cyclic-gmp has been researched along with 8-bromoguanosino-3--5--cyclic-monophosphorothioate* in 85 studies
85 other study(ies) available for cyclic-gmp and 8-bromoguanosino-3--5--cyclic-monophosphorothioate
Article | Year |
---|---|
Intracellular cGMP increase is not involved in thyroid cancer cell death.
Type 5 phosphodiesterase (PDE5) inhibitors (PDE5i) lead to intracellular cyclic-guanosine monophosphate (cGMP) increase and are used for clinical treatment of erectile dysfunction. Studies found that cGMP may up/downregulate the growth of certain endocrine tumor cells, suggesting that PDE5i could impact cancer risk.. We evaluated if PDE5i may modulate thyroid cancer cell growth in vitro.. We used malignant (K1) and benign (Nthy-ori 3-1) thyroid cell lines, as well as the COS7 cells as a reference model. Cells were treated 0-24 h with the PDE5i vardenafil or the cGMP analog 8-br-cGMP (nM-μM range). cGMP levels and caspase 3 cleavage were evaluated by BRET, in cGMP or caspase 3 biosensor-expressing cells. Phosphorylation of the proliferation-associated extracellularly-regulated kinases 1 and 2 (ERK1/2) was evaluated by Western blotting, while nuclear fragmentation by DAPI staining. Cell viability was investigated using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay.. Both vardenafil and 8-br-cGMP effectively induced dose-dependent cGMP BRET signals (p≤0.05) in all the cell lines. However, no differences in caspase 3 activation occurred comparing PDE5i-treated vs untreated cells, at all concentrations and time-points tested (p>0.05). These results match those obtained upon cell treatment with 8-br-cGMP, which failed in inducing caspase 3 cleavage in all the cell lines (p>0.05). Moreover, they reflect the lack of nuclear fragmentation. Interestingly, the modulation of intracellular cGMP levels with vardenafil or the analog did not impact cell viability of both malignant and benign thyroid tumor cell lines, nor the phosphorylation of ERK1/2 (p>0.05).. This study demonstrates that increased cGMP levels are not linked to cell viability or death in K1 and Nthy-ori 3-1 cell lines, suggesting that PDE5i do not impact the growth of thyroid cancer cells. Since different results were previously published, further investigations are recommended to clarify the impact of PDE5i on thyroid cancer cells. Topics: Caspase 3; Cell Death; Cyclic GMP; Humans; Male; Phosphodiesterase Inhibitors; Piperazines; Sulfones; Thyroid Neoplasms; Vardenafil Dihydrochloride | 2023 |
Nitric oxide impacts bovine sperm capacitation in a cGMP-dependent and cGMP-independent manner.
Topics: Animals; Arginine; Cattle; Cryopreservation; Cyclic GMP; Cyclic GMP-Dependent Protein Kinases; Cyclic N-Oxides; Heparin; Imidazoles; Male; Nitric Oxide; Sperm Capacitation; Sperm Motility; Spermatozoa; Thionucleotides | 2019 |
Expression of membrane progesterone receptors (mPRs) in rat peripheral glial cell membranes and their potential role in the modulation of cell migration and protein expression.
The role played by progestogens in modulating Schwann cell pathophysiology is well established. Progestogens exert their effects in these cells through both classical genomic and non-genomic mechanisms, the latter mediated by the GABA-A receptor. However, there is evidence that other receptors may be involved. Membrane progesterone receptors (mPRs) are novel 7-transmembrane receptors coupled to G proteins that have been characterized in different tissues and cells, including the central nervous system (CNS). The mPRs were shown to mediate some of progestogens' neuroprotective effects in the CNS, and to be upregulated in glial cells after traumatic brain injury. Based on this evidence, this paper investigated the possible involvement of mPRs in mediating progestogen actions in S42 Schwann cells. All five mPR isoforms and progesterone receptor membrane component 1 (PGRMC1) were detected in Schwann cells, and were present on the cell membrane. Progesterone and the mPR-specific agonist, Org-OD-02-0 (02) bound to these membranes, indicating the presence of functional mPRs. The mPR agonist 02 rapidly increased cell migration in an in vitro assay, suggesting a putative role of mPRs in the nerve regeneration process. Treatment with pertussis toxin and 8-Br-cAMP blocked 02-induced cell migration, suggesting this progestogen action is mediated by activation of an inhibitory G protein, leading to a decrease in intracellular cAMP levels. In contrast, long-term mPR activation led to increased expression levels of myelin associated glycoprotein (MAG). Taken together, these findings show that mPRs are present and active in Schwann cells and have a role in modulating their physiological processes. Topics: Animals; Cell Membrane; Cell Movement; Cyclic GMP; Myelin-Associated Glycoprotein; Neuroglia; Pertussis Toxin; Protein Isoforms; Rats; Receptors, Progesterone; Thionucleotides; Tumor Cells, Cultured | 2019 |
Andrographolide inhibits hypoxia-induced hypoxia-inducible factor 1α and endothelin 1 expression through the heme oxygenase 1/CO/cGMP/MKP-5 pathways in EA.hy926 cells.
Topics: Anti-Inflammatory Agents; Carbon Monoxide; Cell Hypoxia; Cell Line; Cyclic GMP; Diterpenes; Dual-Specificity Phosphatases; Endothelin-1; Heme Oxygenase-1; Humans; Hypoxia-Inducible Factor 1, alpha Subunit; Mitogen-Activated Protein Kinase Phosphatases; p38 Mitogen-Activated Protein Kinases; Thionucleotides | 2018 |
Modulation of thalamocortical oscillations by TRIP8b, an auxiliary subunit for HCN channels.
Hyperpolarization-activated cyclic nucleotide-gated cation (HCN) channels have important functions in controlling neuronal excitability and generating rhythmic oscillatory activity. The role of tetratricopeptide repeat-containing Rab8b-interacting protein (TRIP8b) in regulation of hyperpolarization-activated inward current, I Topics: Action Potentials; Adenine; Adenylyl Cyclase Inhibitors; Animals; Cardiovascular Agents; Cerebral Cortex; Cyclic AMP; Cyclic GMP; Female; Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels; Male; Membrane Proteins; Mice; Mice, Inbred C57BL; Mice, Transgenic; Models, Neurological; Neural Pathways; Peroxins; Pyrimidines; Sodium Channel Blockers; Tetrodotoxin; Thalamus; Thionucleotides | 2018 |
Nitric oxide modulates ATP-evoked currents in mouse Leydig cells.
Testosterone synthesis within Leydig cells is a calcium-dependent process. Intracellular calcium levels are regulated by different processes including ATP-activated P2X purinergic receptors, T-type Ca2+ channels modulated by the luteinizing hormone, and intracellular calcium storages recruited by a calcium-induced calcium release mechanism. On the other hand, nitric oxide (NO) is reported to have an inhibitory role in testosterone production. Based on these observations, we investigated the interaction between the purinergic and nitrergic systems in Leydig cells of adult mice. For this purpose, we recorded ATP-evoked currents in isolated Leydig cells using the whole cell patch clamp technique after treatment with L-NAME (300 μM and 1 mM), L-arginine (10, 100, 300, and 500 μM), ODQ (300 μM), and 8-Br-cGMP (100 μM). Our results show that NO produced by Leydig cells in basal conditions is insufficient to change the ATP-evoked currents and that extra NO provided by adding 300 μM L-arginine positively modulates the current through a mechanism involving the NO/cGMP signaling pathway. Thus, we report an interaction between the nitrergic and purinergic systems in Leydig cells and suggest that Ca2+ entry via the purinergic receptors can be regulated by NO. Topics: Action Potentials; Adenosine Triphosphate; Animals; Arginine; Cells, Cultured; Cyclic GMP; Leydig Cells; Male; Mice; NG-Nitroarginine Methyl Ester; Nitric Oxide; Patch-Clamp Techniques; Receptors, Purinergic; Thionucleotides | 2018 |
Inhibition of Cgkii Suppresses Seizure Activity and Hippocampal Excitation by Regulating the Postsynaptic Delivery of Glua1.
The imbalance between excitation and inhibition is a defining feature of epilepsy. GluA1 is an AMPA receptor subunit that can strengthen excitatory synaptic transmission when upregulated in the postsynaptic membrane, which has been implicated in the pathogenesis of epilepsy. cGKII, a cGMP-dependent protein kinase, regulates the GluA1 levels at the plasma membrane.. To explore the role of cGKII in epilepsy, we investigated the expression of cGKII in patients with temporal lobe epilepsy (TLE) and in a pilocarpine-induced rat model and then performed behavioral, histological, and electrophysiological analyses by applying either a cGKII agonist or inhibitor in the hippocampus of the animal model.. cGKII expression was upregulated in the epileptogenic brain tissues of both humans and rats. Pharmacological activation or inhibition of cGKII induced changes in epileptic behaviors in vivo and epileptic discharges in vitro. Further studies indicated that cGKII activation disrupted the balance of excitation and inhibition due to strengthened AMPAR-mediated excitatory synaptic transmission. Moreover, cGKII regulated epileptic seizures by phosphorylating GluA1 at Ser845 to modulate the expression and function of GluA1 in the postsynaptic membrane.. These results suggest that cGKII plays a key role in seizure activity and could be a potential therapeutic target for epilepsy. Topics: 4-Aminopyridine; Adolescent; Adult; Animals; Brain; Carbazoles; Child; Cyclic GMP; Cyclic GMP-Dependent Protein Kinase Type II; Disease Models, Animal; Epilepsy; Evoked Potentials; Female; Hippocampus; Humans; Male; Pilocarpine; Rats; Rats, Sprague-Dawley; Receptors, AMPA; Synaptic Transmission; Thionucleotides; Up-Regulation; Young Adult | 2018 |
Balance between
Nitric oxide (NO) contributes to myogenesis by regulating the transition between myoblast proliferation and fusion through cGMP signaling. NO can form Topics: Aldehyde Oxidoreductases; Animals; Cell Differentiation; Cell Fusion; Chick Embryo; Cyclic AMP; Cyclic GMP; Cysteine; Enzyme Inhibitors; Fibroblasts; Gene Expression Regulation, Developmental; Muscle Development; Muscle Fibers, Skeletal; Myoblasts; Nitric Oxide; Nitric Oxide Synthase Type II; Primary Cell Culture; RNA, Small Interfering; S-Nitrosoglutathione; S-Nitrosothiols; Signal Transduction; Soluble Guanylyl Cyclase; Thionucleotides; Triazenes | 2017 |
Synthesis and Characterization of 8-Nitroguanosine 3',5'-Cyclic Monophosphorothioate Rp-Isomer as a Potent Inhibitor of Protein Kinase G1α.
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. Topics: Acetylcholine; Animals; Aorta; Cyclic GMP; Cyclic GMP-Dependent Protein Kinase Type I; Guanosine; Isomerism; Male; Mice, Inbred C57BL; Nitro Compounds; Protein Processing, Post-Translational; Signal Transduction; Thionucleotides; Vasodilation | 2017 |
Scutellarin attenuates endothelium-dependent aasodilation impairment induced by hypoxia reoxygenation, through regulating the PKG signaling pathway in rat coronary artery.
Scutellarin (SCU), a flavonoid from a traditional Chinese medicinal plant. Our previous study has demonstrated that SCU relaxes mouse aortic arteries mainly in an endothelium-depend-ent manner. In the present study, we investigated the vasoprotective effects of SCU against HR-induced endothelial dysfunction (ED) in isolated rat CA and the possible mechanisms involving cyclic guanosine monophosphate (cGMP) dependent protein kinase (PKG). The isolated endothelium-intact and endothelium-denuded rat CA rings were treated with HR injury. Evaluation of endothelium-dependent and -independent vasodilation relaxation of the CA rings were performed using wire myography and the protein expressions were assayed by Western blotting. SCU (10-1 000 μmol·L(-1)) could relax the endothelium-intact CA rings but not endothelium-denuded ones. In the intact CA rings, the PKG inhibitor, Rp-8-Br-cGMPS (PKGI-rp, 4 μmol·L(-1)), significantly blocked SCU (10-1 000 μmol·L(-1))-induced relaxation. The NO synthase (NOS) inhibitor, NO-nitro-L-arginine methylester (L-NAME, 100 μmol·L(-1)), did not significantly change the effects of SCU (10-1 000 μmol·L(-1)). HR treatment significantly impaired ACh-induced relaxation, which was reversed by pre-incubation with SCU (500 μmol·L(-1)), while HR treatment did not altered NTG-induced vasodilation. PKGI-rp (4 μmol·L(-1)) blocked the protective effects of SCU in HR-treated CA rings. Additionally, HR treatment reduced phosphorylated vasodilator-stimulated phosphoprotein (p-VASP, phosphorylated product of PKG), which was reversed by SCU pre-incubation, suggesting that SCU activated PKG phosphorylation against HR injury. SCU induces CA vasodilation in an endothelium-dependent manner to and repairs HR-induced impairment via activation of PKG signaling pathway. Topics: Animals; Apigenin; Cell Adhesion Molecules; Cell Hypoxia; Coronary Vessels; Cyclic GMP; Cyclic GMP-Dependent Protein Kinases; Glucuronates; Microfilament Proteins; NG-Nitroarginine Methyl Ester; Phosphoproteins; Rats; Rats, Sprague-Dawley; Reperfusion Injury; Signal Transduction; Thionucleotides; Vasodilation | 2015 |
cGMP/Protein Kinase G Signaling Suppresses Inositol 1,4,5-Trisphosphate Receptor Phosphorylation and Promotes Endoplasmic Reticulum Stress in Photoreceptors of Cyclic Nucleotide-gated Channel-deficient Mice.
Photoreceptor cyclic nucleotide-gated (CNG) channels play a pivotal role in phototransduction. Mutations in the cone CNG channel subunits CNGA3 and CNGB3 are associated with achromatopsia and cone dystrophies. We have shown endoplasmic reticulum (ER) stress-associated apoptotic cone death and increased phosphorylation of the ER Ca(2+) channel inositol 1,4,5-trisphosphate receptor 1 (IP3R1) in CNG channel-deficient mice. We also presented a remarkable elevation of cGMP and an increased activity of the cGMP-dependent protein kinase (protein kinase G, PKG) in CNG channel deficiency. This work investigated whether cGMP/PKG signaling regulates ER stress and IP3R1 phosphorylation in CNG channel-deficient cones. Treatment with PKG inhibitor and deletion of guanylate cyclase-1 (GC1), the enzyme producing cGMP in cones, were used to suppress cGMP/PKG signaling in cone-dominant Cnga3(-/-)/Nrl(-/-) mice. We found that treatment with PKG inhibitor or deletion of GC1 effectively reduced apoptotic cone death, increased expression levels of cone proteins, and decreased activation of Müller glial cells. Furthermore, we observed significantly increased phosphorylation of IP3R1 and reduced ER stress. Our findings demonstrate a role of cGMP/PKG signaling in ER stress and ER Ca(2+) channel regulation and provide insights into the mechanism of cone degeneration in CNG channel deficiency. Topics: Animals; Apoptosis; Basic-Leucine Zipper Transcription Factors; Carbazoles; Cyclic GMP; Cyclic GMP-Dependent Protein Kinases; Cyclic Nucleotide-Gated Cation Channels; Endoplasmic Reticulum Stress; Ependymoglial Cells; Eye Proteins; Gene Expression Regulation; Guanylate Cyclase; Inositol 1,4,5-Trisphosphate Receptors; Mice; Mice, Knockout; Phosphorylation; Protein Kinase Inhibitors; Receptors, Cell Surface; Retinal Cone Photoreceptor Cells; Signal Transduction; Thionucleotides | 2015 |
cGMP modulates Arabidopsis lateral root formation through regulation of polar auxin transport.
The phytohormone auxin participates in lateral root formation and primary root growth in plants. The auxin gradient formation is mainly regulated by the direction of polar auxin transport (PAT). PAT requires PIN family proteins, which are auxin transport facilitators and contribute to the establishment and maintenance of auxin gradients and mediate multiple developmental processes. Here, we report the effect of the 3', 5'-cyclic guanosine monophosphate (cGMP), an important second messenger, on postembryonic developmental of Arabidopsis lateral root. We find that enhanced cGMP level through the application of the membrane permeable cGMP analog 8-Br-cGMP, promotes the initiation of lateral root primordia and formation of lateral root. 6-Anilino-5,8-quinolinedione (Ly83583, the guanylate cyclase inhibitor) negatively regulates the process. cGMP also mediates acropetal auxin transport and basipetal auxin transport in the root. We further find that 8-Br-cGMP and Ly83583 change the expression of auxin transport genes and alter the polar localization and expression of PIN1 and PIN2 proteins. Moreover, Ly83583 affects actin organization and localization. Taken together, we propose that cGMP affects auxin transport and auxin gradient through modulation PINs proteins localization and expression. cGMP regulates postembryonic formation of Arabidopsis lateral root through the crosstalk with PAT. Topics: Actin Cytoskeleton; Aminoquinolines; Arabidopsis; Arabidopsis Proteins; Biological Transport; Cell Membrane Permeability; Cyclic GMP; Gene Expression Regulation, Plant; Genes, Plant; Green Fluorescent Proteins; Indoleacetic Acids; Membrane Transport Proteins; Plant Roots; Plants, Genetically Modified; Seedlings; Thionucleotides | 2013 |
Phosphodiesterase-5 inhibitor sildenafil prevents neuroinflammation, lowers beta-amyloid levels and improves cognitive performance in APP/PS1 transgenic mice.
Memory deficit is a marker of Alzheimer's disease (AD) that has been highly associated with the dysfunction of cyclic GMP (cGMP) signaling and an ongoing inflammatory process. Phosphodiesterase-5 (PDE5) inhibitors prevent the breakdown of cGMP and are currently studied as a possible target for cognitive enhancement. However, it is still unknown whether inhibition of PDE5 reversed β-amyloid peptide (Aβ)-induced neuroinflammation in APP/PS1 transgenic (Tg APP/PS1) mice. The present study evaluated the cognitive behaviors, inflammatory mediators, and cGMP/PKG/pCREB signaling in 15-month-old Tg APP/PS1 mice and age-matched wild-type (WT) mice that were treated with PDE5 inhibitor sildenafil and the inhibitor of cGMP-dependent protein kinase Rp-8-Br-PET-cGMPS. In comparison with WT mice, Tg APP/PS1 mice were characterized by impaired cognitive ability, neuroinflammatory response, and down-regulated cGMP signaling. Sildenafil reversed these memory deficits and cGMP/PKG/pCREB signaling dysfunction; it also reduced both the soluble Aβ1-40 and Aβ1-42 levels in the hippocampus. These effects of sildenafil were prevented by intra-hippocampal infusion of the Rp-8-Br-PET-cGMPS. These results suggest that sildenafil could restore cognitive deficits in Tg APP/PS1 mice by the regulation of PKG/pCREB signaling, anti-inflammatory response and reduction of Aβ levels. Topics: Alzheimer Disease; Amyloid beta-Peptides; Amyloid beta-Protein Precursor; Analysis of Variance; Animals; Cognition Disorders; Cyclic GMP; Cytokines; Disease Models, Animal; Dose-Response Relationship, Drug; Encephalitis; Exploratory Behavior; Humans; Mice; Mice, Inbred C57BL; Mice, Transgenic; Mutation; Phosphodiesterase 5 Inhibitors; Piperazines; Presenilin-1; Purines; Recognition, Psychology; RNA, Messenger; Sildenafil Citrate; Sulfones; Thionucleotides | 2013 |
Hydrogen peroxide is involved in cGMP modulating the lateral root development of Arabidopsis thaliana.
3',5'-cyclic guanosine monophosphate (cGMP) and hydrogen peroxide (H₂O₂) function as the important signaling molecule which promote the lateral root development of Arabidopsis thaliana. In this study, interestingly, application of 8-Br-cGMP (the membrane permeable cGMP analog) promoted the endogenous H₂O₂ production. In addition, the decrease of endogenous H₂O₂ also inhibited the effect of cGMP on the lateral root development. Thus, H₂O₂ maybe act as a downstream signaling of cGMP molecule which is involved in the lateral root development of Arabidopsis. We further found that H₂O₂ affected cGMP modulating polar auxin transport. When the endogenous H₂O₂ level was inhibited, the effect of cGMP on the acropetal auxin transport and the basipetal auxin transport was removed. Moreover, pin2 was insensitive for cGMP and H₂O₂ suggesting that PIN2 protein plays an important role in cGMP and H₂O₂ modulating the lateral root development of Arabidopsis. Topics: Arabidopsis; Biological Transport; Cyclic GMP; Hydrogen Peroxide; Plant Roots; Signal Transduction; Thionucleotides | 2013 |
Potent and direct presynaptic modulation of glycinergic transmission in rat spinal neurons by atrial natriuretic peptide.
