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










Publication year range
1.
Anesth Analg ; 133(3): 794-810, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34166321

ABSTRACT

BACKGROUND: Remifentanil can induce postinfusion cold hyperalgesia. N-methyl-d-aspartate receptor (NMDAR) activation and upregulation of transient receptor potential melastatin 8 (TRPM8) membrane trafficking in dorsal root ganglion (DRG) are critical to cold hyperalgesia derived from neuropathic pain, and TRPM8 activation causes NMDAR-dependent cold response. Contribution of P2Y1 purinergic receptor (P2Y1R) activation in DRG to cold pain hypersensitivity and NMDAR activation induced by P2Y1R upregulation in neurons are also unraveled. This study explores whether P2Y1R contributes to remifentanil-induced cold hyperalgesia via TRPM8-dependent regulation of NMDAR phosphorylation in DRG. METHODS: Rats with remifentanil-induced cold hyperalgesia were injected with TRPM8 antagonist or P2Y1R antagonist at 10 minutes before remifentanil infusion. Cold hyperalgesia (paw lift number and withdrawal duration on cold plate) was measured at -24, 2, 6, 24, and 48 hours following remifentanil infusion. After the last behavioral test, P2Y1R expression, TRPM8 expression and membrane trafficking, and NMDAR subunit (NR1 and NR2B) expression and phosphorylation in DRG were detected by western blot, and colocalization of P2Y1R with TRPM8 was determined by double-labeling immunofluorescence. Two-way repeated measures analysis of variance (ANOVA) or 2 × 2 factorial design ANOVA with repeated measures was used to analyze behavioral data of cold hyperalgesia. One-way ANOVA followed by Bonferroni post hoc comparisons was used to analyze the data in western blot and immunofluorescence. RESULTS: Remifentanil infusion (1 µg·kg-1·min-1 for 60 minutes) induced cold hyperalgesia (hyperalgesia versus control, paw lift number and withdrawal duration on cold plate at 2-48 hours, P < .0001) with upregulated NR1 (hyperalgesia versus naive, 48 hours, mean ± standard deviation [SD], 114.00% ± 12.48% vs 41.75% ± 5.20%, P < .005) and NR2B subunits expression (104.13% ± 8.37% vs 24.63% ± 4.87%, P < .005), NR1 phosphorylation at Ser896 (91.88% ± 7.08% vs 52.00% ± 7.31%, P < .005) and NR2B phosphorylation at Tyr1472 (115.75% ± 8.68% vs 59.75% ± 7.78%, P < .005), TRPM8 expression (115.38% ± 9.27% vs 40.50% ± 4.07%, P < .005) and membrane trafficking (112.88% ± 5.62% vs 48.88% ± 6.49%, P < .005), and P2Y1R expression (128.25% ± 14.86% vs 45.13% ± 7.97%, P < .005) in DRG. Both TRPM8 and P2Y1R antagonists attenuated remifentanil-induced cold hyperalgesia and downregulated increased NR1 and NR2B expression and phosphorylation induced by remifentanil (remifentanil + RQ-00203078 versus remifentanil + saline, NR1 phosphorylation, 69.38% ± 3.66% vs 92.13% ± 4.85%; NR2B phosphorylation, 72.25% ± 6.43% vs 111.75% ± 11.00%, P < .0001). NMDAR activation abolished inhibition of TRPM8 and P2Y1R antagonists on remifentanil-induced cold hyperalgesia. P2Y1R antagonist inhibited remifentanil-evoked elevations in TRPM8 expression and membrane trafficking and P2Y1R-TRPM8 coexpression (remifentanil + 2'-deoxy-N6-methyl adenosine 3',5'-diphosphate [MRS2179] versus remifentanil + saline, coexpression, 8.33% ± 1.33% vs 22.19% ± 2.15%, P < .0001). CONCLUSIONS: Attenuation of remifentanil-induced cold hyperalgesia by P2Y1R inhibition is attributed to downregulations in NMDAR expression and phosphorylation via diminishing TRPM8 expression and membrane trafficking in DRG.


