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-22748498

ABSTRACT

BACKGROUND AND OBJECTIVE: Nitric oxide (NO) and related nitrovasodilators regulate blood pressure by activation of soluble guanylate cyclase, elevation of cyclic guanosine monophosphate (cGMP), and activation of cGMP-dependent protein kinase (cGPK). Despite the progress of our understanding of the NO/cGMP mediated vasorelaxation, partly through conventional cGPK knock-out mice, the role of cGPK remains unclear. In particular, the downstream target(s) of the kinase are not well defined. We hypothesized that highly selective inhibitors of cGPK delivered in vivo in adult may elucidate the role of the kinase in vasorelaxation and regulation of blood pressure. METHODS AND RESULTS: We have adopted a newly developed method of TAT-mediated protein transduction to study NO/cGMP signaling pathways in mice. In vitro, TAT-cGPK inhibitor peptide blocked autophosphorylation of the kinase. The effect of cGPK inhibition on murine blood pressure (BP) was investigated by continuous infusion of 100 µg of the inhibitor into the internal jugular vein over 72 hours. In 8 animals infused with the inhibitor, the mean BP increased by 38 ± 24/31 ± 30 mm Hg (from 108 ± 14/92 ± 19 to 145 ± 13/123 ± 19 mm Hg) whereas in 8 animals injected with either saline (4) or TAT-green fluorescent protein (4), the BP remained the same (from 117 ± 21/101 ± 26 to 119 ± 22/96 ± 30 mm Hg); P=0.001. Ex vivo, using vascular ring assays, NO-dependent relaxation in murine aortas harvested from animals administered with TAT-cGPK inhibitor was inhibited by 25% (sham 76 ± 11%, inhibitor 51 ± 13%). CONCLUSION: We demonstrated that highly specific peptide inhibitor of cGPK induced adult murine hypertension through inhibition of nitric oxide mediated relaxation.


Subject(s)
Cell-Penetrating Peptides/administration & dosage , Cyclic GMP-Dependent Protein Kinase Type I/antagonists & inhibitors , Drug Delivery Systems/methods , Endothelium, Vascular/drug effects , Gene Products, tat/administration & dosage , Hypertension/drug therapy , Amino Acid Sequence , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/enzymology , Base Sequence , Cell-Penetrating Peptides/genetics , Cells, Cultured , Cyclic GMP-Dependent Protein Kinase Type I/metabolism , Endothelium, Vascular/enzymology , Endothelium, Vascular/physiopathology , Female , Gene Products, tat/genetics , Hypertension/enzymology , Hypertension/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Organ Culture Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...