Atrial and brain natriuretic peptides (ANP and BNP) exist in the central nervous system and modulate neuronal function, although the locus of actions and physiological mechanisms are still unclear. In the present study we used rat spinal sacral dorsal commissural nucleus (SDCN) and hippocampal 'synaptic bouton' preparations, to record both spontaneous and evoked glycinergic inhibitory postsynaptic currents (sIPSCs and eIPSCs) in SDCN neurons, and the evoked excitatory postsynaptic currents (eEPSCs) in hippocampal CA3 neurons. ANP potently and significantly reduced the sIPSC frequency without affecting the amplitude. ANP also potently reduced the eIPSCs amplitude concurrently increasing the failure rate and the paired pulse ratio response. These ANP actions were blocked by anantin, a specific type A natriuretic peptide receptor (NPR-A) antagonist. The results clearly indicate that ANP acts directly on glycinergic presynaptic nerve terminals to inhibit glycine release via presynaptic NPR-A. The ANP effects were not blocked by the membrane permeable cGMP analog (8Br-cGMP) suggesting a transduction mechanisms not simply related to increasing cGMP levels in nerve terminals. BNP did not affect on glycinergic sIPSCs and eIPSCs. Moreover, both ANP and BNP had no effect on glutamatergic EPSCs in hippocampal CA3 neurons. The results indicate a potent and selective presynaptic inhibitory action of ANP on glycinergic transmission in spinal cord sensory circuits. Topics: 4-Aminopyridine; 6-Cyano-7-nitroquinoxaline-2,3-dione; Anesthetics, Local; Animals; Animals, Newborn; Atrial Natriuretic Factor; Cyclic GMP; Excitatory Amino Acid Antagonists; Excitatory Postsynaptic Potentials; Glycine; Hippocampus; Inhibitory Postsynaptic Potentials; Lidocaine; Neurons; Peptides, Cyclic; Potassium Channel Blockers; Presynaptic Terminals; Rats; Rats, Wistar; Spinal Cord; Thionucleotides | 2013 |
Nitric oxide/soluble guanylyl cyclase signaling mediates depolarization-induced protection of rat mesencephalic dopaminergic neurons from MPP⁺ cytotoxicity.
Neuronal electrical activity has been known to affect the viability of neurons in the central nervous system. Here we show that long-lasting membrane depolarization induced by elevated extracellular K(+) recruits nitric oxide (NO)/soluble guanylyl cyclase/protein kinase G signaling pathway, induces 8-nitroguanosine 3',5'-cyclic monophosphate (8-nitro-cGMP)-mediated protein S-guanylation, and confers dopaminergic neuroprotection. Treatment of primary mesencephalic cell cultures with 1-methyl-4-phenylpyridinium (MPP(+)) for 72 h decreased the number of dopaminergic neurons, whereas the cell loss was markedly inhibited by elevated extracellular concentration of K(+) (+40 mM). The neuroprotective effect of elevated extracellular K(+) was significantly attenuated by tetrodotoxin (a Na(+) channel blocker), amlodipine (a voltage-dependent Ca(2+) channel blocker), N(ω)-nitro-l-arginine methyl ester (l-NAME) (a nitric oxide synthase inhibitor), 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ) (a soluble guanylyl cyclase inhibitor), and KT5823 or Rp-8-bromo-β-phenyl-1,N(2)-ethenoguanosine 3',5'-cyclic monophosphorothioate (Rp-8-Br-PET-cGMPS) (protein kinase G inhibitors). Elevated extracellular K(+) increased 8-nitro-cGMP production resulting in the induction of protein S-guanylation in cells in mesencephalic cultures including dopaminergic neurons. In addition, exogenous application of 8-nitro-cGMP protected dopaminergic neurons from MPP(+) cytotoxicity, which was prevented by zinc protoporphyrin IX, an inhibitor of heme oxygenase-1 (HO-1). Zinc protoporphyrin IX also inhibited the neuroprotective effect of elevated extracellular K(+). On the other hand, KT5823 or Rp-8-Br-PET-cGMPS did not inhibit the induction of HO-1 protein expression by 8-nitro-cGMP, although these protein kinase G inhibitors abrogated the neuroprotective effect of 8-nitro-cGMP. These results suggest that protein S-guanylation (leading to HO-1 induction) as well as canonical protein kinase G signaling pathway plays an important role in NO-mediated, activity-dependent dopaminergic neuroprotection. Topics: 1-Methyl-4-phenylpyridinium; Animals; Cyclic GMP; Dopaminergic Neurons; Enzyme Inhibitors; Guanylate Cyclase; Mesencephalon; NG-Nitroarginine Methyl Ester; Nitric Oxide; Nitric Oxide Donors; Rats; Receptors, Cytoplasmic and Nuclear; Signal Transduction; Soluble Guanylyl Cyclase; Thionucleotides | 2013 |
Electrophysiological characterization of Grueneberg ganglion olfactory neurons: spontaneous firing, sodium conductance, and hyperpolarization-activated currents.
Mammals rely on their acute olfactory sense for their survival. The most anterior olfactory subsystem in the nose, the Grueneberg ganglion (GG), plays a role in detecting alarm pheromone, cold, and urinary compounds. GG neurons respond homogeneously to these stimuli with increases in intracellular [Ca(2+)] or transcription of immediate-early genes. In this electrophysiological study, we used patch-clamp techniques to characterize the membrane properties of GG neurons. Our results offer evidence of functional heterogeneity in the GG. GG neurons fire spontaneously and independently in several stable patterns, including phasic and repetitive single-spike modes of discharge. Whole cell recordings demonstrated two distinct voltage-gated fast-inactivating Na(+) currents with different steady-state voltage dependencies and different sensitivities to tetrodotoxin. Hodgkin-Huxley simulations showed that these Na(+) currents confer dual mechanisms of action potential generation and contribute to different firing patterns. Additionally, GG neurons exhibited hyperpolarization-activated inward currents that modulated spontaneous firing in vitro. Thus, in GG neurons, the heterogeneity of firing patterns is linked to the unusual repertoire of ionic currents. The membrane properties described here will aid the interpretation of chemosensory function in the GG. Topics: Action Potentials; Animals; Animals, Newborn; Biophysical Phenomena; Biophysics; Cesium; Chlorides; Computer Simulation; Cyclic GMP; Cyclic Nucleotide-Gated Cation Channels; Electric Stimulation; Ganglia, Sensory; Glial Fibrillary Acidic Protein; Green Fluorescent Proteins; Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels; In Vitro Techniques; Mice; Mice, Transgenic; Models, Neurological; Nerve Growth Factors; Olfactory Receptor Neurons; Patch-Clamp Techniques; Potassium Channels; RNA, Messenger; S100 Calcium Binding Protein beta Subunit; S100 Proteins; Sodium Channel Blockers; Sodium Channels; Tetrodotoxin; Thionucleotides | 2012 |
Single molecule analysis of serotonin transporter regulation using antagonist-conjugated quantum dots reveals restricted, p38 MAPK-dependent mobilization underlying uptake activation.
The presynaptic serotonin (5-HT) transporter (SERT) is targeted by widely prescribed antidepressant medications. Altered SERT expression or regulation has been implicated in multiple neuropsychiatric disorders, including anxiety, depression and autism. Here, we implement a generalizable strategy that exploits antagonist-conjugated quantum dots (Qdots) to monitor, for the first time, single SERT proteins on the surface of serotonergic cells. We document two pools of SERT proteins defined by lateral mobility, one that exhibits relatively free diffusion, and a second, localized to cholesterol and GM1 ganglioside-enriched microdomains, that displays restricted mobility. Receptor-linked signaling pathways that enhance SERT activity mobilize transporters that, nonetheless, remain confined to membrane microdomains. Mobilization of transporters arises from a p38 MAPK-dependent untethering of the SERT C terminus from the juxtamembrane actin cytoskeleton. Our studies establish the utility of ligand-conjugated Qdots for analysis of the behavior of single membrane proteins and reveal a physical basis for signaling-mediated SERT regulation. Topics: Actins; Animals; beta-Cyclodextrins; Cell Line, Transformed; Cholera Toxin; Cholesterol; Cyclic GMP; Cytochalasin D; Enzyme Inhibitors; Gangliosidosis, GM1; Imidazoles; Ligands; Membrane Microdomains; Microscopy, Confocal; Neurons; Normal Distribution; Nucleic Acid Synthesis Inhibitors; p38 Mitogen-Activated Protein Kinases; Protein Transport; Pyridines; Quantum Dots; Rats; Serotonin; Serotonin Plasma Membrane Transport Proteins; Thionucleotides | 2012 |
Interaction between bradykinin and natriuretic peptides via RGS protein activation in HEK-293 cells.
In this study, the interaction of natriuretic peptides (NP) and bradykinin (BK) signaling pathways was identified by measuring membrane potential (V(m)) and intracellular Ca(2+) using the patch-clamp technique and flow cytometry in HEK-293 cells. BK and NP receptor mRNA was identified using RT-PCR. BK (100 nM) depolarized cells activating bradykinin receptor type 2 (B(2)R) and Ca(2+)-dependent Cl(-) channels inhibitable by 5-nitro-2-(3-phenylpropylamino)benzoic acid (NPPB; 10 μM). The BK-induced Ca(2+) signal was blocked by the B(2)R inhibitor HOE 140. [Des-Arg(9)]-bradykinin, an activator of B(1)R, had no effect on intracellular Ca(2+). NP [atrial natriuretic peptide (ANP), brain natriuretic peptide (BNP), C-type natriuretic peptide (CNP), and urodilatin] depolarized HEK-293 cells inhibiting K(+) channels. ANP, urodilatin, BNP [binding to natriuretic peptide receptor (NPR)-A] and 8-bromo-(8-Br)-cGMP inhibited the BK-induced depolarization while CNP (binding to NPR-Bi) failed to do so. The inhibitory effect on BK-triggered depolarization could be reversed by blocking PKG using the specific inhibitor KT 5823. BK-stimulated depolarization as well as Ca(2+) signaling was completely blocked by the phospholipase C (PLC) inhibitor U-73122 (10 nM). The inositol 1,4,5-trisphosphate receptor blocker 2-aminoethoxydiphenyl borate (2-APB; 50 μM) completely inhibited the BK-induced Ca(2+) signaling. UTP, another activator of the PLC-mediated Ca(2+) signaling pathway, was blocked by U-73122 as well but not by 8-Br-cGMP, indicating an intermediate regulatory step for NP via PKG in BK signaling such as regulators of G-protein signaling (RGS) proteins. When RGS proteins were inhibited by CCG-63802 in the presence of BK and 8-Br-cGMP, cells started to depolarize again. In conclusion, as natural antagonists of the B(2)R signaling pathway, NP may also positively interact in pathological conditions caused by BK. Topics: Boron Compounds; Bradykinin; Bradykinin B2 Receptor Antagonists; Carbazoles; Chloride Channels; Cyclic GMP; Estrenes; Flow Cytometry; HEK293 Cells; Humans; Inositol 1,4,5-Trisphosphate Receptors; Membrane Potentials; Natriuretic Peptides; Nitrobenzoates; Patch-Clamp Techniques; Potassium Channel Blockers; Protein Kinase Inhibitors; Pyrrolidinones; RGS Proteins; Signal Transduction; Thionucleotides; Type C Phospholipases | 2012 |
Activation of cGMP-PKG signaling pathway contributes to neuronal hyperexcitability and hyperalgesia after in vivo prolonged compression or in vitro acute dissociation of dorsal root ganglion in rats.
Injury or inflammation affecting sensory neurons in the dorsal root ganglia (DRG) causes hyperexcitability of DRG neurons that can lead to spinal central sensitization and neuropathic pain. Recent studies have indicated that, following chronic compression of DRG (CCD) or acute dissociation of DRG (ADD) treatment, both hyperexcitability of neurons in intact DRG and behaviorally expressed hyperalgesia are maintained by activity in cGMP-PKG signaling pathway. Here, we provide evidence supporting the idea that CCD or ADD treatment activates cGMP-PKA signaling pathway in the DRG neurons. The results showed that CCD or ADD results in increase of levels of cGMP concentration and expression of PKG-I mRNA, as well as PKG-I protein in DRG. CCD or ADD treated-DRG neurons become hyperexcitable and exhibit increased responsiveness to the activators of cGMP-PKG pathway, 8-Br-cGMP and Sp-cGMP. Hyperexcitability of the injured neurons is inhibited by cGMP-PKG pathway inhibitors, ODQ and Rp-8-pCPT-cGMPS. In vivo delivery of Rp-8-pCPT-cGMPS into the compressed ganglion within the intervertebral foramen suppresses CCD-induced thermal hyperalgesia. These findings indicate that the in vivo CCD or in vitro ADD treatment can activate the cGMP-PKG signaling pathway, and that continuing activation of cGMP-PKG pathway is required to maintain DRG neuronal hyperexcitability and/or hyperalgesia after these two dissimilar forms of injury-related stress. Topics: Animals; Cyclic GMP; Cyclic GMP-Dependent Protein Kinases; Ganglia, Spinal; Hyperalgesia; Rats; Rats, Sprague-Dawley; Signal Transduction; Thionucleotides | 2012 |
Cellular mechanisms underlying nitric oxide-induced vasodilation of descending vasa recta.
It has been observed that vasoactivity of explanted descending vasa recta (DVR) is modulated by intrinsic nitric oxide (NO) and superoxide (O(2)(-)) production (Cao C, Edwards A, Sendeski M, Lee-Kwon W, Cui L, Cai CY, Patzak A, Pallone TL. Am J Physiol Renal Physiol 299: F1056-F1064, 2010). To elucidate the cellular mechanisms by which NO, O(2)(-) and hydrogen peroxide (H(2)O(2)) modulate DVR pericyte cytosolic Ca(2+) concentration ([Ca](cyt)) and vasoactivity, we expanded our mathematical model of Ca(2+) signaling in pericytes. We incorporated simulations of the pathways that translate an increase in [Ca](cyt) to the activation of myosin light chain (MLC) kinase and cell contraction, as well as the kinetics of NO and reactive oxygen species formation and their effects on [Ca](cyt) and MLC phosphorylation. The model reproduced experimentally observed trends of DVR vasoactivity that accompany exposure to N(ω)-nitro-L-arginine methyl ester, 8-Br-cGMP, Tempol, and H(2)O(2). Our results suggest that under resting conditions, NO-induced activation of cGMP maintains low levels of [Ca](cyt) and MLC phosphorylation to minimize basal tone. This results from stimulation of Ca(2+) uptake from the cytosol into the SR via SERCA pumps, Ca(2+) efflux into the extracellular space via plasma membrane Ca(2+) pumps, and MLC phosphatase (MLCP) activity. We predict that basal concentrations of O(2)(-) and H(2)O(2) have negligible effects on Ca(2+) signaling and MLC phosphorylation. At concentrations above 1 nM, O(2)(-) is predicted to modulate [Ca(cyt)] and MCLP activity mostly by reducing NO bioavailability. The DVR vasoconstriction that is induced by high concentrations of H(2)O(2) can be explained by H(2)O(2)-mediated downregulation of MLCP and SERCA activity. We conclude that intrinsic generation of NO by the DVR wall may be sufficient to inhibit vasoconstriction by maintaining suppression of MLC phosphorylation. Topics: Animals; Calcium; Computer Simulation; Cyclic GMP; Cyclic N-Oxides; Hydrogen Peroxide; Kidney Medulla; Models, Biological; Myosin-Light-Chain Kinase; Myosin-Light-Chain Phosphatase; NG-Nitroarginine Methyl Ester; Nitric Oxide; Pericytes; Rats; Reactive Oxygen Species; Renal Artery; Sarcoplasmic Reticulum Calcium-Transporting ATPases; Spin Labels; Thionucleotides; Vasodilation | 2011 |
Activity-dependent regulation of surface glucose transporter-3.
Glucose transporter 3 (GLUT3) is the main facilitative glucose transporter in neurons. Glucose provides neurons with a critical energy source for neuronal activity. However, the mechanism by which neuronal activity controls glucose influx via GLUT3 is unknown. We investigated the influence of synaptic stimulation on GLUT3 surface expression and glucose import in primary cultured cortical and hippocampal neurons. Synaptic activity increased surface expression of GLUT3 leading to an elevation of intracellular glucose. The effect was blocked by NMDA receptor (NMDAR) and neuronal nitric oxide synthase (nNOS) inhibition. The Akt inhibitor I (Akt-I) blocked NMDAR-induced GLUT3 surface expression while a nNOS-phosphomimetic mutant (S1412D) enhanced GLUT3 expression at cell surface. These results suggest that NMDAR/Akt-dependent nNOS phosphorylation is coupled to GLUT3 trafficking. We demonstrated that activation of cGMP-dependent protein kinase (cGK) increased the surface expression of GLUT3, which was repressed by Rp-8-pCPT-cGMPS, a potent cell-permeable inhibitor of cGKs. These studies characterize the molecular basis for activity-dependent increases in surface GLUT3 after stimulation of the NMDARs. NMDAR-induced increase in surface GLUT3 represents a novel pathway for control of energy supply during neuronal activity that is critical for maintaining glucose homeostasis during neuronal transmission. Topics: 4-Aminopyridine; Analysis of Variance; Animals; Bicuculline; Biotinylation; Cerebral Cortex; Cyclic GMP; Disks Large Homolog 4 Protein; Dogs; Drug Interactions; Embryo, Mammalian; Enzyme Inhibitors; Excitatory Amino Acid Antagonists; GABA-A Receptor Antagonists; Gene Expression Regulation; Glucose Transporter Type 3; Green Fluorescent Proteins; Hexoses; Hippocampus; Humans; Intracellular Signaling Peptides and Proteins; Membrane Proteins; Neurons; Nitric Oxide; Nitric Oxide Synthase Type I; Potassium Channel Blockers; Protein Transport; Rats; Rats, Sprague-Dawley; Receptors, N-Methyl-D-Aspartate; Serine; Synapses; Synaptosomes; Thionucleotides; Time Factors; Transfection; Valine | 2011 |
cGMP regulates hydrogen peroxide accumulation in calcium-dependent salt resistance pathway in Arabidopsis thaliana roots.
3',5'-cyclic guanosine monophosphate (cGMP) is an important second messenger in plants. In the present study, roles of cGMP in salt resistance in Arabidopsis roots were investigated. Arabidopsis roots were sensitive to 100 mM NaCl treatment, displaying a great increase in electrolyte leakage and Na(+)/K(+) ratio and a decrease in gene expression of the plasma membrane (PM) H(+)-ATPase. However, application of exogenous 8Br-cGMP (an analog of cGMP), H(2)O(2) or CaCl(2) alleviated the NaCl-induced injury by maintaining a lower Na(+)/K(+) ratio and increasing the PM H(+)-ATPase gene expression. In addition, the inhibition of root elongation and seed germination under salt stress was removed by 8Br-cGMP. Further study indicated that 8Br-cGMP-induced higher NADPH levels for PM NADPH oxidase to generate H(2)O(2) by regulating glucose-6-phosphate dehydrogenase (G6PDH) activity. The effect of 8Br-cGMP and H(2)O(2) on ionic homeostasis was abolished when Ca(2+) was eliminated by glycol-bis-(2-amino ethyl ether)-N,N,N',N'-tetraacetic acid (EGTA, a Ca(2+) chelator) in Arabidopsis roots under salt stress. Taken together, cGMP could regulate H(2)O(2) accumulation in salt stress, and Ca(2+) was necessary in the cGMP-mediated signaling pathway. H(2)O(2), as the downstream component of cGMP signaling pathway, stimulated PM H(+)-ATPase gene expression. Thus, ion homeostasis was modulated for salt tolerance. Topics: Arabidopsis; Calcium; Cell Membrane; Cyclic GMP; Germination; Glucosephosphate Dehydrogenase; Homeostasis; Hydrogen Peroxide; NADP; Plant Roots; Potassium; Proton-Translocating ATPases; Salt Tolerance; Seeds; Signal Transduction; Sodium; Sodium Chloride; Stress, Physiological; Thionucleotides | 2011 |
Mechanisms of xylanase-induced nitric oxide and phosphatidic acid production in tomato cells.