Subject(s)
Ganglia, Spinal/metabolism , Hyperalgesia/metabolism , Pain Threshold , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, Purinergic P2Y1/metabolism , TRPM Cation Channels/metabolism , Analgesics/pharmacology , Animals , Behavior, Animal , Cold Temperature , Disease Models, Animal , Ganglia, Spinal/drug effects , Ganglia, Spinal/physiopathology , Hyperalgesia/chemically induced , Hyperalgesia/physiopathology , Hyperalgesia/prevention & control , Male , Pain Threshold/drug effects , Phosphorylation , Protein Transport , Purinergic P2Y Receptor Antagonists/pharmacology , Rats, Sprague-Dawley , Receptors, Purinergic P2Y1/drug effects , Remifentanil , Signal Transduction , TRPM Cation Channels/antagonists & inhibitors
2.
Am J Physiol Heart Circ Physiol ; 320(2): H699-H712, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33306443

ABSTRACT

Brain capillary pericytes have been suggested to play a role in the regulation of cerebral blood flow under physiological and pathophysiological conditions. ATP has been shown to cause constriction of capillaries under ischemic conditions and suggested to be involved in the "no-reflow" phenomenon. To investigate the effects of extracellular ATP on pericyte cell contraction, we studied purinergic receptor activation of cultured bovine brain capillary pericytes. We measured intracellular Ca2+ concentration ([Ca2+]i) responses to purinergic agonists with the fluorescent indicators fura-2 and Cal-520 and estimated contraction of pericytes as relative change in cell area, using real-time confocal imaging. Addition of ATP caused an increase in cytosolic calcium and contraction of the brain capillary pericytes, both reversible and inhibited by the purinergic receptor antagonist pyridoxalphosphate-6-azophenyl-2',4'-disulfonic acid (PPADS). Furthermore, we demonstrated that ATP-induced contraction could be eliminated by intracellular calcium chelation with BAPTA, indicating that the contraction was mediated via purinergic P2-type receptor-mediated [Ca2+]i signaling. ATP stimulation induced inositol triphosphate signaling, consistent with the notion of P2Y receptor activation. Receptor profiling studies demonstrated the presence of P2Y1 and P2Y2 receptors, using ATP, UTP, ADP, and the subtype specific agonists MRS2365 (P2Y1) and 2-thio-UTP (P2Y2). Addition of specific P2X agonists only caused an [Ca2+]i increase at high concentrations, attributed to activation of inositol triphosphate signaling. Our results suggest that contraction of brain capillary pericytes in vitro by activation of P2Y-type purinergic receptors is caused by intracellular calcium release. This adds more mechanistic understanding of the role of pericytes in vessel constriction and points toward P2Y receptors as potential therapeutic targets.NEW & NOTEWORTHY The study concerns brain capillary pericytes, which have been suggested to play a role in the regulation of cerebral blood flow. We show that extracellular ATP causes contraction of primary brain pericytes by stimulation of purinergic receptors and subsequent release of intracellular Ca2+ concentration ([Ca2+]i). The contraction is mainly mediated through activation of P2Y-receptor subtypes, including P2Y1 and P2Y2. These findings add more mechanistic understanding of the role of pericytes in regulation of capillary blood flow. ATP was earlier suggested to be involved in capillary constriction in brain pathologies, and our study gives a detailed account of a part of this important mechanism.


Subject(s)
Adenosine Triphosphate/pharmacology , Brain/blood supply , Calcium Signaling/drug effects , Cell Shape/drug effects , Pericytes/drug effects , Purinergic P2Y Receptor Agonists/pharmacology , Receptors, Purinergic P2Y/drug effects , Animals , Capillaries/cytology , Cattle , Cells, Cultured , Inositol 1,4,5-Trisphosphate/metabolism , Pericytes/metabolism , Phenotype , Receptors, Purinergic P2Y/metabolism , Receptors, Purinergic P2Y1/drug effects , Receptors, Purinergic P2Y1/metabolism , Receptors, Purinergic P2Y2/drug effects , Receptors, Purinergic P2Y2/metabolism
3.
J Neurosci ; 40(49): 9364-9371, 2020 12 02.
Article in English | MEDLINE | ID: mdl-33122390