The second messenger nitric oxide (NO), phosphatidic acid (PA) and reactive oxygen species (ROS) are involved in the plant defense response during plant-pathogen interactions. NO has been shown to participate in PA production in response to the pathogen-associated molecular pattern xylanase in tomato cell suspensions. Defense responses downstream of PA include ROS production. The goal of this work was to study the signaling mechanisms involved in PA production during the defense responses triggered by xylanase and mediated by NO in the suspension-cultured tomato cells. We analyzed the participation of protein kinases, guanylate cyclase and the NO-mediated posttranslational modification S-nitrosylation, by means of pharmacology and biochemistry. We showed that NO, PA and ROS levels are significantly diminished by treatment with the general protein kinase inhibitor staurosporine. This indicates that xylanase-induced protein phosphorylation events might be the important components leading to NO formation, and hence for the downstream regulation of PA and ROS levels. When assayed, a guanylate cyclase inhibitor or a cGMP analog did not alter the PA accumulation. These results suggest that a cGMP-mediated pathway is not involved in xylanase-induced PA formation. Finally, the inhibition of protein S-nitrosylation did not affect NO formation but compromised PA and ROS production. Data collectively indicate that upon xylanase perception, cells activate a protein kinase pathway required for NO formation and that, S-nitrosylation-dependent mechanisms are involved in downstream signaling leading to PA and ROS. Topics: Cell Culture Techniques; Cyclic GMP; Enzyme Inhibitors; Gene Expression Regulation, Developmental; Gene Expression Regulation, Plant; Guanylate Cyclase; Host-Pathogen Interactions; Nitric Oxide; Phosphatidic Acids; Phosphorylation; Plant Immunity; Protein Kinases; Protein Processing, Post-Translational; Reactive Oxygen Species; Signal Transduction; Solanum lycopersicum; Staurosporine; Thionucleotides; Xylosidases | 2011 |
Investigating the role of protein kinase-G in the antidepressant-like response of sildenafil in combination with muscarinic acetylcholine receptor antagonism.
The cGMP/PK-G pathway plays a crucial role in neuroprotection and neurotrophin support, and is possibly involved in antidepressant action. Recently we reported on a novel antidepressant-like response following simultaneous administration of sildenafil (phosphodiesterase 5 (PDE5) inhibitor, thereby increasing cGMP levels), and atropine (muscarinic acetylcholine receptor antagonist) in the rat forced swim test (FST). However, it is unclear whether the antidepressant-like activity of sildenafil+atropine is mediated via the activation of PK-G, an important down-stream effector for cGMP, and whether this may target known pathways in antidepressant action. We investigated whether the antidepressant-like response of sildenafil+/-atropine could be reversed by Rp-8-Br-PET-cGMP, a PK-G inhibitor, and also whether a combination of 8-Br-cGMP (PK-G activator)+/-atropine would likewise be active in the FST, and whether this combination could be attenuated by a PK-G inhibitor. 8-Br-cGMP alone, but not sildenafil alone, reduced immobility and selectively increased swimming in the FST. The antidepressant-like action of sildenafil was only evident following co-administration of atropine, and selectively increased climbing behaviour. Importantly, PK-G inhibition prevented the antidepressant-like effects of both 8-Br-cGMP and the sildenafil/atropine combination. These results confirm cholinergic-cGMP-PK-G interactions in the antidepressant-like effects of sildenafil, putatively acting via noradrenergic mechanisms, whereas direct PK-G activation induces antidepressant-like effects that are associated with enhancement of serotonergic neurotransmission. Topics: Animals; Atropine; Behavior, Animal; Cyclic GMP; Cyclic GMP-Dependent Protein Kinases; Depression; Disease Models, Animal; Drug Administration Routes; Drug Interactions; Drug Therapy, Combination; Freezing Reaction, Cataleptic; Male; Muscarinic Antagonists; Phosphodiesterase Inhibitors; Piperazines; Purines; Rats; Rats, Sprague-Dawley; Sildenafil Citrate; Statistics, Nonparametric; Sulfones; Swimming; Thionucleotides | 2010 |
A G-protein-coupled neuropeptide Y-like receptor suppresses behavioral and sensory response to multiple stressful stimuli in Drosophila.
Recent studies suggest that human neuropeptide Y (NPY) plays a prominent role in management of stress response and emotion, and higher NPY levels observed in combat-exposed veterans may help coping with posttraumatic stress. Neuropeptide F (NPF), the counterpart of NPY in Drosophila melanogaster, also displays parallel activities, including promotion of resilience to diverse stressors and prevention of uncontrolled aggressive behavior. However, it remains unclear how NPY family peptides modulate physical and emotional responses to various stressors. Here we show that NPFR1, a G-protein-coupled NPF receptor, exerts an inhibitory effect on larval aversion to diverse stressful stimuli mediated by different subtypes of fly and mammalian transient receptor potential (TRP) family channels. Imaging analysis in larval sensory neurons and cultured human cells showed that NPFR1 attenuates Ca(2+) influx mediated by fly TRPA and rat TRPV1 channels. Our findings suggest that suppression of TRP channel-mediated neural excitation by the conserved NPF/NPFR1 system may be a major mechanism for attaining its broad anti-stress function. Topics: 8-Bromo Cyclic Adenosine Monophosphate; Animals; Animals, Genetically Modified; Behavior, Animal; Calcium; Capsaicin; Carbohydrates; Cell Line, Transformed; Cyclic GMP; Drosophila melanogaster; Drosophila Proteins; Feeding Behavior; Gene Expression Regulation, Developmental; Humans; Larva; Locomotion; Luminescent Proteins; Neurons; Neuropeptides; Pain Measurement; Physical Stimulation; Receptors, Neuropeptide; Receptors, Neuropeptide Y; Sensation; Sensory System Agents; Signal Transduction; Social Behavior; Stress, Physiological; Thionucleotides; Time Factors; Transfection; TRPV Cation Channels | 2010 |
Modulation of cardiac ERG1 K(+) channels by cGMP signaling.
Different K(+) currents have been implicated in the myocardial action potential repolarization including the I(Kr). ERG1 alpha subunits, identified as the molecular correlate of I(Kr), have been shown to form heteromultimeric channels in the heart and their activity is modulated by a complex interplay of signal transduction events. Using electrophysiological techniques, we examined the effects of the cGMP-analogue 8-Br-cGMP on rat and guinea-pig papillary action potential duration (APD), on the biophysical properties of heterologously expressed homo- and heteromeric ERG1 channels, and on cardiac I(Kr). 8-Br-cGMP prolonged APD by about 25% after pharmacological inhibition of L-type Ca(2+) currents and I(Ks). The prolongation was completely abolished by prior application of the hERG channel blocker E-4031 or the protein kinase G (PKG) inhibitor Rp-8-Br-cGMPS. Expression analysis revealed the presence of both ERG1a and -1b subunits in rat papillary muscle. Both 8-Br-cGMP and ANP inhibited heterologously expressed ERG1b and even stronger ERG1a/1b channels, whereas ERG1a channels remained unaffected. The inhibitory 8-Br-cGMP effects were PKG-dependent and involved a profound ERG current reduction, which was also observed with cardiac AP clamp recordings. Measurements of I(Kr) from isolated mouse cardiomyocytes using Cs(+) as charge carrier exhibited faster deactivation kinetics in atrial than in ventricular myocytes consistent with a higher relative expression of ERG1b transcripts in atria than in ventricles. 8-Br-cGMP significantly reduced I(Kr) in atrial, but not in ventricular myocytes. These findings provide first evidence that through heteromeric assembly ERG1 channels become a critical target of cGMP-PKG signaling linking cGMP accumulation to cardiac I(Kr) modulation. Topics: Action Potentials; Animals; Cyclic GMP; Cyclic GMP-Dependent Protein Kinase Type I; Cyclic GMP-Dependent Protein Kinases; Female; Guinea Pigs; Heart Ventricles; Male; Mice; Mice, Inbred C57BL; Myocardium; Myocytes, Cardiac; Piperidines; Pyridines; Rats; Rats, Wistar; Signal Transduction; Thionucleotides; Trans-Activators; Transcriptional Regulator ERG | 2010 |
Nitric oxide and cGMP signal transduction positively regulates the motility of human neuronal precursor (NT2) cells.
Developmental studies in both vertebrates and invertebrates implicate an involvement of nitric oxide (NO) signaling in cell proliferation, neuronal motility, and synaptic maturation. However, it is unknown whether NO plays a role in the development of the human nervous system. We used a model of human neuronal precursor cells from a well-characterized teratocarcinoma cell line (NT2). The precursor cells proliferate during retinoic acid treatment as spherical aggregate culture that stains for nestin and betaIII-tubulin. Cells migrate out of the aggregates to acquire fully differentiated neuronal phenotypes. The cells express neuronal nitric oxide synthase and soluble guanylyl cyclase (sGC), an enzyme that synthesizes cGMP upon activation by NO. The migration of the neuronal precursor cell is blocked by the use of nNOS, sGC, and protein kinase G (PKG) inhibitors. Inhibition of sGC can be rescued by a membrane permeable analog of cGMP. In gain of function experiments the application of a NO donor and cGMP analog facilitate cell migration. Our results from the differentiating NT2 model neurons point towards a vital role of the NO/cGMP/PKG signaling cascade as positive regulator of cell migration in the developing human brain. Topics: Cell Differentiation; Cell Line, Tumor; Cell Movement; Cell Proliferation; Cell Survival; Cyclic GMP; Dose-Response Relationship, Drug; Enzyme Inhibitors; Gene Expression Regulation; Humans; Models, Biological; Neoplastic Stem Cells; Neurons; Nitric Oxide; Nitric Oxide Donors; Nitric Oxide Synthase Type I; Nitroso Compounds; Signal Transduction; Thionucleotides; Time Factors; Tretinoin; Tubulin | 2009 |
ANP stimulates hepatocyte Ca2+ efflux via plasma membrane recruitment of PKGIalpha.
In rat hepatocytes, atrial natriuretic peptide (ANP) elevates cGMP through activation of particulate guanylyl cyclase and attenuates Ca(2+) signals by stimulating net plasma membrane Ca(2+) efflux. We show here that ANP-stimulated hepatocyte Ca(2+) efflux is mediated by protein kinase G (PKG) isotype I. Furthermore, we show that ANP recruits endogenous PKGIalpha, but not PKGIbeta, to the plasma membrane. These effects are mimicked by 8-bromo-cGMP, but not by the soluble guanylyl cyclase activators, sodium nitroprusside and YC-1. We propose that ANP, through localized cGMP elevation, promotes plasma membrane recruitment of PKGIalpha, which, in turn, stimulates Ca(2+) efflux. Topics: Animals; Atrial Natriuretic Factor; Calcium; Cell Membrane; Cyclic GMP; Cyclic GMP-Dependent Protein Kinase Type I; Cyclic GMP-Dependent Protein Kinases; Hepatocytes; Indazoles; Male; Nitroprusside; Rats; Rats, Wistar; Thionucleotides | 2008 |
Stimulation of histamine H1 receptor up-regulates histamine H1 receptor itself through activation of receptor gene transcription.
Histamine is a major mediator in allergy acting mainly through the histamine H(1) receptor (H1R). Although H1R up-regulation has been suggested as an important step for induction of allergic symptoms, little is known about the regulation of H1R level. Here we report that the activation of H1R up-regulates H1R through augmentation of H1R mRNA expression in HeLa cells. Histamine stimulation significantly increased both H1R promoter activity and mRNA level without alteration in mRNA stability. H1R protein was also up-regulated by histamine. An H1R antagonist but not histamine H(2) receptor antagonist blocked histamine-induced up-regulation of both promoter activity and mRNA expression. A protein kinase C (PKC) activator, phorbol-12-myristate-13-acetate, increased H1R mRNA expression, whereas an activator of PKA or PKG (8-Br-cAMP or 8-Br-cGMP, respectively) did not. Furthermore, histamine-induced up-regulation of both promoter activity and mRNA level were completely suppressed by the PKC inhibitor Ro-31-8220. H1R antagonists have long been thought to block H1R and inhibit immediate allergy symptoms. In addition to this short-term effect, our data propose their long-term inhibitory effect against allergic diseases by suppressing PKC-mediated H1R gene transcription. This finding provides new insights into the therapeutic target of H1R antagonist in allergic diseases. Topics: 8-Bromo Cyclic Adenosine Monophosphate; Carbazoles; Cyclic GMP; Enzyme Inhibitors; HeLa Cells; Histamine; Histamine H1 Antagonists; Humans; Indoles; Luciferases; Protein Kinase C; Pyrilamine; Pyrroles; Receptors, Histamine H1; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; Tetradecanoylphorbol Acetate; Thionucleotides; Transcription, Genetic; Transfection; Tritium; Up-Regulation | 2007 |
Acute hypoxia induces vasodilation and increases coronary blood flow by activating inward rectifier K(+) channels.
We examined the effects of acute hypoxia on vascular tone and coronary blood flow (CBF) in rabbit coronary arteries. In the pressurized arterial preparation of small arteries (<100 mum) and the Langendorff-perfused rabbit hearts, hypoxia induced coronary vasodilation and increased CBF in the presence of glibenclamide (K(ATP) channel blocker), Rp-8-Br-PET-cGMPs [cyclic guanosine monophosphate (cGMP)-dependent protein kinase inhibitor, Rp-cGMPs], and methionyl transfer RNA synthetase (MRS) 1334 (adenosine A(3) receptor inhibitor); these increases were inhibited by the inward rectifier K(+) (Kir) channel inhibitor, Ba(2+). These effects were blocked by the adenylyl cyclase inhibitor SQ 22536 and by the cyclic adenosine monophosphate (cAMP)-dependent protein kinase (PKA) inhibitors Rp-8-CPT-cAMPs (Rp-cAMPs) and KT 5720. However, cGMP-dependent protein kinase was not involved in the hypoxia-induced increases of the vascular diameter and CBF. In summary, our results suggest that acute hypoxia can induce the opening of Kir channels in coronary artery that has small diameter (<100 mum) by activating the cAMP and PKA signalling pathway, which could contribute to vasodilation and, therefore, increased CBF. Topics: Acute Disease; Adenine; Animals; Blood Pressure; Carbazoles; Coronary Circulation; Cyclic AMP-Dependent Protein Kinases; Cyclic GMP; Cyclic GMP-Dependent Protein Kinases; Enzyme Inhibitors; Female; Glyburide; Hypoxia; In Vitro Techniques; Indoles; Male; Potassium Channel Blockers; Potassium Channels, Inwardly Rectifying; Pyrroles; Rabbits; Signal Transduction; Thionucleotides; Vasodilation | 2007 |
Serotonin transporter phosphorylation by cGMP-dependent protein kinase is altered by a mutation associated with obsessive compulsive disorder.
Human serotonin transporter (hSERT) activity expressed in HeLa cells was stimulated by agents that release nitric oxide, stimulate soluble guanylyl cyclase, or activate cGMP-dependent protein kinase (PKG). This stimulation was blocked by a PKG inhibitor. A naturally occurring mutation, I425V, associated with obsessive-compulsive disorder and other neuropsychiatric disorders, activated hSERT and eliminated stimulation via the PKG pathway. Inhibitors of soluble guanylyl cyclase or PKG decreased activity of the I425V mutant, but not wild type, indicating that both wild-type and mutant transporters could exist in both high and low activity forms. Mutation of Thr-276 in the fifth transmembrane domain (TM5) to alanine or aspartate prevented activation of wild-type hSERT through the PKG pathway and also blocked the inhibition of I425V activity by inhibitors of the pathway. The accessibility of positions in TM5 near Thr-276 was modified in T276D, but not in I425V. These results are consistent with the hypothesis that PKG phosphorylates hSERT at Thr-276 and increases its activity by modifying the substrate permeation pathway formed, in part, by TM5. The effect of the I425V mutation may shift the balance of hSERT toward the phosphorylated form, possibly by interfering with the action of a phosphatase. However, association of hSERT with protein phosphatase 2A was not decreased in the I425V mutant. Topics: Biotinylation; Cell Adhesion Molecules; Cyclic GMP; Cyclic GMP-Dependent Protein Kinases; Dose-Response Relationship, Drug; Drug Interactions; Enzyme Activation; Enzyme Inhibitors; HeLa Cells; Humans; Immunoprecipitation; Microfilament Proteins; Mutagenesis, Site-Directed; Mutation; Nitric Oxide Donors; Penicillamine; Phosphoproteins; Phosphorylation; Protein Transport; Serotonin; Serotonin Plasma Membrane Transport Proteins; Thionucleotides | 2007 |
Regulation of spontaneous contractile activity in the bovine epididymal duct by cyclic guanosine 5'-monophosphate-dependent pathways.
Passage of spermatozoa through the epididymis is obligatory for sperm maturation processes and is based on spontaneous phasic contractions (SC) of the epididymal duct. Here, the functional role of cyclic GMP (cGMP) signaling in modulating SC in the bovine epididymal caput and corpus region was examined by muscle tension recording and immunological and autoradiographic techniques. The cGMP-analog 8-bromo (Br)-cGMP, as well as the nitric oxide (NO) donor sodium nitroprusside and the natriuretic peptides (NPs) atrial NP and C-type NP, displayed distally increasing SC-relaxant effects. In agreement, a distally increasing epididymal expression of the cGMP-dependent protein kinase I (PKG I), endothelial NO synthase (eNOS), and the atrial NP receptor was found. Immunoreactivity for PKG, soluble guanylate cyclase, and eNOS could be localized to the epididymal muscle cells as well as to the epithelial basal cells only at the corpus level. The SC-relevant action of NO and the NPs was cGMP dependent, and the action of 8-Br-cGMP, in turn, was modified by epithelial and luminal factors. The NOS inhibitor L-NAME (N(omega)-nitro-L-arginine methyl ester) caused an increase in SC frequency, indicating basal activity of NO generating enzymes. The SC-inhibitory effect of 8-Br-cGMP was clearly reduced by the PKG inhibitor Rp-8-Br-cGMPS as well as by iberiotoxin, thapsigargin, and indomethacin, pointing to PKG as main SC-relevant target of cGMP, and to large-conductance calcium-activated K(+) channels, the sarcoplasmic-endoplasmic reticulum Ca(2+)-ATPase and cyclooxygenase-1 as possible targets of PKG. These data support an essential role of cGMP signaling in the control of epididymal peristalsis, thereby enabling fine tuning of sperm transport and maturation. Topics: Animals; Cattle; Cyclic GMP; Cyclic GMP-Dependent Protein Kinases; Epididymis; Guanylate Cyclase; Male; Muscle Contraction; Natriuretic Peptide, C-Type; Nitric Oxide; Nitroprusside; Signal Transduction; Thionucleotides | 2006 |
Nitric oxide donor-induced persistent inhibition of cell adhesion protein expression and NFkappaB activation in endothelial cells.
Nitric oxide (NO), applied by inhalation or released from NO donors, has been used to reduce the expression of cell adhesion molecules (CAM) and ameliorate other consequences of ischemia/reperfusion (I/R) injury. In this study, we have assessed the time frames of pretreatment and of the duration of the preconditioned state using human umbilical vein endothelial cells (HUVECs) and the NO donor, SNAP, in combination with cysteine. The induction of vascular cell adhesion molecule (VCAM), intercellular adhesion molecule (ICAM) and E-selectin by the cytokines TNFalpha and IL-1beta, and by bacterial lipopolysaccharide (LPS) was reduced by SNAP/Cys preincubation (30 min, 1mM) to less than 10% of controls. This refractory state in respect to cytokine-induced CAM expression persisted for 6h after washout of the NO donor in the combination TNFalpha/VCAM, and a partial block was still observed after 8h. The effect was not mediated by the cGMP pathway, as was demonstrated by using the inhibitor of guanylyl cyclase, ODQ, and the cGMP analogue, 8-Br-cGMP. The TNFalpha-induced expression of CAM was exclusively dependent on the transcription factor NFkappaB since the inhibitor of NFkappaB activation, BAY 11-7082, completely blocked the induction. The TNFalpha-induced phosphorylation and degradation of the inhibitor of kappaB (IkappaBalpha) was suppressed for up to 8h after SNAP/Cys pretreatment. The inhibitory S-nitrosation of IkappaB kinase (IKKbeta), as assessed by the biotin-switch-procedure and immunoprecipitation, was only detectable immediately after SNAP/Cys incubation but not at later time points. In summary, a short preincubation of HUVEC with SNAP/Cys results in a persistent suppression of NFkappaB-dependent expression of CAM. The stabilization of IkappaBalpha over the same time span may be causally related to this effect. Topics: Cell Adhesion Molecules; Cells, Cultured; Cyclic GMP; Cysteine; Endothelial Cells; Guanylate Cyclase; Humans; I-kappa B Kinase; I-kappa B Proteins; Interleukin-1; Lipopolysaccharides; NF-kappa B; NF-KappaB Inhibitor alpha; Nitric Oxide Donors; Oxadiazoles; Quinoxalines; S-Nitroso-N-Acetylpenicillamine; Thionucleotides; Tumor Necrosis Factor-alpha; Vascular Cell Adhesion Molecule-1 | 2006 |
Nitric oxide potentiates cAMP-gated cation current in feeding neurons of Pleurobranchaea californica independent of cAMP and cGMP signaling pathways.
Critical roles for nitric oxide (NO) in regulating cell and tissue physiology are broadly appreciated, but aspects remain to be explored. In the mollusk Pleurobranchaea, NO synthase activity is high in CNS ganglia containing motor networks for feeding and locomotion, where a cAMP-gated cation current (I(Na,cAMP)) is also prominent in many neurons. We examined effects of NO on I(Na,cAMP) using voltage-clamp methods developed to analyze cAMP signaling in the live neuron, focusing on the identified metacerebral giant neuron of the feeding network. NO donors enhanced the I(Na,cAMP) response to injected cAMP by an averaged 85%. In dose-response measures, NO increased the current stimulated by cAMP injection without altering either apparent cAMP binding affinity or cooperativity of current activation. NO did not detectably alter levels of native cAMP or synthesis or degradation rates as observable in both current saturation and decay rate of I(Na,cAMP) responses to cAMP injection. NO actions were not exerted by cGMP signaling, as they were not mimicked by cGMP analogue nor blocked by inhibitors of guanylate cyclase and protein kinase G. NO potentiation of I(Na,cAMP) was broadly distributed among many other neurons of the feeding motor network in the buccal ganglion. However, NO did not affect a second type of I(Na,cAMP) found in locomotor neurons of the pedal ganglia. These results suggest that NO acts through a novel mechanism to regulate the gain of cAMP-dependent neuromodulatory pathways that activate I(Na,cAMP) and may thereby affect the set points of feeding network excitability and reactivity to exogenous input. Topics: Animals; Carbazoles; Cyclic GMP; Diethylamines; Dose-Response Relationship, Radiation; Drug Interactions; Electric Stimulation; Enzyme Inhibitors; Feeding Behavior; Ganglia, Invertebrate; In Vitro Techniques; Indoles; Ion Channel Gating; Membrane Potentials; Neurons; Nitric Oxide; Oxadiazoles; Patch-Clamp Techniques; Pleurobranchaea; Signal Transduction; Thionucleotides; Time Factors | 2006 |
Induction of glial fibrillary acidic protein expression in astrocytes by nitric oxide.
Increased expression of glial fibrillary acidic protein (GFAP) represents astroglial activation and gliosis during neurodegeneration. However, the molecular mechanism behind increased expression of GFAP in astrocytes is poorly understood. The present study was undertaken to explore the role of nitric oxide (NO) in the expression of GFAP. Bacterial lipopolysachharides (LPSs) induced the production of NO and the expression of GFAP in mouse primary astrocytes. Either a scavenger of NO [2-(4-carboxyphenyl)-4,4,5,5-tetramethylimidazoline-1-oxyl-3-oxide (PTIO)] or an inhibitor of inducible nitric oxide synthase [l-N6-(I-iminoethyl)-lysine hydrochloride] blocked this induction of GFAP expression. Similarly, other inducers of NO production such as interferon-gamma, interleukin-1beta, human immunodeficiency virus type 1 gp120, fibrillar amyloid beta peptides, and double-stranded RNA (polyinosinic-polycytidilic acid) also induced the expression of GFAP through NO. The role of NO in the expression of GFAP was supported further by increased expression of GFAP by S-nitroso glutathione (GSNO), an NO donor. Interestingly, inhibition of nuclear factor kappaB (NF-kappaB) suppressed LPS- but not GSNO-induced expression of GFAP, suggesting that NO does not require NF-kappaB to induce GFAP and that NF-kappaB functions upstream of NO production. However, inhibition of LPS- and GSNO-induced expression of GFAP either by NS-2028 [a specific inhibitor of guanylate cyclase (GC)] or by KT5823 [a specific inhibitor of cGMP-activated protein kinase (PKG)], and induction of GFAP expression by either 8-Br cGMP (a cell-permeable cGMP analog) or MY-5445 (a specific inhibitor of cGMP phosphodiesterase) suggests that NO induces GFAP via GC-cGMP-PKG. This study illustrates a novel biological role of NO in regulating the expression of GFAP in astrocytes through the GC-cGMP-PKG pathway that may participate in the pathogenesis of neurodegenerative disorders. Topics: Animals; Animals, Newborn; Astrocytes; Cell Survival; Cells, Cultured; Cerebral Cortex; Corpus Striatum; Cyclic GMP; Cyclic N-Oxides; Cytokines; Dose-Response Relationship, Drug; Drug Interactions; Electrophoretic Mobility Shift Assay; Enzyme Inhibitors; Fluorescent Antibody Technique; Free Radical Scavengers; Gene Expression; Glial Fibrillary Acidic Protein; HIV Envelope Protein gp120; Imidazoles; Lipopolysaccharides; Lysine; Male; Mice; Mice, Inbred C57BL; Nitric Oxide; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; Tetrazolium Salts; Thiazoles; Thionucleotides; Time Factors | 2006 |
Cigarette smoke exposure impairs VEGF-induced endothelial cell migration: role of NO and reactive oxygen species.
Endothelial dysfunction is one of the earliest pathological effects of cigarette smoking. Vascular endothelial growth factor (VEGF) has been shown to be an important regulator of endothelial healing and growth. Accordingly, we tested the hypothesis that cigarette smoke exposure impairs VEGF actions in endothelial cells. In human umbilical vein endothelial cells (HUVECs), cigarette smoke extracts (CSE) inhibited VEGF-induced tube formation in the matrigel assay. CSE did not affect HUVECs proliferation, but significantly reduced cellular migration in response to VEGF. This impaired migratory activity was associated with a reduced expression of alpha(v)beta(3), alpha(v)beta(5), alpha(5)beta(1) and alpha(2)beta(1) integrins. The Akt/eNOS/NO pathway has been shown to be important for VEGF-induced endothelial cell migration. We found that CSE inhibited Akt/eNOS phosphorylation and NO release in VEGF-stimulated HUVECs. This was associated with an increased generation of reactive oxygen species (ROS). Importantly, in HUVECs exposed to CSE, treatment with antioxidants (NAC, vitamin C) reduced ROS formation and rescued VEGF-induced NO release, cellular migration and tube formation. Moreover, treatment with NO donors (SNAP, SNP) or a cGMP analog (8-Br-cGMP) rescued integrin expression, cellular migration and tube formation in endothelial cells exposed to CSE. (1) Cigarette smoke exposure impairs VEGF-induced endothelial cell migration and tube formation. (2) The mechanism involves increased generation of ROS, decreased expression of surface integrins together with a blockade of the Akt/eNOS/NO pathway. (3) These findings could contribute to explain the negative effect of cigarette smoking on endothelial function and vessel growth. Topics: Antioxidants; Cardiovascular Diseases; Cell Movement; Cells, Cultured; Cyclic GMP; Drug Interactions; Endothelial Cells; Humans; Integrins; Neovascularization, Physiologic; Nicotiana; Nitric Oxide; Nitric Oxide Donors; Nitric Oxide Synthase Type III; Reactive Oxygen Species; Risk Factors; S-Nitroso-N-Acetylpenicillamine; Signal Transduction; Smoke; Smoking; Thionucleotides; Vascular Endothelial Growth Factor A | 2006 |
Impaired cerebrovascular reactivity after cortical spreading depression in rats: Restoration by nitric oxide or cGMP.
We investigated the role of the NO/cGMP system in the vasodilatory response to hypercapnia after cortical spreading depression (CSD) in barbiturate anesthetized rats in vivo. Regional cerebral blood flow (rCBF) was measured by laser Doppler flowmetry (LDF). Hypercapnia (arterial pCO2 50-60 mm Hg) increased rCBF by 2.8+/-1.0%/mm Hg (n = 34). Fifteen minutes after CSD, resting rCBF was reduced to 87%, and rCBF response to hypercapnia was abolished (p < 0.001, n = 28). Within 1 h after CSD, only little restoration of vascular reactivity occurred. Topical application of the NO-donors S-nitroso-N-acetylpenicillamine (SNAP), 3-morpholinosydnonimine (SIN1), or spermine/NO complex (Sperm/NO), or of the cell permeable guanosine 3',5'-cyclic monophosphate (cGMP) analogue 8-Br-cGMP reestablished resting rCBF to values measured before CSD, and reversed CSD-induced attenuation of the cerebrovascular response to hypercapnia. Restoration of resting rCBF to pre-CSD level by the NO-independent vasodilator papaverine had no effect on the attenuated hypercapnic response. In conclusion, we have shown that the compromised vascular reactivity to hypercapnia after CSD can be reversed to normal reactivity by restoration of the basal NO or cGMP concentration in the cortex, suggesting a reduction of the cerebrovascular NO or cGMP concentration following CSD. Topics: Analysis of Variance; Animals; Cerebral Cortex; Cerebrovascular Circulation; Cerebrovascular Disorders; Cortical Spreading Depression; Cyclic GMP; Drug Interactions; Hypercapnia; Laser-Doppler Flowmetry; Male; Molsidomine; Nitric Oxide; Nitric Oxide Donors; Penicillamine; Rats; Rats, Wistar; Spermine; Thionucleotides; Time Factors | 2006 |
Puerarin, an isoflavonoid derived from Radix puerariae, potentiates endothelium-independent relaxation via the cyclic AMP pathway in porcine coronary artery.
Puerarin, an isoflavonoid derived from the Chinese medicinal herb Radix puerariae, has been suggested to be useful in the management of various cardiovascular disorders. The present study examined the effect of acute exposure (30 min) to puerarin on vascular relaxation. Rings from porcine coronary artery of either sex were used. The highest concentration of puerarin (100 microM) produced a small but statistically significant relaxation of U46619-contracted rings. Vascular relaxations were also studied in the presence of lower concentrations of puerarin (0.1, 1 and 10 microM) which had no direct relaxation effect. Puerarin enhanced vasorelaxation to endothelium-independent relaxing agents, sodium nitroprusside and cromakalim. However, puerarin had no effect on vasorelaxation induced by endothelium-dependent relaxing agents, bradykinin and calcium ionophore A23187. The potentiating action of puerarin (10 microM) on sodium nitroprusside-mediated relaxation was not affected by the nitric oxide synthase inhibitor, N(omega)-nitro-L-arginine methyl ester (L-NAME; 300 microM), or by the disruption of the endothelium with Triton X-100. The effect of puerarin was reversible following a washout period. The potentiating effects were comparable with the 3'-5'-cyclic adenosine monophosphate (cyclic AMP) analogues, 8-bromoadenosine-3'-5'-cyclic monophosphate (8-Br-cyclic AMP; 10 muM) and Sp-isomer [S nomenclature refers to phosphorus] of adenosine-3', 5'-cyclic monophosphorothioate (Sp-cyclic AMPS; 3 microM), but not the 3'-5'-cyclic guanosine monophosphate (cyclic GMP) analogue, 8-bromoguanosine-3'-5'-cyclic monophosphate (8-Br-cyclic GMP; 3 microM). The cyclic AMP antagonist, Rp-isomer [R nomenclature refers to phosphorus] of 8-bromoadenosine-3', 5'-cyclic monophosphorothioate (Rp-8-Br-cyclic AMPS; 10 microM), but not cyclic GMP antagonist, Rp-isomer of 8-bromoguanosine-3', 5'-cyclic monophosphorothioate (Rp-8-Br-cyclic GMPS; 10 microM), reversed the effects of puerarin (10 microM) on the enhancement of vasorelaxation to sodium nitroprusside. Our results demonstrated that puerarin enhanced sodium nitroprusside-induced relaxation, possibly via the cyclic AMP-dependent pathway. Topics: 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid; 8-Bromo Cyclic Adenosine Monophosphate; Animals; Coronary Vessels; Cyclic AMP; Cyclic GMP; Dose-Response Relationship, Drug; Drug Synergism; Endothelium, Vascular; Enzyme Inhibitors; Female; In Vitro Techniques; Isoflavones; Male; NG-Nitroarginine Methyl Ester; Nitric Oxide Synthase; Nitroprusside; Plant Roots; Pueraria; Signal Transduction; Swine; Thionucleotides; Vasoconstriction; Vasoconstrictor Agents; Vasodilation; Vasodilator Agents | 2006 |
Effects of nitric oxide-active drugs on the discharge of subthalamic neurons: microiontophoretic evidence in the rat.
The presence of nitric oxide (NO) synthase and of soluble guanylyl cyclase, the main NO-activated metabolic pathway, has been demonstrated in many cells of the subthalamic nucleus. In this study, the effects induced on the firing of 96 subthalamic neurons by microiontophoretically administering drugs modifying NO neurotransmission were explored in anaesthetized rats. Recorded neurons were classified into regularly and irregularly discharging on the basis of their firing pattern. Nomega-nitro-L-arginine methyl ester (L-NAME; a NO synthase inhibitor), 3-morpholino-sydnonimin-hydrocloride (SIN-1; a NO donor), S-nitroso-glutathione (SNOG; another NO donor) and 8-Br-cGMP (a cell-permeable analogue of cGMP, the main second-messenger of NO neurotransmission) were iontophoretically applied while performing single-unit extracellular recordings. The activity of most neurons was influenced in a statistically significant way: in particular, both current-related inhibitory L-NAME-induced effects (20/39 tested cells) and excitatory effects of SIN-1 (25/41 tested neurons), SNOG (19/32 tested cells) and 8-Br-cGMP (13/19 tested neurons) were demonstrated. Neither statistically significant differences between the responses of regularly and irregularly discharging cells, nor specific topographical clustering of responding neurons, were demonstrated. Neurons administered drugs oppositely modulating the NO neurotransmission often displayed responses to only one treatment. We hypothesize that NO neurotransmission could exert a modulatory influence upon subthalamic neurons, with a prevalent excitatory effect. However, in the light of the presence of some responses of opposite sign to the same drug displayed by different subthalamic neurons, more complex effects of NO neurotransmission could be suggested, probably due to interactions with other classical neurotransmitter systems. Topics: Action Potentials; Analysis of Variance; Animals; Cyclic GMP; Dose-Response Relationship, Drug; Enzyme Activation; Enzyme Inhibitors; Iontophoresis; Male; Molsidomine; Neurons; NG-Nitroarginine Methyl Ester; Nitric Oxide; Nitric Oxide Donors; Rats; Rats, Wistar; S-Nitrosoglutathione; Subthalamic Nucleus; Thionucleotides; Time Factors | 2006 |
Nitric oxide directly inhibits ghrelin-activated neurons of the arcuate nucleus.
The hypothalamic arcuate nucleus (Arc) is a target site for signals regulating energy homeostasis. The orexigenic hormone ghrelin directly activates neurons of the medial arcuate nucleus (ArcM) in rats. Nitric oxide (NO) is a neuromodulator implicated in the control of food intake and body weight. NO is produced by nitric oxide synthase (NOS) and induces the formation of cyclic guanosine monophosphate (cGMP) via a stimulation of soluble guanylate cyclase (sGC). Both enzymes NOS and sGC have been identified in the Arc. Using extracellular recordings we characterized the effects of NO signaling on ArcM neurons and their co-sensitivity to ghrelin. The artificial NO donor sodium nitroprusside (10(-4) M) reversibly inhibited 91% of all ArcM neurons by a direct postsynaptic mechanism. 52% of ArcM neurons were excited by ghrelin. In all but one of these neurons SNP caused inhibitory responses. The SNP-induced inhibitions were mediated by cGMP since they were blocked by the specific sGC inhibitor ODQ (1H-[1,2,4]oxadiazole[4,3-a]quinoxalin-1-one, 10(-4) M). Furthermore, the membrane permeating cGMP analogue 8-Br-cGMP (10(-4) M) mimicked the inhibitory responses of SNP. In immunohistological in vitro studies SNP induced a cGMP formation, which could also be blocked by ODQ. The current studies demonstrate that NO/cGMP signaling inhibits a large population of ArcM neurons including ghrelin-excited cells. Since an activation of the latter neurons is regarded as a correlate of negative energy balance, NO may represent an anorectic neuromodulator in the Arc and/or restrain the action of signals promoting energy intake. NO signaling in the Arc is also induced following inflammation suggesting a possible role of Arc-intrinsic NO in disease-related anorexia. Topics: Action Potentials; Analysis of Variance; Animals; Arcuate Nucleus of Hypothalamus; Calcium; Cyclic GMP; Dose-Response Relationship, Drug; Drug Interactions; Enzyme Inhibitors; Gene Expression; Ghrelin; Immunohistochemistry; Magnesium; Male; Neural Inhibition; Neurons; Nitric Oxide; Nitric Oxide Donors; Nitroprusside; Oxadiazoles; Peptide Hormones; Quinoxalines; Rats; Rats, Wistar; Reaction Time; Thionucleotides | 2006 |
Antiproliferative effects of calcitonin gene-related peptide in aortic and pulmonary artery smooth muscle cells.
Pulmonary hypertension is characterized by vascular remodeling involving smooth muscle cell proliferation and migration. Calcitonin gene-related peptide (CGRP) and nitric oxide (NO) are potent vasodilators, and the inhibition of aortic smooth muscle cell (ASMC) proliferation by NO has been documented, but less is known about the effects of CGRP. The mechanism by which overexpression of CGRP inhibits proliferation in pulmonary artery smooth muscle cells (PASMC) and ASMC following in vitro transfection by the gene coding for prepro-CGRP was investigated. Increased expression of p53 is known to stimulate p21, which inhibits G(1) cyclin/cdk complexes, thereby inhibiting cell proliferation. We hypothesize that p53 and p21 are involved in the growth inhibitory effect of CGRP. In this study, CGRP was shown to inhibit ASMC and PASMC proliferation. In PASMC transfected with CGRP and exposed to a PKA inhibitor (PKAi), cell proliferation was restored. p53 and p21 expression increased in CGRP-treated cells but decreased in cells treated with CGRP and PKAi. PASMC treated with CGRP and a PKG inhibitor (PKGi) recovered from inhibition of proliferation induced by CGRP. ASMC treated with CGRP and then PKAi or PKGi recovered only when exposed to the PKAi and not PKGi. Although CGRP is thought to act through a cAMP-dependent pathway, cGMP involvement in the response to CGRP has been reported. It is concluded that p53 plays a role in CGRP-induced inhibition of cell proliferation and cAMP/PKA appears to mediate this effect in ASMC and PASMC, whereas cGMP appears to be involved in PASMC proliferation. Topics: 8-Bromo Cyclic Adenosine Monophosphate; Animals; Aorta; Calcitonin Gene-Related Peptide; Cell Division; Cells, Cultured; Cyclic AMP; Cyclic AMP-Dependent Protein Kinases; Cyclic GMP; Cyclic GMP-Dependent Protein Kinases; Enzyme Inhibitors; Male; Myocytes, Smooth Muscle; Pulmonary Artery; Rats; Rats, Sprague-Dawley; Thionucleotides; Transfection | 2005 |
Chronic hyperalgesic priming in the rat involves a novel interaction between cAMP and PKCepsilon second messenger pathways.
Toward the goal of defining new pharmacological targets for the treatment of chronic pain conditions, in previous studies we established a model, termed 'hyperalgesic priming,' in which an acute inflammatory stimulus causes a long-lasting latent susceptibility to hyperalgesia induced by subsequent exposures to the inflammatory mediator, prostaglandin E2 (PGE2). Those investigations suggested the hypothesis that priming induces a novel linkage between the PGE2-activated second messenger cascade and the epsilon isoform of protein kinase C (PKCepsilon). In the present study, comparison of dose-response relations for hyperalgesia produced by PGE2, forskolin, 8-Br-cAMP, or the protein kinase A (PKA) catalytic subunit, in primed versus normal animals, demonstrated that priming-induced enhancement of the PGE2-activated second messenger cascade occurs downstream to adenylate cyclase and upstream to PKA. Therefore, PGE2-induced hyperalgesia in the primed animal is enhanced by the recruitment of a novel cAMP/PKCepsilon signaling pathway in addition to the usual cAMP/PKA pathway. These observations suggest that pharmacological disruption of the novel interaction between cAMP and PKCepsilon might provide a route toward the development of highly specific methods to reverse cellular processes that underlie chronic pain states. Topics: 8-Bromo Cyclic Adenosine Monophosphate; Analysis of Variance; Animals; Behavior, Animal; Carrageenan; Colforsin; Cyclic AMP; Cyclic GMP; Dinoprostone; Disease Models, Animal; Dose-Response Relationship, Drug; Drug Interactions; Enzyme Inhibitors; Hyperalgesia; Male; Models, Biological; Pain Measurement; Pain Threshold; Protein Kinase C; Protein Kinase C-epsilon; Rats; Rats, Sprague-Dawley; Second Messenger Systems; Thionucleotides | 2005 |
Acute impairment of contractile responses by 17beta-estradiol is cAMP and protein kinase G dependent in vascular smooth muscle cells of the porcine coronary arteries.
The aim of the present study was to investigate the involvement of adenosine 3',5'-cyclic monophosphate (cAMP) cascade in the acute impairment of contraction by 17beta-estradiol in porcine coronary arteries, and to elucidate the signaling pathway leading to the activation of this cascade by the hormone. Isometric tension was recorded in isolated rings of porcine coronary arteries. The contraction to U46619 was reduced significantly following 30 min incubation with 1 nM 17beta-estradiol or 1 nM isoproterenol. There was no additive effect when 17beta-estradiol and isoproterenol were administered together. The effect of 17beta-estradiol was mimicked by both the cyclic AMP analogue 8-Br-cAMP and the guanosine 3',5'-cyclic monophosphate (cyclic GMP) analogue 8-Br-cGMP. In rings with and without endothelium, the modulatory effect of 17beta-estradiol was abolished by the adenylyl cyclase inhibitor, SQ 22536, but was unaffected by the guanylyl cyclase inhibitor, ODQ. Both the cAMP antagonist Rp-8-Br-cAMPS and the cGMP antagonist inhibitor Rp-8-Br-cGMPS inhibited the effect of 17beta-estradiol. The effect of 17beta-estradiol was unaffected by the protein kinase A inhibitor, KT5720, but was abolished by the protein kinase G (PKG) inhibitor, KT5823, which also abolished the effect of isoproterenol. These data support our earlier findings that 17beta-estradiol (1 nM) acutely impairs contractile responses of porcine coronary arteries in vitro. This acute effect of 17beta-estradiol involves cAMP in vascular smooth muscles and the activation of PKG. Topics: 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid; 8-Bromo Cyclic Adenosine Monophosphate; Adenine; Adenylyl Cyclase Inhibitors; Animals; Carbazoles; Coronary Vessels; Cyclic AMP; Cyclic AMP-Dependent Protein Kinases; Cyclic GMP; Cyclic GMP-Dependent Protein Kinases; Drug Interactions; Estradiol; Indoles; Isometric Contraction; Isoproterenol; Muscle, Smooth, Vascular; Swine; Thionucleotides; Time Factors | 2005 |
Formation of releasable NO stores by S-nitrosoglutathione in arteries exhibiting tolerance to glyceryl-trinitrate.
S-Nitrosating nitric oxide (NO) donors like S-nitrosoglutathione (GSNO) induce a persistent inhibition of vascular tone, through the formation of releasable NO stores. In this study, we investigate whether GSNO also induces NO stores-related effects in vessels exhibiting tolerance to glyceryl-trinitrate. Rat aortic rings treated with glyceryl-trinitrate (100 microM for 1 h) exhibited increased level of superoxide and a decrease in NO elevation and relaxation induced by subsequent addition of glyceryl-trinitrate. In glyceryl-trinitrate-treated rings as in controls, pre-exposure to GSNO (1 microM for 30 min) induced a persistent hyporesponsiveness to noradrenaline and a relaxant response to N-acetylcysteine (a low molecular weight thiol which can displace NO from NO stores), both of which being inhibited by guanylyl-cyclase or cyclic GMP-dependent protein kinase inhibitors. These data indicate that GSNO can promote the formation of releasable NO stores in arteries exhibiting increased superoxide level and tolerance to glyceryl-trinitrate. Formation of releasable NO stores is of potential interest to restore the protective effect of NO in organic nitrate-tolerant blood vessels. Topics: Acetylcysteine; Analysis of Variance; Animals; Aorta, Thoracic; Cyclic GMP; Diethylamines; Dose-Response Relationship, Drug; Drug Tolerance; In Vitro Techniques; Male; Nitric Oxide; Nitrogen Oxides; Nitroglycerin; Norepinephrine; Oxadiazoles; Quinoxalines; Rats; Rats, Wistar; S-Nitrosoglutathione; Superoxide Dismutase; Thionucleotides; Vasoconstriction; Vasodilation | 2005 |
Sildenafil citrate and sildenafil nitrate (NCX 911) are potent inhibitors of superoxide formation and gp91phox expression in porcine pulmonary artery endothelial cells.
Acute respiratory distress syndrome (ARDS) is associated with increased superoxide (O(2)(*-)) formation in the pulmonary vasculature and negation of the bioavailability of nitric oxide (NO). Since NO inhibits NADPH oxidase expression through a cyclic GMP-mediated mechanism, sildenafil, a type V phosphodiesterase inhibitor, may be therapeutically effective in ARDS through an augmentation of NO-mediated inhibition of NADPH oxidase. Therefore, the effect of sildenafil citrate and NO-donating sildenafil (NCX 911) on O(2)(*-) formation and gp91(phox) (active catalytic subunit of NADPH oxidase) expression was investigated in cultured porcine pulmonary artery endothelial cells (PAECs). PAECs were incubated with 10 nM TXA(2) analogue, 9,11-dideoxy-9alpha,11alpha-methanoepoxy-prostaglandin F(2alpha) (U46619) (+/-sildenafil or NCX 911), for 16 h and O(2)(*-) formation measured spectrophometrically and gp91(phox) using Western blotting. The role of the NO-cGMP axis was studied using morpholinosydnonimine hydrochloride (SIN-1), the diethylamine/NO complex (DETA-NONOate), the guanylyl cyclase inhibitor, 1H-{1,2,4}oxadiazolo{4,3-a}quinoxalin-1-one (ODQ), and the protein kinase G inhibitor, 8-bromoguanosine-3',5'-cyclic monophosphorothioate, Rp-isomer (Rp-8-Br-cGMPS). NO release was studied using a fluorescence assay and O(2)(*-)-NO interactions by measuring nitrites. After a 16-h incubation with 10 nM U46619, both NCX 911 and sildenafil elicited a concentration-dependent inhibition of O(2)(*-) formation and gp91(phox) expression, NCX 911 being more potent (IC(50); 0.26 nM) than sildenafil citrate (IC(50); 1.85 nM). These inhibitory effects were reversed by 1 microM ODQ and 10 microM Rp-8-Br-cGMPS. NCX 911 stimulated the formation of cGMP in PAECs and generated NO in a cell-free system to a greater degree than sildenafil citrate. The inhibitory effect of sildenafil was augmented by 1 muM SIN-1 and blocked partially by the eNOS inhibitor 10 microM N(5)-(1-iminoethyl)-ornithine (L-NIO). Acutely, sildenafil and NCX 911 also inhibited O(2)(*-) formation, again blocked by 1 microM ODQ. NCX 911 reacted with O(2)(*-) generated by xanthine oxidase, an effect that was inhibited by superoxide dismutase (500 U ml(-1)). Since O(2)(*-) formation plays contributory role in ARDS, both sildenafil citrate and NCX 911 may be indicated for treating ARDS through suppression of NADPH oxidase expression and therefore of O(2)(*-) formation and preservation of NO bioavailability. Topics: 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid; Animals; Cells, Cultured; Cyclic GMP; Endothelial Cells; Guanylate Cyclase; Male; Membrane Glycoproteins; NADPH Oxidases; Nitric Oxide; Nitric Oxide Synthase; Ornithine; Oxadiazoles; Piperazines; Protein Kinase Inhibitors; Pulmonary Artery; Purines; Quinoxalines; Sildenafil Citrate; Sulfones; Superoxides; Swine; Thionucleotides | 2005 |
C-type natriuretic peptide applied to the brain enhances exocrine pancreatic secretion through a vagal pathway.
C-type natriuretic peptide (CNP) is the major natriuretic peptide in the brain and its mRNA has been reported in the central nervous system, which supports local synthesis and its role as a neuromodulator. The aim of the present work was to study the effect of centrally applied CNP on pancreatic secretion. Rats were fitted with a lateral cerebroventricular cannula one-week before secretion studies. The central administration of CNP dose-dependently enhanced pancreatic fluid and protein output. CNP response was diminished by atropine and hexamethonium, but it was abolished by vagotomy. Neither adrenergic antagonists nor the administration of (D-p-Cl-Phe(6),Leu(17))-vasoactive intestinal peptide (VIP antagonist) or N(omega) Nitro-L arginine methyl ester (L-NAME) (nitric oxide synthase inhibitor) affected CNP response. The effect induced by CNP was mimicked by 8-Br-cGMP but not by c-ANP-(4-23) amide (selective agonist of the natriuretic peptide receptor C). Furthermore, CNP interacted with cholecystokinin (CCK) and secretin in the brain to modify pancreatic secretion. Present findings show that centrally applied CNP enhanced pancreatic secretion through a vagal pathway and suggest that CNP response is mediated by the activation of natriuretic peptide guanylyl cyclase coupled receptors in the brain. Topics: Animals; Atrial Natriuretic Factor; Atropine; Brain; Cyclic GMP; Drug Interactions; Enzyme Inhibitors; Ganglionic Blockers; Hexamethonium; Natriuretic Peptide, C-Type; NG-Nitroarginine Methyl Ester; Nitric Oxide Synthase; Pancreas; Parasympatholytics; Peptide Fragments; Proteins; Rats; Rats, Sprague-Dawley; Receptors, Atrial Natriuretic Factor; Secretin; Sincalide; Thionucleotides; Time Factors; Vagotomy; Vagus Nerve; Vasoactive Intestinal Peptide | 2005 |
Protein kinase G II-mediated proliferative effects in human cultured prostatic stromal cells.
This study investigates the effect of protein kinase G (PKG) activation upon proliferation of human cultured prostatic stromal cells. The PKG II activator (8-pCPT-cGMP; IC50 of 113+/-42 nM) and the phosphodiesterase inhibitor, zaprinast (up to 50 microM), but not the PKG I isoform activators (APT-cGMP and PET-cGMP), reduced foetal calf serum-stimulated proliferation. The effect of 8-pCPT-cGMP (30 microM) was blocked by Rp-8-Br-cGMPS (5 microM) and Rp-8-pCPT-cGMP (5 microM), but not Rp-cAMPS (5 microM). 8-pCPT-cGMP (30 microM) and zaprinast (50 microM), but not PET-cGMP (30 microM), caused a significant increase in atypical nuclei and an increase in annexin-V staining. These data indicate that activation of PKG II induces apoptosis of human cultured prostatic stromal cells. Topics: Aged; Apoptosis; Azides; Cell Division; Cell Nucleus; Cells, Cultured; Cyclic AMP; Cyclic GMP; Cyclic GMP-Dependent Protein Kinases; Enzyme Inhibitors; Humans; Isoenzymes; Male; Phosphoric Diester Hydrolases; Prostate; Prostatic Hyperplasia; Purinones; Serum; Stromal Cells; Thionucleotides | 2004 |
Reduced inflammatory hyperalgesia with preservation of acute thermal nociception in mice lacking cGMP-dependent protein kinase I.
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. Topics: Animals; Cyclic GMP; Cyclic GMP-Dependent Protein Kinases; Disease Models, Animal; Formaldehyde; Hot Temperature; Hyperalgesia; Inflammation; Mice; Mice, Knockout; Neurons; Pain; Reaction Time; Substance P; Thionucleotides | 2004 |
Mouse photoreceptor synaptic ribbons lose and regain material in response to illumination changes.
Abstract Chemical synapses equipped with ribbons are tonically active, high-output synapses. The ribbons may play a role in the trafficking of synaptic vesicles. Recent findings in retinal rod cells of BALB/c mice indicate that ribbons are large and smooth in the dark phase, and, due to the formation and release of protrusions, small during the light phase. As a consequence of these changes, ribbons may traffick fewer vesicles in the light than in the dark phases. The aim of the present study was to find out whether the above ribbon changes in this mouse strain are strictly illumination-dependent and which signalling processes may be involved. Here, we show that ribbons form protrusions and release them into the cytoplasm within 30-60 min after lights on, the reverse occurring within 30 min after lights off. Under constant light or constant dark, no circadian rhythm of synaptic ribbon changes is observed. The illumination-dependence of ribbon structure is supported by in vitro experiments showing that in dark-adapted retinas, light induces the same morphological changes as in vivo. In vitro, the effect of light on the ribbons can be counteracted by cyclic guanosine monophosphate and melatonin. In dark-adapted retinas, light effects can be produced by decreasing the calcium ion concentrations in the incubation media. These results suggest that in retinal rod cells, the well known phototransduction signalling mechanisms may be responsible for the ribbon changes presently and previously reported. Topics: Animals; Calcimycin; Calcium Chloride; Chelating Agents; Circadian Rhythm; Cyclic GMP; Dark Adaptation; Darkness; Drug Interactions; Egtazic Acid; Ionophores; Light; Lighting; Male; Melatonin; Mice; Mice, Inbred BALB C; Microscopy, Electron; Models, Biological; Organ Culture Techniques; Photic Stimulation; Photoreceptor Cells; Retina; Synapses; Thionucleotides; Time Factors | 2004 |
Elevation of intracellular cAMP evokes activity-dependent release of adenosine in cultured rat forebrain neurons.
Adenosine is an important regulator of neuronal excitability. Zaprinast is a cyclic nucleotide phosphodiesterase inhibitor, and has been shown in the hippocampal slice to suppress excitation. This action can be blocked by an adenosine receptor antagonist, and therefore is presumably due to adenosine release stimulated by exposure to zaprinast. To explore the mechanism of this phenomenon further, we examined the effect of zaprinast on adenosine release itself in cultured rat forebrain neurons. Zaprinast significantly stimulated extracellular adenosine accumulation. The effect of zaprinast on adenosine appeared to be mediated by increasing intracellular cyclic adenosine monophosphate (cAMP) and activation of protein kinase A (PKA): (i) zaprinast stimulated intracellular cAMP accumulation; (ii) a cAMP antagonist (Rp-8-Br-cAMP) significantly reduced the zaprinast effect on adenosine; (iii) an inhibitor of phosphodiesterase (PDE)1 (vinpocetine) and an activator of adenylate cyclase (forskolin) mimicked the effect of zaprinast on adenosine. We also found that zaprinast had no effect on adenosine in astrocyte cultures, and tetrodotoxin completely blocked zaprinast-evoked adenosine accumulation in neuronal cultures, suggesting that neuronal activity was likely to be involved. Consistent with a dependence on neuronal activity, NMDA receptor antagonists (MK-801 and D-APV) and removal of extracellular glutamate by glutamate-pyruvate transaminase blocked the effect of zaprinast. In addition, zaprinast was shown to stimulate glutamate release. Thus, our data suggest that zaprinast-evoked adenosine accumulation is likely to be mediated by stimulation of glutamate release by a cAMP- and PKA-dependent mechanism, most likely by inhibition of PDE1 in neurons. Furthermore, regulation of cAMP, either by inhibiting cAMP-PDE activity or by stimulating adenylate cyclase activity, may play an important role in modulating neuronal excitability. These data suggest the existence of a homeostatic negative feedback loop in which increases in neuronal activity are damped by release of adenosine following activation of glutamate receptors. Topics: Adenosine; Animals; Animals, Newborn; Astrocytes; Cells, Cultured; Cyclic AMP; Cyclic GMP; Dizocilpine Maleate; Dose-Response Relationship, Drug; Drug Interactions; Embryo, Mammalian; Enzyme Activation; Excitatory Amino Acid Agonists; Excitatory Amino Acid Antagonists; Extracellular Space; Female; Glutamic Acid; Intracellular Space; Male; Membrane Potentials; Models, Neurological; N-Methylaspartate; Neurons; Patch-Clamp Techniques; Phosphodiesterase Inhibitors; Pregnancy; Prosencephalon; Purinones; Rats; Rats, Sprague-Dawley; Thionucleotides | 2004 |
Nitric oxide modulates local reflexes of the tailfan of the crayfish.
Electrical stimulation of sensory neurons that innervate receptors on the tailfan of crayfish evokes a reflex response of motor neurons that produce movements of the blades of the tailfan, the uropods. We analyzed the modulatory effects of nitric oxide (NO) on the spike frequency of the reflex response. Bath application of L-arginine and SNAP, which elevate endogenous and exogenous NO levels, increased the frequency of the evoked response, whereas the application of L-NAME and PTIO, which reduce NO levels, decreased the frequency of the response. To determine through what pathway and target NO exerted these effects we bath applied ODQ, an inhibitor of soluble guanylyl cyclase (sGC), which decreased the frequency of response, and 8-br-cGMP, which increased the spike frequency of response. To provide further evidence that NO acts via sGC, we elevated NO levels with L-arginine while simultaneously inhibiting sGC with ODQ. This application reduced the response to control levels, indicating that NO in the terminal ganglion of crayfish acts via sGC to modulate cGMP levels, which in turn regulate the responses of the uropod motor neurons. Topics: Analysis of Variance; Animals; Arginine; Astacoidea; Cyclic GMP; Cyclic N-Oxides; Drug Interactions; Electric Stimulation; Enzyme Inhibitors; Excitatory Postsynaptic Potentials; Female; Free Radical Scavengers; Imidazoles; In Vitro Techniques; Male; Muscles; Neurons; NG-Nitroarginine Methyl Ester; Nitric Oxide; Nitric Oxide Donors; Oxadiazoles; Penicillamine; Picolines; Quinoxalines; Reflex; Thionucleotides | 2004 |
Modulation of soluble guanylate cyclase activity by phosphorylation.
The levels of the cGMP in smooth muscle of the gut reflect continued synthesis by soluble guanylate cyclase (GC) and breakdown by phosphodiesterase 5 (PDE5). Soluble GC is a haem-containing, heterodimeric protein consisting alpha- and beta-subunits: each subunit has N-terminal regulatory domain and a C-terminal catalytic domain. The haem moiety acts as an intracellular receptor for nitric oxide (NO) and determines the ability of NO to activate the enzyme and generate cGMP. In the present study the mechanism by which protein kinases regulate soluble GC in gastric smooth muscle was examined. Sodium nitroprusside (SNP) acting as a NO donor stimulated soluble GC activity and increased cGMP levels. SNP induced soluble GC phosphorylation in a concentration-dependent fashion. SNP-induced soluble GC phosphorylation was abolished by the selective cGMP-dependent protein kinase (PKG) inhibitors, Rp-cGMPS and KT-5823. In contrast, SNP-stimulated soluble GC activity and cGMP levels were significantly enhanced by Rp-cGMPS and KT-5823. Phosphorylation and inhibition of soluble GC were PKG specific, as selective activator of cAMP-dependent protein kinase, Sp-5, 6-DCl-cBiMPS had no effect on SNP-induced soluble GC phosphorylation and activity. The ability of PKG to stimulate soluble GC phosphorylation was demonstrated in vitro by back phosphorylation technique. Addition of purified phosphatase 1 inhibited soluble GC phosphorylation in vitro, and inhibition was reversed by a high concentration (10 microM) of okadaic acid. In gastric smooth muscle cells, inhibition of phosphatase activity by okadaic acid increased soluble GC phosphorylation in a concentration-dependent fashion. The increase in soluble GC phosphorylation inhibited SNP-stimulated soluble GC activity and cGMP formation. The results implied the feedback inhibition of soluble GC activity by PKG-dependent phosphorylation impeded further formation of cGMP. Topics: Carbazoles; Cyclic AMP; Cyclic AMP-Dependent Protein Kinases; Cyclic GMP; Enzyme Inhibitors; Guanylate Cyclase; Humans; Indoles; Muscle, Smooth; Nitric Oxide Donors; Nitroprusside; Okadaic Acid; Phosphoric Monoester Hydrolases; Phosphorylation; Radioimmunoassay; Receptors, Cytoplasmic and Nuclear; Solubility; Soluble Guanylyl Cyclase; Stomach; Thionucleotides | 2004 |
Chorion-induced myometrial relaxation is mediated by large-conductance Ca2+-activated K+ channel opening in the guinea pig.
We previously demonstrated that chorion releases a factor that inhibits both spontaneous and oxytocin-stimulated myometrial contractility. Here, we investigate the mechanism of action of this unidentified substance.. Myometrial strips from pregnant guinea pigs were mounted in an organ bath and contractility stimulated with oxytocin.. Guinea pig chorion produced a time-dependent decrease in oxytocin-induced myometrial contractility. The ability of the chorion to reduce contractility was unaltered by inhibiting chorionic synthesis of either nitric oxide (N [omega]-nitro-L-arginine), carbon monoxide (tin-protoporphyrin), prostaglandins (indomethacin), or the myometrial cyclic guanosine monophosphate pathway (1H-[1,2,4]oxadiazolo[4,3-a]quinoxalime-1-one and Rp-8Br-cGMP). In contrast, iberiotoxin, an inhibitor of large conductance Ca(2+)-activated K(+) channels reduced the quiescent effect of chorion by 40%; in contrast, inhibition of adenosine triphosphate-sensitive (glibenclamide) and voltage-gated K(+) channels (4amynopyridine) had no effect.. Chorion-induced relaxation of oxytocin-stimulated myometrial contraction is, in great part, the product of a paracrine substance that opens myometrial large conductance Ca(2+)-activated K(+) channels. Topics: Animals; Chorion; Culture Media, Conditioned; Culture Techniques; Cyclic GMP; Cyclic GMP-Dependent Protein Kinases; Enzyme Inhibitors; Female; Guinea Pigs; Ion Channel Gating; Muscle Relaxation; Myometrium; Peptides; Potassium Channel Blockers; Potassium Channels, Calcium-Activated; Pregnancy; Tetraethylammonium; Thionucleotides; Uterine Contraction | 2003 |
Inhibition of cyclic guanosine 5'-monophosphate-dependent protein kinase I (PKG-I) in lumbar spinal cord reduces formalin-induced hyperalgesia and PKG upregulation.
Nitric oxide-mediated nociception has been suggested to involve formation of cyclic guanosine 5'-monophosphate (cGMP) and activation of cGMP-dependent protein kinase (PKG). To further evaluate this pathway we assessed the effects of the PKG-inhibiting cGMP analog Rp-8-Br-cGMPS in the rat formalin assay and analyzed the regulation of PKG expression in rat lumbar spinal cord. Spinally delivered Rp-8-Br-cGMPS (0.1-0.5 micro mol i.t.) reduced the nociceptive behavior in a dose-dependent manner. Similar effects were achieved with Rp-8-Br-PET-cGMPS (0.5 micro mol i.t.), another PKG-inhibitory cGMP analog. In contrast, Rp-8-Br-cAMPS (0.5 micro mol i.t.), an inhibitor of protein kinase A, had no effect in this model. Formalin treatment resulted in a rapid (within 1h), long-lasting (up to 96h) upregulation of PKG-I protein expression. This increase was prevented in animals pretreated with Rp-8-Br-cGMPS (0.5 micro mol i.t.) or morphine (2.5-5mg/kg i.p.) 10min prior to formalin injection. Spinal delivery of 8-Br-cGMP, a PKG-activating cGMP analog, without subsequent formalin treatment also caused an increase of PKG-I protein expression. Hence, the upregulation of PKG-I might possibly be mediated by cGMP itself. Our data suggest that PKG-I activation is involved in the synaptic transmission of nociceptive stimuli in the spinal cord and that PKG-I inhibitors might be interesting novel drugs for pain treatment. Topics: Animals; Blotting, Western; Cyclic GMP; Cyclic GMP-Dependent Protein Kinases; Dose-Response Relationship, Drug; Enzyme Inhibitors; Formaldehyde; Hyperalgesia; Lumbar Vertebrae; Nitric Oxide; Pain Measurement; Rats; Spinal Cord; Thionucleotides; Time Factors | 2003 |
The role of two novel regulatory sites in the activation of the cGMP-dependent protein kinase from Plasmodium falciparum.
The Plasmodium falciparum cGMP-dependent protein kinase (PfPKG) uniquely contains three cGMP binding sites, but also has a 'degenerate' fourth site. The role of each cGMP-binding site in PfPKG activation remains unknown. We have analysed the effect of mutation of each cGMP-binding site (individually and in combination) on PfPKG activation in vitro. The most striking result was that mutation of cGMP site 3 resulted in a 10-49-fold increase in the K (a((cGMP))) value and a 45-55% decrease in maximal activity compared with wild-type. Mutations involving only cGMP-binding sites 1 and 2 had less effect on both the K (a((cGMP))) values and the maximal activities. These results suggest that, although all three cGMP-binding sites are involved in PfPKG activation, cGMP-binding site 3 has the greatest influence on activation. A mutation in the fourth, degenerate cGMP-binding site decreased PfPKG maximal activity by 40%, but did not change the K (a((cGMP))) value for the PfPKG mutant, suggesting that this site does not bind cGMP, but is required for full activation of PfPKG. The distinct activation properties of PfPKG from mammalian isoforms may be exploitable in the design of a parasite-specific inhibitor and development of a novel anti-malarial drug. Topics: Alanine; Allosteric Regulation; Allosteric Site; Amino Acid Sequence; Amino Acid Substitution; Animals; Binding, Competitive; Conserved Sequence; Cyclic GMP; Cyclic GMP-Dependent Protein Kinases; Enzyme Activation; Gene Duplication; Molecular Sequence Data; Mutagenesis, Site-Directed; Plasmodium falciparum; Protein Binding; Protozoan Proteins; Recombinant Proteins; Serine; Thionucleotides; Threonine | 2003 |
Role of phosphodiesterase 3 in NO/cGMP-mediated antiinflammatory effects in vascular smooth muscle cells.
Atherosclerosis involves cellular immune responses and altered vascular smooth muscle cell (VSMC) function. Nitric oxide (NO)/cGMP is uniquely capable of inhibiting key processes in atherosclerosis. In this study, we determined the effects of NO/cGMP and their molecular mechanisms in the regulation of NF-kappaB-dependent gene expression in VSMCs. We found that cGMP-elevating agents such as the NO donor S-nitroso-N-acetylpenicillamine (SNAP) and C-type natriuretic peptide (CNP), reduced TNF-alpha-induced NF-kappaB-dependent reporter gene expression in rat aortic VSMCs in a cGMP-dependent manner. The effects of SNAP and CNP on NF-kappaB are mediated by cAMP-dependent protein kinase (PKA) but not cGMP-dependent protein kinase (PKG) based on the findings that the selective PKA inhibitor, PKI, abolished the effects of SNAP and CNP on NF-kappaB, whereas the PKG inhibitor Rp-8-Br-PET-cGMP had no effect. Inhibition of cGMP-inhibited cAMP-hydrolyzing phosphodiesterase 3 (PDE3) blocked SNAP- and CNP-elicited effects on NF-kappaB-dependent transcription. Furthermore, cGMP analogues such as 8-pCPT-cGMP, which selectively activates PKG but does not inhibit PDE3, had no effect on NF-kappaB-mediated transcription. Activation of PKA by SNAP or cAMP-elevating agents not only inhibited TNF-alpha-induced NF-kappaB-dependent reporter gene expression but also reduced endogenous NF-kappaB-dependent adhesion molecule and chemokine expression. These results suggest that SNAP and CNP exert inhibitory effects on NF-kappaB-dependent transcription by activation of PKA via cGMP-dependent inhibition of PDE3 activity. Therefore, PDE3 is a novel mediator of inflammation in VSMCs. Topics: 3',5'-Cyclic-AMP Phosphodiesterases; Animals; Blotting, Western; Cells, Cultured; Chemokine CCL2; Cyclic AMP-Dependent Protein Kinases; Cyclic GMP; Cyclic GMP-Dependent Protein Kinases; Cyclic Nucleotide Phosphodiesterases, Type 3; Enzyme Inhibitors; Gene Expression Regulation; Guanylate Cyclase; Inflammation Mediators; Luciferases; Muscle, Smooth, Vascular; Natriuretic Peptide, C-Type; NF-kappa B; Nitric Oxide; Nitric Oxide Donors; Oxadiazoles; Quinoxalines; Rats; RNA, Messenger; S-Nitroso-N-Acetylpenicillamine; Thionucleotides; Transcription, Genetic; Tumor Necrosis Factor-alpha; Vascular Cell Adhesion Molecule-1 | 2003 |
Negative functional effects of cyclic GMP are altered by cyclic AMP phosphodiesterases in rabbit cardiac myocytes.
In this study, we tested the hypothesis that the negative functional effects of cyclic GMP on cardiac myocytes would be affected by the actions of cyclic GMP on cyclic AMP phosphodiesterases. Ventricular myocytes from eight rabbits were used to determine the functional and cyclic AMP changes caused by 10(-7), 10(-6), 10(-5) M 8-Bromo-cGMP alone and after the administration of 10(-6) M milrinone (cyclic GMP-inhibited cyclic AMP phosphodiesterase inhibitor) or 10(-6) M erythro-9-(2-Hydroxy-3-3-nonyl)adenine (EHNA, cyclic GMP-stimulated cyclic AMP phosphodiesterase inhibitor). 8-Br-cGMP dose-dependently reduced %shortening by 35+/-4% of baseline at 10(-5) M. This effect was significantly blunted by EHNA at all doses. The maximum rate of shortening was reduced by 31+/-3% by 10(-5) M 8-Br-cGMP. This effect of 8-Br-cGMP was significantly enhanced (42+/-4%) in the milrinone group. A similar pattern was observed in the maximum rate of relaxation data. Cyclic AMP levels were significantly increased from a baseline level of 4.0+/-0.8 pmol/10(5) myocytes by milrinone (+60%), EHNA (+61%) and 8-Br-cGMP (+47%). The combination of EHNA plus 8-Br-cGMP increased cyclic AMP levels significantly more that the combination of milrinone plus 8-Br-cGMP. Exogenous cyclic GMP reduces myocyte function, while raising cyclic AMP possibly through cyclic GMP-inhibited cyclic AMP phosphodiesterase effects. Blocking cyclic GMP-inhibited cyclic AMP phosphodiesterase enhances the functional effects cyclic GMP, while blocking cyclic GMP-stimulated cyclic AMP phosphodiesterase reduced these effects. The study demonstrated a functional interaction between cyclic GMP and cyclic AMP related to the cyclic GMP affected cyclic AMP phosphodiesterases. Topics: 3',5'-Cyclic-AMP Phosphodiesterases; Adenine; Animals; Cell Size; Cyclic AMP; Cyclic GMP; Dose-Response Relationship, Drug; Milrinone; Myocytes, Cardiac; Phosphodiesterase Inhibitors; Rabbits; Thionucleotides | 2003 |
The NO signaling pathway differentially regulates KCC3a and KCC3b mRNA expression.
Nitric oxide (NO) donors and protein kinase G (PKG) acutely up-regulate K-Cl cotransporter-1 and -3 (KCC1 and KCC3) mRNA expression in vascular smooth muscle cells (VSMCs). Here, we report the presence, relative abundance, and regulation by sodium nitroprusside (SNP) of the novel KCC3a and KCC3b mRNAs, in primary cultures of rat VSMCs. KCC3a and KCC3b mRNAs were expressed in an approximate 3:1 ratio, as determined by semiquantitative RT-PCR analysis. SNP as well as YC-1 and 8-Br-cGMP, a NO-independent stimulator of soluble guanylyl cyclase (sGC) and PKG, respectively, increased KCC3a and KCC3b mRNA expression by 2.5-fold and 8.1-fold in a time-dependent manner, following a differential kinetics. Stimulation of the NO/sGC/PKG signaling pathway with either SNP, YC-1, or 8-Br-cGMP decreased the KCC3a/KCC3b ratio from 3.0+/-0.4 to 0.9+/-0.1. This is the first report on a differential regulation by the NO/sGC/PKG signaling pathway of a cotransporter and of KCC3a and KCC3b mRNA expression. Topics: Animals; Cells, Cultured; Cyclic GMP; Cyclic GMP-Dependent Protein Kinases; Gene Expression Regulation; Guanylate Cyclase; Indazoles; Muscle, Smooth, Vascular; Nitric Oxide; Nitroprusside; Protein Isoforms; Rats; Receptors, Cytoplasmic and Nuclear; RNA, Messenger; Signal Transduction; Soluble Guanylyl Cyclase; Symporters; Thionucleotides | 2003 |
Hemin, a heme oxygenase substrate analog, both inhibits and enhances neutrophil random migration and chemotaxis.
Carbon monoxide (CO), is an endogenously produced gas, generated by the rate-limiting enzyme heme oxygenase (HO), present in man throughout the respiratory tract. CO can elicit important physiological responses like bronchial relaxation and vasodilation. Both HO expression and CO levels in the airways increase in response to hypoxic challenge and to a wide variety of inflammatory stimuli, such as intermittent allergic rhinitis, asthma and upper respiratory tract infections. A role for CO in airway regulation and inflammation has therefore been suggested. However, information about CO-induced effects on cells involved in airway inflammation is scarce. The present study was designed to investigate if the HO substrate analog hemin could affect neutrophil random migration, and N-formyl-L-methionyl-L-leucyl-L-phenylalanine (fMLP) induced chemotaxis.. Hemin was added to and incubated with whole blood and the effects of the anticipated CO production were then evaluated on isolated neutrophils using a chemotaxis chamber.. A biphasic dose-response curve emerged for both the neutrophil spontaneous random migration and the fMLP-induced chemotaxis. Low concentrations of hemin (10(-11) m to 10(-9) m) enhanced the migratory response, whereas higher concentrations (10(-7) m and 10(-5) m) inhibited migration. The inhibition induced by hemin on fMLP-induced migration was abolished after pre-treatment with Rp-8Br-cyclicGMPS, an inhibitor of cyclicGMP.. The present data indicate that endogenously produced CO can affect both spontaneous and stimulated neutrophil migration, partly via a cyclicGMP-related process, hence strengthening the idea of a role for CO in airway inflammation. Topics: Cell Movement; Chemotaxis, Leukocyte; Cyclic GMP; Dose-Response Relationship, Drug; Enzyme Inhibitors; Heme Oxygenase (Decyclizing); Hemin; Humans; N-Formylmethionine Leucyl-Phenylalanine; Neutrophils; Reference Values; Thionucleotides | 2002 |
Long-term potentiation in hippocampus involves sequential activation of soluble guanylate cyclase, cGMP-dependent protein kinase, and cGMP-degrading phosphodiesterase.
Previous studies indicate that cGMP is involved in long-term potentiation (LTP). However, the effects of application of tetanus to induce LTP on cGMP content and the mechanisms by which cGMP may modulate LTP have not been reported. The aim of this work was to study the time course of the changes in cGMP content and of the activity of soluble guanylate cyclase (sGC) (the enzyme that synthesizes cGMP) during LTP. Moreover, we also studied how the changes in cGMP affect cGMP-dependent protein kinase (PKG) and cGMP-degrading phosphodiesterase and the possible role of these changes in LTP. Application of tetanus induced a rise in cGMP, reaching a maximum 10 sec after tetanus. cGMP content decreased below basal levels 5 min after tetanus and remained decreased after 60 min. Activity of sGC increased 5 min after tetanus and returned to basal at 60 min. Tetanus increased the activity of cGMP-degrading phosphodiesterase at 5 and 60 min. GMP, the product of degradation, was increased at 5 and 60 min. Activation of phosphodiesterase and a decrease in cGMP were prevented by inhibiting PKG with Rp-8-bromoguanosine-cGMPS (Rp-8-Br-cGMPS). Inhibition of sGC [with ODQ (oxadiazolo quinoxalin-1-one) or NS 2028 (4H-8-bromo-1,2,4-oxadiazolo(3,4-d)benz(b)(1,4)oxazin-1-one)], of PKG (with Rp-8-Br-cGMPS), or of cGMP-degrading phosphodiesterase [with zaprinast or MBAM (4-[[3',4'-(methylenedioxy)benzyl]amino]-6-methoxyquinazoline) ] impairs LTP. The results indicate that induction of LTP involves transient activation of sGC and an increase in cGMP, followed by activation of cGMP-dependent protein kinase, which, in turn, activates cGMP-degrading phosphodiesterase, resulting in long-lasting reduction of cGMP content. Topics: 3',5'-Cyclic-GMP Phosphodiesterases; Animals; Cyclic GMP; Cyclic GMP-Dependent Protein Kinases; Electric Stimulation; Enzyme Activation; Enzyme Inhibitors; Excitatory Amino Acid Antagonists; Excitatory Postsynaptic Potentials; Guanylate Cyclase; Hippocampus; In Vitro Techniques; Long-Term Potentiation; Male; Nitric Oxide Synthase; Rats; Rats, Wistar; Receptors, Cytoplasmic and Nuclear; Receptors, N-Methyl-D-Aspartate; Soluble Guanylyl Cyclase; Thionucleotides | 2002 |
Differential effects of soluble and particulate guanylyl cyclase on Ca(2+) sensitivity in airway smooth muscle.
Maximal relaxation of airway smooth muscle (ASM) in response to atrial natriuretic peptide (ANP), which stimulates particulate guanylyl cyclase (pGC), is less than that produced by nitric oxide (NO) and other compounds that stimulate soluble guanylyl cyclase (sGC). We hypothesized that stimulation of pGC relaxes ASM only by decreasing intracellular Ca(2+) concentration ([Ca(2+)](i)), whereas stimulation of sGC decreases both [Ca(2+)](i) and the force developed for a given [Ca(2+)](i) (i.e., the Ca(2+) sensitivity) during muscarinic stimulation. We measured the relationship between force and [Ca(2+)](i) (using fura 2) under control conditions (using diltiazem to change [Ca(2+)](i)) and during exposure to ANP, diethylamine-NO (DEA-NO), sodium nitroprusside (SNP), and the Sp diastereoisomer of beta-phenyl-1,N(2)-etheno-8-bromoguanosine-3',5'-cyclic monophosphorothionate (Sp-8-Br-PET-cGMPS), a cell-permeant analog of cGMP. Addition of DEA-NO, SNP, or Sp-8-Br-PET-cGMPS decreased both [Ca(2+)](i) and force, causing a significant rightward shift of the force-[Ca(2+)](i) relationship. In contrast, with ANP exposure, the force-[Ca(2+)](i) relationship was identical to control, such that ANP produced relaxation solely by decreasing [Ca(2+)](i). Thus, during muscarinic stimulation, stimulation of pGC relaxes ASM exclusively by decreasing [Ca(2+)](i), whereas stimulation of sGC decreases both [Ca(2+)](i) and Ca(2+) sensitivity. Topics: Animals; Atrial Natriuretic Factor; Calcium; Cyclic GMP; Enzyme Activation; Female; Guanylate Cyclase; In Vitro Techniques; Isometric Contraction; Male; Muscle Relaxation; Muscle, Smooth; Nitric Oxide; Nitric Oxide Donors; Respiratory System; Spectrometry, Fluorescence; Swine; Thionucleotides; Trachea; Vasodilator Agents | 2002 |
Effects of nitric oxide donors and inhibitors of nitric oxide signalling on endothelin- and serotonin-induced contractions in human placental arteries.
In order to explore the role of nitric oxide (NO) in the control of fetoplacental vascular tone in normal pregnancy we have examined the effects of NO donors on uteroplacental arteries pre-contracted with the vasoconstrictor endothelin-1 (ET-1) or serotonin (5-HT). We have furthermore examined the effects of guanylate cyclase inhibitors on the NO-induced relaxation. Segments of placental arteries (n=102) obtained from 39 placentas immediately after delivery were mounted in organ baths and superfused with Krebs-Ringer solution at 37 degrees C. The vessel segments were exposed to drugs for various intervals and the tension was recorded isometrically and registered on a polygraph. Cyclic guanosine monophosphate (cGMP) analysis was performed after extraction of vessel segments using a specific radioimmunoassay. The placental artery segments responded to ET-1 and 5-HT with a dose-dependent vasoconstriction. After pre-contraction with ET-1 (10(-7) M) or 5-HT (10(-6) M), the vessels relaxed in response to the NO donors glyceryltrinitrate (GTN) (10(-6) M) and S-nitroso-N-acetyl-penicillamine (SNAP) (10(-5) M). In the presence of the non-specific guanylate cyclase inhibitor LY 83583 (10(-6) M), the vessels responded with a small contraction. In the presence of the soluble guanylate cyclase (sGC) inhibitor 1H[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ) the non-treated vessels responded with a relaxation. 1H[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one gave no obvious relaxation in pre-contracted vessels. Addition of 8-Br-cGMP, the cell-permeant analogue of cGMP, with or without pre-contraction had no effect on the vessels. Cyclic guanosine monophosphate analysis showed that GTN treatment caused an increase in cGMP after 12 min. Our results indicate that NO acts as a vasodilator in placental vessels. The cGMP-dependent mechanisms may be involved in NO-induced relaxation but cGMP-independent mechanisms appear also to be involved. Topics: Adult; Aminoquinolines; Cyclic GMP; Dose-Response Relationship, Drug; Endothelin-1; Female; Humans; Muscle, Smooth, Vascular; Nitric Oxide; Nitric Oxide Donors; Nitroglycerin; Placenta; Placental Circulation; Pregnancy; S-Nitroso-N-Acetylpenicillamine; Serotonin; Signal Transduction; Superoxide Dismutase; Thionucleotides; Vasoconstriction; Vasoconstrictor Agents; Vasodilation | 2002 |
Inhibition of antigen-specific T cell proliferation and cytokine production by protein kinase A type I.
cAMP inhibits biochemical events leading to T cell activation by triggering of an inhibitory protein kinase A (PKA)-C-terminal Src kinase pathway assembled in lipid rafts. In this study, we demonstrate that activation of PKA type I by Sp-8-bromo-cAMPS (a cAMP agonist) has profound inhibitory effects on Ag-specific immune responses in peripheral effector T cells. Activation of PKA type I inhibits both cytokine production and proliferative responses in both CD4(+) and CD8(+) T cells in a concentration-dependent manner. The observed effects of cAMP appeared to occur endogenously in T cells and were not dependent on APC. The inhibition of responses was not due to apoptosis of specific T cells and was reversible by a PKA type I-selective cAMP antagonist. This supports the notion of PKA type I as a key enzyme in the negative regulation of immune responses and a potential target for inhibiting autoreactive T cells. Topics: Antigen-Presenting Cells; Antigens, Bacterial; Apoptosis; CD4-Positive T-Lymphocytes; CD8-Positive T-Lymphocytes; Cyclic AMP; Cyclic AMP-Dependent Protein Kinase Type II; Cyclic AMP-Dependent Protein Kinases; Cyclic GMP; Cytokines; Dinoprostone; Down-Regulation; Enterotoxins; Enzyme Activation; Epitopes, T-Lymphocyte; Humans; Interferon-gamma; Interleukin-2; Lymphocyte Activation; Protein-Tyrosine Kinases; Proto-Oncogene Proteins pp60(c-src); Thionucleotides; Tumor Necrosis Factor-alpha | 2002 |
Stimulation of nitric oxide-cGMP pathway excites striatal cholinergic interneurons via protein kinase G activation.
Conflicting data have been collected so far on the action of nitric oxide (NO) on cholinergic interneurons of the striatum. In the present in vitro electrophysiological study, we reported that intracellularly recorded striatal cholinergic interneurons are excited by both hydroxylamine and S-nitroso-N-acetylpenicillamine, two NO donors. This excitation persisted unchanged in the presence of glutamate, dopamine, and substance P receptor antagonists as well as after blockade of tetrodotoxin (TTX)- and calcium channel-sensitive transmitter release, suggesting that NO produces its effects by modulating directly resting ion conductances in the somatodendritic region of striatal cholinergic cells. The depolarizing effect of hydroxylamine was greatly reduced by lowering external concentrations of sodium ions (from 126 to 38 mm) and did not reverse polarity in the voltage range from -120 to -40 mV. The sodium transporter blockers bepridil and 3',4'-dichlorobenzamil were conversely ineffective in preventing NO-induced membrane depolarization. Intracellular cGMP elevation is required for the action of hydroxylamine on striatal cholinergic cells, as demonstrated by the findings that the membrane depolarization produced by this pharmacological agent was prevented by bath and intracellular application of two inhibitors of soluble guanylyl cyclase and was mimicked and occluded by zaprinast, a cGMP phosphodiesterase inhibitor. Finally, intracellular Rp-8-Br-cGMPS, a protein kinase G (PKG) inhibitor, blocked the hydroxylamine-induced membrane depolarization of cholinergic interneurons, whereas both okadaic acid and calyculin A, two protein phosphatase inhibitors, enhanced it, indicating that intracellular PKG and phosphatases oppositely regulate the sensitivity of striatal cholinergic interneurons to NO. The characterization of the cellular mechanisms involved in the regulation of striatal interneuron activity is a key step for the understanding of the role of these cells in striatal microcircuitry. Topics: 3',5'-Cyclic-GMP Phosphodiesterases; Acetylcholine; Animals; Corpus Striatum; Cyclic GMP; Cyclic GMP-Dependent Protein Kinases; Electric Stimulation; Enzyme Inhibitors; Excitatory Amino Acid Antagonists; In Vitro Techniques; Interneurons; Male; Membrane Potentials; Neurotransmitter Agents; Nitric Oxide; Nitric Oxide Donors; Nitric Oxide Synthase; Patch-Clamp Techniques; Phosphoprotein Phosphatases; Rats; Rats, Wistar; Sodium; Tetrodotoxin; Thionucleotides | 2001 |
The induction of carbon monoxide-mediated airway relaxation by PACAP 38 in isolated guinea pig airways.
Pituitary adenylate cyclase--activating peptide 38 (PACAP 38) displays several biologic activities relevant to obstructive airway disease. Carbon monoxide (CO) has recently emerged as a potent, endogenously produced mediator of bronchodilation. In this study, we have analyzed the occurrence of PACAP 38 and the corresponding occurrence of heme oxygenase (HO), the rate-limiting enzyme for CO production, in guinea pig trachea, using immunocytochemistry. We have also investigated whether the dilatory effects of PACAP 38 are dependent on CO, using an in vitro setup for tracheal studies. A moderate supply of PACAP-like immunoreactive nerve fibers was seen in association with tracheal smooth muscle. HO-like immunoreactivity was observed in the respiratory epithelium and in association with smooth muscle bundles. PACAP 38 induced a concentration-dependent relaxation of precontracted tracheal segments. This dilation was nearly abolished after pretreatment with zincprotoporphyrine, an inhibitor of heme oxygenase. The same effect was accomplished with Rp-8Br-cyclicGMPS, an inhibitor of cyclicGMP, whereas the nitric oxide synthase inhibitor N(G)-monomethyl-L-arginine had no effect on the PACAP 38--induced dilation. The presented data suggest that PACAP 38 can induce bronchodilation by means of a CO-dependent, cyclicGMP-related mechanism, thereby providing a link between neurotransmission and local CO release in the airway smooth muscle. Topics: Animals; Carbon Monoxide; Cyclic GMP; Guinea Pigs; Heme Oxygenase (Decyclizing); Male; Muscle Relaxation; Muscle, Smooth; Neuropeptides; Pituitary Adenylate Cyclase-Activating Polypeptide; Thionucleotides; Trachea | 2001 |
Mechanism of a nitric oxide donor NOR 1-induced relaxation in longitudinal muscle of rat proximal colon.
We previously suggested that nitric oxide (NO)-mediated relaxation of the rat proximal colon is not associated with change in cyclic GMP content. We further studied the intracellular mechanism of NO-induced relaxation by measuring changes in tension and intracellular Ca2+ concentration ([Ca2+]i), simultaneously. NOR 1, NO donor, relaxed the longitudinal muscle of the rat proximal colon, which was precontracted by carbachol, with a concomitant decrease in [Ca2+]. ODQ, an inhibitor of soluble guanylate cyclase, partially inhibited the relaxant effect of only higher concentrations of NOR 1, but Rp-8-Br-cGMPS, an inhibitor of cyclic GMP-dependent protein kinase (PKG), did not have any effects on the relaxant effect of NOR 1. When the preparations were transferred to normal solution after the treatment with thapsigargin, an inhibitor of sarcoplasmic reticulum (SR) Ca2+-ATPase, in the absence of Ca2+, contraction with a concomitant increase in [Ca2+]i occurred. NOR 1 did not show significant effects on the tension and [Ca2+]i in thapsigargin-treated preparations. In high K+-precontracted preparations, NOR 1 relaxed the preparations with a slight change in [Ca2+]i. The relaxant effect was significantly inhibited by ODQ and Rp-8-Br-cGMPS. These results suggest that NO induces the relaxation preferentially by acting thapsigargin-sensitive function of SR and in turn decreasing [Ca2+]i, although a cyclic GMP-PKG pathway is suggested under the experimental conditions of a high K+ concentration. Topics: Animals; Calcium; Carbachol; Colon; Culture Techniques; Cyclic GMP; DNA-Binding Proteins; Dose-Response Relationship, Drug; Male; Models, Biological; Muscle Contraction; Muscle Relaxation; Muscle, Skeletal; Nerve Tissue Proteins; Nitric Oxide; Nitric Oxide Donors; Oxadiazoles; Quinoxalines; Rats; Rats, Wistar; Thapsigargin; Thionucleotides | 2001 |
p11 expression in human bronchial epithelial cells is increased by nitric oxide in a cGMP-dependent pathway involving protein kinase G activation.
The effect of nitric oxide on p11 expression was studied in an immortalized human bronchial epithelial cell line (BEAS-2B cells). Three nitric oxide donors were used: spermine NONOate (SP), (+/-)-S-nitroso-N-acetylpenicillamine (SNAP), and S-nitrosoglutathione (SNOG). All three nitric oxide donors had similar effects resulting in dose-dependent and time-dependent accumulation of p11 protein and an increase of steady-state p11 mRNA. Studies using a reporter gene containing the region from -1499 to +89 of the p11 promoter demonstrated an increase in transcriptional activity after stimulation with NO donors for 4 h. These effects were abolished at the promoter and protein level using protein kinase G inhibitors (KT5823 and R(p)-8-pCPT-cGMPS). Incubation of transfected cells with a cell permeable cGMP analogue (8-Br-cGMP) resulted in a dose-related increase of promoter activity. An electrophoretic mobility shift assay of nuclear proteins extracted from BEAS-2B cells identified an AP-1 site located at -82 to -77 of the p11 promoter region as an NO- and cGMP- dependent response element. These data were confirmed using a c-jun dominant negative mutant vector and a c-jun expression plasmid. Therefore, we conclude that nitric oxide-induced p11 expression in human bronchial epithelial cells is mediated at least in part through increased binding of activator protein one to the p11 promoter. Topics: Alkaloids; Annexin A2; Bronchi; Calcium-Binding Proteins; Carbazoles; Cell Nucleus; Chloramphenicol O-Acetyltransferase; Cyclic GMP; Cyclic GMP-Dependent Protein Kinases; Dose-Response Relationship, Drug; Enzyme Activation; Enzyme Inhibitors; Epithelial Cells; Genes, Dominant; Genes, jun; Genes, Reporter; Immunoblotting; Indoles; Mutation; Nitric Oxide; Nitrogen Oxides; Plasmids; Promoter Regions, Genetic; Protein Binding; RNA, Messenger; S100 Proteins; Spermine; Thionucleotides; Time Factors; Transcription Factor AP-1; Transfection | 2001 |
Role of adenosine A(2B) receptors in vasodilation of rat pial artery and cerebral blood flow autoregulation.
This study was aimed to investigate the underlying mechanism of vasodilation induced by the activation of A(2B) adenosine receptors in relation to cerebral blood flow (CBF) autoregulation. Changes in pial arterial diameters were observed directly through a closed cranial window. N(omega)-nitro-L-arginine methyl ester (L-NAME, nitric oxide synthase inhibitor) significantly suppressed the concentration-dependent vasodilations induced by adenosine and 5'-N-ethylcarboxamido-adenosine (NECA) but not the vasodilation by CGS-21680 (A(2A)-receptor agonist). Moreover, NECA-induced vasodilation was suppressed by alloxazine (1 micromol/l) but not by ZM-241385 (1 micromol/l, A(2A) antagonist), which suggests mediation by A(2B)- receptor activation. Otherwise, the level of nitrite/nitrate was concentration dependently increased in the artificial cerebrospinal fluid (CSF) when adenosine and NECA were suffused over the cortical surface. L-NAME and alloxazine, but not ZM-241385, largely inhibited their releases. The lower limit of CBF autoregulation was little affected following pretreatment with L-NAME or alloxazine. Thus it is suggested that adenosine-induced vasodilation via activation of A(2B)-adenosine receptors of the rat pial artery is coupled to the production of nitric oxide, which contributes little to CBF autoregulation. Topics: Animals; Arteries; Cerebrovascular Circulation; Cyclic AMP; Cyclic GMP; Enzyme Inhibitors; Flavins; Homeostasis; Male; NG-Nitroarginine Methyl Ester; Nitrates; Nitrites; Pia Mater; Rats; Rats, Sprague-Dawley; Receptor, Adenosine A2B; Receptors, Purinergic P1; Thionucleotides; Triazines; Triazoles; Vasodilation | 2000 |
Nitric oxide/cyclic GMP pathway attenuates ATP-evoked intracellular calcium increase in supporting cells of the guinea pig cochlea.
We demonstrate here that nitric oxide (NO) attenuates ATP-evoked calcium transients in Deiters' and Hensen's cells, "supporting" (nonsensory) cells of the guinea pig cochlea, by means of activation of soluble guanylyl cyclase and protein kinase G. The enzymatic activities associated with the nitric oxide/cGMP/protein kinase G pathway had previously been demonstrated to be present in Deiters' and Hensen's cells. We now isolate these cells and measure changes in intracellular free calcium by using the calcium indicator fluo-3. In Deiters' cells, calcium increased rapidly in response to the application of ATP. The increase was attenuated when the pathway was stimulated by NO donors (diethylamine NONOate or sodium nitroprusside) or the cyclic GMP analog, 8-bromo-cyclic GMP. When the activation of the pathway was blocked by the additional presence of inhibitors of soluble guanylyl cyclase (LY83583) or protein kinase G (Rp-8-bromo-cyclic GMP or KT5823), the response to ATP was restored. The reactions also occurred in calcium-free media. Hensen's cells responded similarly. These results provide evidence that intracellular calcium is regulated by the NO/cGMP/protein kinase G pathway in the inner ear. Topics: 8-Bromo Cyclic Adenosine Monophosphate; Adenosine Triphosphate; Alkaloids; Animals; Biological Transport; Calcium; Carbazoles; Cell Survival; Cochlea; Cyclic GMP; Cyclic GMP-Dependent Protein Kinases; Enzyme Inhibitors; Guanylate Cyclase; Guinea Pigs; Homeostasis; In Vitro Techniques; Indoles; Nitric Oxide; Protein Kinase Inhibitors; Protein Kinases; Thionucleotides | 2000 |
Nitric oxide provokes tumor necrosis factor-alpha expression in adult feline myocardium through a cGMP-dependent pathway.
The mechanism(s) responsible for the persistent coexpression of tumor necrosis factor-alpha (TNF-alpha) and nitric oxide (NO) in the failing heart is unknown.. To determine whether NO was sufficient to provoke TNF-alpha biosynthesis, we examined the effects of an NO donor, S-nitroso-N-acetyl penicillamine (SNAP), in buffer-perfused Langendorff hearts. SNAP (1 micromol/L) treatment resulted in a time- and dose-dependent increase in myocardial TNF-alpha mRNA and protein biosynthesis in adult cat hearts. The effects of SNAP were completely abrogated by a NO quenching agent, 2-(4-carboxyphenyl)-4, 4,5,5-tetramethylimidazoline-1-oxyl 3-oxide (C-PTIO), and mimicked by sodium nitroprusside. Electrophoretic mobility shift assays demonstrated that SNAP treatment led to the rapid induction of nuclear factor kappa-beta (NF-kappaB) but not AP-1. The importance of the cGMP pathway in terms of mediating NO-induced TNF-alpha biosynthesis was shown by studies that demonstrated that 8-bromo-cGMP mimicked the effects of SNAP and that the effects of SNAP could be completely abrogated using a cGMP antagonist, 1H-(1,2, 4)oxadiazolo(4,3-a)quinoxalin-1-one (ODQ), or protein kinase G antagonist (Rp-8-Br-cGMPS). SNAP and 8-Br-cGMP were both sufficient to lead to the site-specific phosphorylation (serine 32) and degradation of IkappaBalpha in isolated cardiac myocytes. Finally, protein kinase G was sufficient to directly phosphorylate IkappaBalpha on serine 32, a critical step in the activation of NF-kappaB.. These studies show that NO provokes TNF-alpha biosynthesis through a cGMP-dependent pathway, which suggests that the coincident expression of TNF-alpha and NO may foster self-sustaining positive autocrine/paracrine feedback inflammatory circuits within the failing heart. Topics: Animals; Benzoates; Cats; Cyclic GMP; Cyclic GMP-Dependent Protein Kinases; DNA-Binding Proteins; Electrophoresis; I-kappa B Proteins; Imidazoles; In Vitro Techniques; Myocardium; NF-kappa B; NF-KappaB Inhibitor alpha; Nitric Oxide; Nitric Oxide Donors; Nitroprusside; Oxadiazoles; Penicillamine; Phosphorylation; Protein Kinases; Quinoxalines; RNA, Messenger; Thionucleotides; Tumor Necrosis Factor-alpha | 2000 |
Neurokinin A inhibits oxytocin and GABA release from the posterior pituitary by stimulating nitric oxide synthase.
Neurokinin A (NKA) is a tachykinin that participates in the control of neuroendocrine functions. The posterior pituitary lobe (PP) contains abundant nitric oxide synthase (NOS), suggesting that nitric oxide (NO) may play a role in controlling the release of neuropeptides and neurotransmitters. In the present project, we investigated the in vitro effect of NKA on oxytocin release from hypothalamic explants and PP of male rats and the possible involvement of NO in the action of NKA. Since NKA inhibits gamma-aminobutyric acid (GABA) release from PP, we also examined the role of NO in the effect of NKA on basal and K(+)-evoked GABA release. NKA (10(-7)-10(-5) M) significantly decreased oxytocin release from PP, whereas it did not affect its release from hypothalamic explants. The inhibitory effect of NKA on oxytocin release from PP was completely blocked by the NOS inhibitors N(G)-monomethyl-L-arginine (L-NMMA, 0.5 mM) or N(G)-nitro-L-arginine-methyl-ester (L-NAME, 1 mM). Sodium nitroprusside (0.5 mM), an NO releaser, had no effect on basal GABA release but significantly decreased K(+)-evoked GABA release. L-NMMA (0.3 mM) and L-NAME (0.5 mM) increased K(+)-evoked GABA release, indicating that NO plays an inhibitory role in GABA release from PP. The inhibition in both basal and K(+)-evoked GABA release induced by NKA (10(-7) M) was reduced by L-NAME (1 mM). Also, NKA (10(-7) M) increased NO synthesis as measured by [(14)C] citrulline production. Considered all together, our data indicate that NO may mediate the inhibitory effect of NKA on the release of both oxytocin and GABA from PP. Topics: Animals; Cyclic GMP; Dose-Response Relationship, Drug; Enzyme Inhibitors; gamma-Aminobutyric Acid; Hypothalamus; In Vitro Techniques; Male; Neurokinin A; NG-Nitroarginine Methyl Ester; Nitric Oxide; Nitric Oxide Synthase; omega-N-Methylarginine; Oxytocin; Pituitary Gland, Posterior; Potassium; Rats; Rats, Wistar; Thionucleotides | 2000 |
Effects of guanylyl cyclase and protein kinase G inhibitors on vasodilatation in non-tolerant and tolerant bovine coronary arteries.
The effects in bovine coronary arteries of the soluble guanylyl cyclase inhibitor 1 H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ) were examined in order to establish the relative importance of the enzyme (a) in the vasodilator actions of glyceryl trinitrate and S-nitroso-N-acetylpenicillamine and (b) in induction of tolerance to these agents. ODQ strongly inhibited responses to both relaxants with IC50's of the order of 0.5 microM; in contrast, the protein kinase G inhibitor, 8-bromoguanosine-3',5'-monophosphorothioate (Rp-8-Br-cGMPS) had little effect on the responses. Tolerance after pre-incubation with glyceryl trinitrate (10 microM) was unaffected by co-pre-incubation with ODQ (1.0 microM), but similar experiments with S-nitroso-N-acetylpenicillamine were inconclusive because tolerance was associated with depressed contractile activity. It is concluded that in bovine coronary arteries soluble guanylyl cyclase is essential for vasorelaxation to both glyceryl trinitrate and S-nitroso-N-acetylpenicillamine but is unimportant for induction of tolerance to glyceryl trinitrate. Our results add weight to the hypothesis of impaired biotransformation rather than guanylyl cyclase desensitisation as the mechanism of in vitro nitrate tolerance. Topics: Animals; Cattle; Coronary Vessels; Cyclic GMP; Cyclic GMP-Dependent Protein Kinases; Drug Interactions; Drug Tolerance; Enzyme Inhibitors; Guanylate Cyclase; Muscle, Smooth, Vascular; Nitroglycerin; Oxadiazoles; Penicillamine; Protein Kinase Inhibitors; Quinoxalines; S-Nitroso-N-Acetylpenicillamine; Thionucleotides; Vasodilation; Vasodilator Agents | 1999 |
Natriuretic peptides and cGMP modulate K+, Na+, and H+ fluxes in Zea mays roots.
Recent evidence suggests that in plants, as in vertebrates, natriuretic peptides (NPs) regulate homeostasis. In this study noninvasive ion-selective vibrating microelectrodes were used to measure net fluxes of K+, Na+, and H+ in Zea mays root conductive tissue. Immunoreactant plant natriuretic peptides (irPNP) cause immediate net H+ influx and delayed net K+ and Na+ uptake. Delayed net K+ influx was also observed in response to 8-Br-cGMP, however, not accompanied by significant changes in net H+ fluxes. Furthermore, 8-Br-cGMP does not stimulate the plasma membrane H+-ATPase implying that cGMP directly affects cation channels. The data are consistent with NP and cGMP-dependent stimulation of nonselective cation channels with P(K) > P(Na) and point to a complex role for NPs in plant homeostasis. Topics: Affinity Labels; Animals; Atrial Natriuretic Factor; Cations, Monovalent; Cyclic GMP; Electrophoresis, Polyacrylamide Gel; Hydrogen; Immunoblotting; Models, Biological; Peptides; Plant Proteins; Plant Roots; Potassium; Rats; Sodium; Thionucleotides; Zea mays | 1999 |
Natriuretic peptides regulate the expression of tissue factor and PAI-1 in endothelial cells.
In the present study, we demonstrate that brain natriuretic peptide (BNP) and C-type natriuretic peptide (CNP) interact with angiotensin II (Ang II) in regulative blood coagulation and fibrinolysis by suppressing the expressions of both tissue factor (TF) and plasminogen activator inhibitor-1 (PAI-1) induced by Ang II. The expressions of TF and PAI-1 mRNA were analyzed by northern blotting methods, and the activities of TF on the surface of rat aortic endothelial cells (RAECs) and PAI-1 in the culture media were respectively measured by chromogenic assay. Both BNP and CNP suppressed the expressions of TF and PAI-1 mRNA induced by Ang II in a time- and concentration-dependent manner via cGMP cascade, which suppressions were accompanied by respective decrease in activities of TF and PAI-1. However, neither the expression of tissue factor pathway inhibitor (TFPI) nor tissue-type plasminogen activator (TPA) mRNA was affected by the treatment of BNP and CNP. Topics: Angiotensin II; Animals; Blotting, Northern; Cells, Cultured; Culture Media, Conditioned; Cyclic GMP; Endothelium, Vascular; Gene Expression Regulation; Lipopolysaccharides; Lipoproteins; Male; Natriuretic Peptide, Brain; Natriuretic Peptide, C-Type; Plasminogen Activator Inhibitor 1; Rats; Rats, Sprague-Dawley; RNA, Messenger; Second Messenger Systems; Thionucleotides; Thrombin; Thromboplastin; Tissue Plasminogen Activator | 1999 |
Effect of cGMP inhibitors on the actions of nitrodilators in rat aorta.
1. The involvement of cGMP in vasodilatation produced by a range of nitrodilators was investigated using two different protein kinase G inhibitors, R(p) 8-bromoguanosine-3'5'-cyclic monophosphothioate (RBrcGMPS) and KT 5823. 2. The nitric oxide donors sodium nitroprusside (SNP), glyceryltrinitrate (GTN) and s-nitroso-acetylpenicillamine (SNAP), the endothelium-dependent vasodilator acetylcholine (ACh) as well as the cGMP analogues 8-(4-chlorophenylthio)-cGMP(CPTcGMP) and beta-phenyl-1-N2-etheno-8-bromo-cGMP (PETcGMP) all relaxed rat aortic rings preconstricted with phenylephrine (0.1 micromol/L). 3. The protein kinase G inhibitor KT 5823 (10 micromol/L) produced a very small inhibition of the vasodilatation produced by GTN, but had no effect against vasodilatation produced by SNP, CPTcGMP or PETcGMP, which suggests that KT 5823 is not a useful tool in this system. 4. In contrast, RBrcGMPS (0.5 mmol/L) produced a rightward shift of the concentration-response curves to SNP, CPTcGMP and PETcGMP. RBrcGMPS (0.5 mmol/L) also completely abolished vasodilatation to ACh and GTN but, surprisingly, had no effect on vasodilatation produced by SNAP. 5. The guanylate cyclase inhibitor 1H-[1,2,4] oxadiazolo[4,3-a]quinoxalin-1-one (ODQ; 1 and 10 micromol/L) completely inhibited the relaxation produced by GTN, whereas SNAP still had an appreciable relaxant effect after ODQ (1 micromol/L). 6. The differential effect of RBrcGMPS and ODQ on the nitrodilators suggests that there are differences in the mechanism of dilatation between the nitrodilators. Topics: Alkaloids; Animals; Aorta, Thoracic; Carbazoles; Cyclic GMP; Cyclic GMP-Dependent Protein Kinase Type I; Cyclic GMP-Dependent Protein Kinases; Guanylyl Imidodiphosphate; Indoles; Male; Nitroprusside; Rats; Rats, Sprague-Dawley; Thionucleotides; Vasodilator Agents | 1998 |
24R,25-dihydroxyvitamin D3 increases cyclic GMP contents, leading to an enhancement of osteocalcin synthesis by 1,25-dihydroxyvitamin D3 in cultured human osteoblastic cells.
The effect of the physiological vitamin D metabolite 24R, 25-dihydroxyvitamin D3 [24R,25(OH)2D3] on human osteoblastic cells was assessed. Physiological concentrations (10(-9)-10(-8) M) of 24R, 25(OH)2D3 significantly increased the cyclic guanosine 5'-monophosphate (cGMP) content in the human osteoblastic cells by approximately 200% in 5 to 15 min. In contrast, 24S, 25-dihydroxyvitamin D3 had only a weak effect on the cGMP content, and 1,25-dihydroxyvitamin D3 [1,25(OH)2D3] did not affect the content. The production of osteocalcin was not induced by 10(-9)-10(-8) M of 24R,25(OH)2D3 in the absence of 1,25(OH)2D3. However, the same concentration of 24R,25(OH)2D3 showed stimulatory effects on osteocalcin synthesis in the presence of 10(-9) M 1, 25(OH)2D3. Rp-8Br-cyclic GMP, a specific inhibitor of cyclic GMP-dependent protein kinase, significantly inhibited the cooperative effect of 24R,25(OH)2D3 with 1,25(OH)2D3 on the osteocalcin synthesis, although Rp-8Br-cyclic AMP, a specific inhibitor of cyclic AMP-dependent protein kinase, did not affect the cooperative effect. In addition, okadaic acid enhanced the osteocalcin synthesis induced by 1,25(OH)2D3. These observations suggest that 24R,25(OH)2D3 has a unique activity of increasing cGMP contents in osteoblastic cells, and that the increase in cGMP contents may lead to the cooperative effect of 24R,25(OH)2D3 with 1, 25(OH)2D3 on osteocalcin synthesis. These data support the hypothesis that 24R,25(OH)2D3 has a physiological role in human bone and mineral metabolism. Topics: 24,25-Dihydroxyvitamin D 3; 8-Bromo Cyclic Adenosine Monophosphate; Calcitriol; Cells, Cultured; Cyclic GMP; Drug Synergism; Humans; Okadaic Acid; Osteoblasts; Osteocalcin; Thionucleotides | 1998 |
Evidence that additional mechanisms to cyclic GMP mediate the decrease in intracellular calcium and relaxation of rabbit aortic smooth muscle to nitric oxide.
1. The role of cyclic GMP in the ability of nitric oxide (NO) to decrease intracellular free calcium concentration [Ca2+]i and divalent cation influx was studied in rabbit aortic smooth muscle cells in primary culture. In cells stimulated with angiotensin II (AII, 10(-1) M), NO (10(-10) - 10(-6) M) increased cyclic GMP levels measured by radioimmunoassay and decreased [Ca2+]i and cation influx as indicated by fura-2 fluorimetry. 2. Zaprinast (10(-4) M), increased NO-stimulated levels of cyclic GMP by 3-20 fold. Although the phosphodiesterase inhibitor lowered the level of [Ca2+]i reached after administration of NO, the initial decreases in [Ca2+]i initiated by NO were not significantly different in magnitude or duration from those that occurred in the absence of zaprinast. 3. The guanylyl cyclase inhibitor, H-(1,2,4) oxadiazolo(4,3-a) quinoxallin-1-one (ODQ, 10(-5) M), blocked cyclic GMP accumulation and activation of protein kinase G, as measured by back phosphorylation of the inositol trisphosphate receptor. ODQ and Rp-8-Br-cyclic GMPS, a protein kinase G inhibitor, decreased the effects of NO, 10(-10) - 10(-8) M, but the decrease in [Ca2+]i or cation influx caused by higher concentrations of NO (10(-7) - 10(-6) M) were unaffected. Relaxation of intact rabbit aorta rings to NO (10(-7) - 10(-5) M) also persisted in the presence of ODQ without a significant increase in cyclic GMP. Rp-8-Br-cyclic GMPS blocked the decreases in cation influx caused by a cell permeable cyclic GMP analog, but ODQ and/or the protein kinase G inhibitor had no significant effect on the decrease caused by NO. 4. Although inhibitors of cyclic GMP, protein kinase G and phosphodiesterase can be shown to affect the decrease in [Ca2+]i and cation influx via protein kinase G, these studies indicate that when these mechanisms are blocked, cyclic GMP-independent mechanisms also contribute significantly to the decrease in [Ca2+]i and smooth muscle relaxation to NO. Topics: 1-Methyl-3-isobutylxanthine; 3',5'-Cyclic-GMP Phosphodiesterases; Angiotensin II; Animals; Aorta; Calcium; Cyclic GMP; Dose-Response Relationship, Drug; Enzyme Inhibitors; Guanylate Cyclase; Manganese; Muscle Relaxation; Muscle, Smooth; Nitric Oxide; Oxadiazoles; Phosphodiesterase Inhibitors; Protein Kinase C; Purinones; Quinoxalines; Rabbits; Thionucleotides; Vasoconstrictor Agents; Vasodilator Agents | 1998 |
cGMP-kinase mediates cGMP- and cAMP-induced Ca2+ desensitization of skinned rat artery.
(Rp)-8-Bromo-guanosine 3',5'-cyclic monophosphorothioate (Rp-8-Br-cGMPS) inhibited competitively both isozymes of type I alpha and I beta cGMP-dependent protein kinase (cGMP-kinase) purified from porcine aorta with apparent Ki values (microM) of 3.7 for I alpha and 1.8 for I beta. The compound also inhibited bovine heart type II cAMP-dependent protein kinase (cAMP-kinase), but with a Ki of 25 microM. Thus, it is a selective inhibitor of cGMP-kinase. In alpha-toxin-skinned smooth muscle preparations from rat mesenteric artery, 8-Br-cGMP (10(-7) M) and 8-Br-cAMP (10(-6) M) produced a rightward shift of the concentration-contraction curves for Ca2+, denoting a decrease in Ca2+ sensitivity of the contractile elements. The shift by 8-Br-cAMP as well as by 8-Br-cGMP was completely reversed by Rp-8-Br-cGMPS, while a selective inhibitor of activation of cAMP-kinase, (Rp)-adenosine-3',5'-cyclic monophosphorothioate (Rp-cAMPS), was without effects on the shift produced by these two compounds. These findings indicate the pivotal role that the activation of cGMP-kinase plays in the production of a decrease in Ca2+ sensitivity of contractile elements. Topics: 8-Bromo Cyclic Adenosine Monophosphate; Analysis of Variance; Animals; Aorta; Calcium; Cattle; Cyclic AMP; Cyclic AMP-Dependent Protein Kinase Type II; Cyclic AMP-Dependent Protein Kinases; Cyclic GMP; Cyclic GMP-Dependent Protein Kinases; Dose-Response Relationship, Drug; Enzyme Activation; Heart; Isoenzymes; Male; Mesenteric Arteries; Muscle Contraction; Muscle, Smooth, Vascular; Myocardium; Rats; Rats, Wistar; Swine; Thionucleotides; Type C Phospholipases | 1997 |
Involvement of cyclic nucleotide-dependent protein kinases in cyclic AMP-mediated vasorelaxation.
1. The involvement of cyclic AMP-dependent protein kinase (PKA) and cyclic GMP-dependent protein kinase (PKC) in the effects of cyclic AMP-elevating agents on vascular smooth muscle relaxation, cyclic nucleotide dependent-protein kinase activities and ATP-induced calcium signalling ([Ca2+]i was studied in rat aorta. Cyclic AMP-elevating agents used were a beta-adrenoceptor agonist (isoprenaline), a phosphodiesterase 3 (PDE3) inhibitor (SK&F 94120) and a PDE4 inhibitor (rolipram). 2. In rat intact aorta, the relaxant effect induced by isoprenaline (0.01-0.03 microM) was decreased by a specific inhibitor of PKA, H-89, whereas a specific inhibitor of PKG, Rp-8-Br-cyclic GMPs, was without effect. NO significant difference in PKA and PKG activity ratios was detected in aortic rings when isoprenaline 10 microM was used. At the same concentration, isoprenaline did not modify ATP-induced changes in [Ca2+]i in smooth muscle cells. Neither H-89 nor Rp-8-Br-cyclic GMPs modified this response. These findings suggest that PKA is only involved in the relaxant effect induced by low concentrations of isoprenaline (0.01-0.3 microM), whereas for higher concentrations, other mechanisms independent of PKA and PKG were involved. 3. The relaxant effects induced by SK&F 94120 and rolipram were inhibited by Rp-8-Br-cyclic GMPS with no significant effect of H-89. Neither SK&F 94120, nor rolipram at 30 microM significantly modified the activity ratios of PKA and PKG. Rolipram inhibited the ATP-induced transient increase in [Ca2+]i. This decrease was abolished by Rp-8-Br-cyclic GMPS whereas H-89 had no significant effect. These results suggests that PKG is involved in the vascular effects induced by the inhibitors of PDE3 and PDE4. Moreover, since it was previously shown that PDE3 and PDE4 inhibitors only increased cyclic AMP levels with no change in cyclic GMP level, these data also suggest a cross-activation of PKG by cyclic AMP in rat aorta. 4. The combinations of 5 microM SK&F 94120 with rolipram markedly potentiated the relaxant effect of rolipram. This relaxation was decreased by H-89 and not significantly modified by Rp-8-Br-cyclic GMPS. Moreover, the association of the two PDE inhibitors significantly increased the activity ratio of PKA without changing the PKG ratio. The present findings show that PKA rather than PKG is involved in this type of vasorelaxation. The differences in the participation of PKA vs PKG observed when inhibitors of PDE3 and PDE4 were used alone or to Topics: Adenosine Triphosphate; Adrenergic beta-Agonists; Animals; Aorta; Calcium; Cardiotonic Agents; Cyclic AMP; Cyclic AMP-Dependent Protein Kinases; Cyclic GMP; Cyclic GMP-Dependent Protein Kinases; Endothelium, Vascular; Enzyme Inhibitors; In Vitro Techniques; Isoproterenol; Isoquinolines; Male; Muscle Relaxation; Muscle, Smooth, Vascular; Phosphodiesterase Inhibitors; Pyrazines; Pyrrolidinones; Rats; Rats, Wistar; Rolipram; Signal Transduction; Sulfonamides; Thionucleotides | 1997 |
Induction of neuroprotective kappa B-dependent transcription by secreted forms of the Alzheimer's beta-amyloid precursor.
A significant fraction of the beta-amyloid precursor protein is proteolytically processed to yield large secreted forms (sAPP). These proteins have pleiotropic effects which potentially involve control of gene expression. We have investigated the influence of sAPP on the class of transcription factors which bind kappa B enhancer sequences. Transcription dependent on a kappa B element was enhanced by sAPP in several cell lines, as measured by expression of a transfected chloramphenicol acetyltransferase reporter gene. Secreted APP also induced an increase in kappa B DNA-binding activity in hippocampal neurons treated with sAPP. Both effects were mimicked by an analog of cyclic GMP and inhibited by an antagonist of cyclic GMP-dependent protein kinase. Such activation of kappa B-dependent transcription was correlated in two ways with the ability of sAPP to protect neuronal cells against calcium-mediated damage: (1) tumor necrosis factor beta also protected against calcium-mediated insults and induced kappa B-dependent transcription; (2) antisense oligonucleotide-mediated reduction of an endogenous inhibitor of NF-kappa B activated kappa B-binding activity and attenuated calcium-mediated toxicity in both a neuronal cell line and in primary neurons. These findings suggest that a kappa B-binding transcription factor can act as a coordinator of neuroprotective gene expression in response to cytokines. Topics: Amyloid beta-Protein Precursor; Base Sequence; Calcimycin; Calcium; Cell Line; Cell Survival; Chloramphenicol O-Acetyltransferase; Cyclic GMP; Cyclic GMP-Dependent Protein Kinases; DNA-Binding Proteins; Enhancer Elements, Genetic; Glioma; Humans; I-kappa B Proteins; Kidney; Kinetics; Neuroblastoma; Neurons; NF-kappa B; NF-KappaB Inhibitor alpha; Oligonucleotides, Antisense; Recombinant Fusion Proteins; Thionucleotides; Transcription, Genetic; Transfection; Tumor Cells, Cultured | 1996 |
The role of nitric oxide in the central control of blood pressure.
In these studies blood pressure responses to intracerebroventricular (i.c.v.) infusions were recorded in anesthetized rats. NO donors caused a fall in blood pressure, whereas L-NAME, which blocks the enzyme (NOS) that produces NO, caused a rise in blood pressure. Calcium, i.c.v., stimulates NOS to lower blood pressure. The depressor action of NO is reduced by blocking the action of cGMP. This central NO/cGMP system is tonically active to maintain blood pressure at a normal level. Topics: Amino Acid Oxidoreductases; Animals; Arginine; Blood Pressure; Calcium Chloride; Cerebral Ventricles; Cyclic GMP; Dose-Response Relationship, Drug; Injections, Intraventricular; Male; NG-Nitroarginine Methyl Ester; Nitric Oxide; Nitric Oxide Synthase; Rats; Rats, Inbred WKY; Thionucleotides | 1995 |
Activity-dependent long-term enhancement of transmitter release by presynaptic 3',5'-cyclic GMP in cultured hippocampal neurons.
Long-term potentiation (LTP) in hippocampus is a type of synaptic plasticity that is thought to be involved in learning and memory. Several lines of evidence suggest that LTP involves 3',5'-cyclic GMP (cGMP), perhaps as an activity-dependent presynaptic effector of one or more retrograde messengers (refs 2-12, but see ref. 13). However, previous results are also consistent with postsynaptic effects of cGMP. This is difficult to test in hippocampal slices, but more rigorous tests are possible in dissociated cell culture. We have therefore developed a reliable method for producing N-methyl-D-aspartate (NMDA) receptor-dependent LTP at synapses between individual hippocampal pyramidal neurons in culture. We report that inhibitors of guanylyl cyclase or of cGMP-dependent protein kinase block potentiation by either tetanic stimulation or low-frequency stimulation paired with postsynaptic depolarization. Conversely, application of 8-Br-cGMP to the bath or injection of cGMP into the presynaptic neuron produces activity-dependent long-lasting potentiation. The potentiation by cGMP involves an increase in transmitter release that is in part independent of changes in the presynaptic action potential. These results support a presynaptic role for cGMP in LTP. Topics: Action Potentials; Aminoquinolines; Animals; Cells, Cultured; Cyclic GMP; Guanylate Cyclase; Long-Term Potentiation; Magnesium; Neurons; Neurotransmitter Agents; Presynaptic Terminals; Pyramidal Cells; Rats; Rats, Sprague-Dawley; Receptors, N-Methyl-D-Aspartate; Synapses; Thionucleotides | 1995 |
Rp-8-Br-guanosine-3',5'-cyclic monophosphorothioate inhibits relaxation elicited by nitroglycerin in rabbit aorta.
To ascertain whether the activation of cyclic GMP-dependent protein kinase is involved in the relaxant effects of nitroglycerin, the effects of Rp-8-Br-guanosine-3',5'-cyclic monophosphorothioate (Rp-8-Br-cGMPS), an inhibitor of activation of G-kinase by cyclic GMP, were studied. In the isolated rabbit aorta contracted by phenylephrine, Rp-8-Br-cGMPS (30 microM) competitively inhibited the relaxation elicited by 8-Br-cGMP, but not that elicited by 8-Br-cyclic AMP, indicating that Rp-8-Br-cGMPS is a specific inhibitor of activation of cyclic GMP-dependent protein kinase by cyclic GMP. The relaxation elicited by nitroglycerin was inhibited by Rp-8-Br-cGMPS. Topics: 8-Bromo Cyclic Adenosine Monophosphate; Animals; Aorta; Cyclic GMP; Cyclic GMP-Dependent Protein Kinases; Enzyme Activation; Male; Muscle Relaxation; Muscle, Smooth, Vascular; Nitroglycerin; Rabbits; Rats; Thionucleotides | 1994 |
Modulation of bradykinin-induced calcium signals by oxidative stress in PC12 cells.
The influence of oxidative stress on agonist-stimulated changes of intracellular free calcium and inositol trisphosphate in the neurosecretory PC12 cell line was investigated. The oxidant H2O2 modulated the bradykinin-induced calcium signal by decreasing the initial peak and the plateau phase in the same manner as tetraphorbolacetate, an activator of protein kinase C. Inositol trisphosphate formation, induced by bradykinin was also decreased by oxidative stress. Thiol protecting agents were able to restore the altered signal. In contrast to this, radical quenching substances had no influence on calcium signals in stressed cells. Inhibitors of several protein kinases, such as protein kinase C, protein kinase A, or cyclic GMP-dependent protein kinase showed the ability to protect the plateau phase of calcium signals against oxidative stress, but not the peak response. These results indicate that under the influence of oxidative stress multiple targets within the signal transduction cascades are affected. Topics: Adenosine Triphosphate; Alkaloids; Animals; Antioxidants; Bradykinin; Calcium; Cyclic AMP; Cyclic GMP; Free Radical Scavengers; Hydrogen Peroxide; Inositol 1,4,5-Trisphosphate; Kinetics; Nickel; PC12 Cells; Protein Kinase C; Signal Transduction; Staurosporine; Tetradecanoylphorbol Acetate; Thionucleotides; Time Factors | 1993 |