ABSTRACT

Mechanosensitivity is a well-known feature of astrocytes, however, its underlying mechanisms and functional significance remain unclear. There is evidence that astrocytes are acutely sensitive to decreases in cerebral perfusion pressure and may function as intracranial baroreceptors, tuned to monitor brain blood flow. This study investigated the mechanosensory signaling in brainstem astrocytes, as these cells reside alongside the cardiovascular control circuits and mediate increases in blood pressure and heart rate induced by falls in brain perfusion. It was found that mechanical stimulation-evoked Ca2+ responses in astrocytes of the rat brainstem were blocked by (1) antagonists of connexin channels, connexin 43 (Cx43) blocking peptide Gap26, or Cx43 gene knock-down; (2) antagonists of TRPV4 channels; (3) antagonist of P2Y1 receptors for ATP; and (4) inhibitors of phospholipase C or IP3 receptors. Proximity ligation assay demonstrated interaction between TRPV4 and Cx43 channels in astrocytes. Dye loading experiments showed that mechanical stimulation increased open probability of carboxyfluorescein-permeable membrane channels. These data suggest that mechanosensory Ca2+ responses in astrocytes are mediated by interaction between TRPV4 and Cx43 channels, leading to Cx43-mediated release of ATP which propagates/amplifies Ca2+ signals via P2Y1 receptors and Ca2+ recruitment from the intracellular stores. In astrocyte-specific Cx43 knock-out mice the magnitude of heart rate responses to acute increases in intracranial pressure was not affected by Cx43 deficiency. However, these animals displayed lower heart rates at different levels of cerebral perfusion, supporting the hypothesis of connexin hemichannel-mediated release of signaling molecules by astrocytes having an excitatory action on the CNS sympathetic control circuits.SIGNIFICANCE STATEMENT There is evidence suggesting that astrocytes may function as intracranial baroreceptors that play an important role in the control of systemic and cerebral circulation. To function as intracranial baroreceptors, astrocytes must possess a specialized membrane mechanism that makes them exquisitely sensitive to mechanical stimuli. This study shows that opening of connexin 43 (Cx43) hemichannels leading to the release of ATP is the key central event underlying mechanosensory Ca2+ responses in astrocytes. This astroglial mechanism plays an important role in the autonomic control of heart rate. These data add to the growing body of evidence suggesting that astrocytes function as versatile surveyors of the CNS metabolic milieu, tuned to detect conditions of potential metabolic threat, such as hypoxia, hypercapnia, and reduced perfusion.


Subject(s)
Astrocytes/physiology , Mechanotransduction, Cellular/physiology , Adenosine Triphosphate/metabolism , Animals , Blood Pressure/drug effects , Brain Stem/cytology , Brain Stem/drug effects , Brain Stem/physiology , Calcium Signaling/drug effects , Calcium Signaling/physiology , Cerebrovascular Circulation/physiology , Connexin 43/antagonists & inhibitors , Connexin 43/genetics , Female , Heart Rate/physiology , Male , Mechanotransduction, Cellular/drug effects , Mice , Mice, Knockout , Peptides/antagonists & inhibitors , Peptides/genetics , Physical Stimulation , Rats , Receptors, Purinergic P2Y1/drug effects , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/genetics
4.
Biomed Pharmacother ; 130: 110537, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32717630

ABSTRACT

Rhizoma Bletillae, the tubes of Bletilla striata, has been traditionally used in China as a hemostatic agent. In preliminary studies, the major active fraction responsible for its hemostatic effect have been confirmed to be Rhizoma Bletillae polysaccharide (RBp), but the hemostatic mechanism of action of RBp is still unknown.The main aim of this study was to clarify its mechanism of hemostatic effect. RBp was prepared by 80 % ethanol precipitation of the water extract of Rhizoma Bletillae followed by the Sevag method to remove proteins. The average molecular weight (Mw) of the crude RBp maintained at a range of 30.06-200 KDa. The hemostatic effects of RBp were evaluated by testing its effect on the platelet aggregation of rat platelet-rich plasma (PRP). PRP was dealt with different concentrations of RBp and platelet aggregation was measured by the turbidimetric method. The hemostatic mechanism of RBp was investigated by examining its effect on platelet shape, platelet secretion, and activation of related receptors (P2Y1, P2Y12 and TXA2) by electron microscopy and the turbidimetric method. RBp significantly enhanced the platelet aggregations at concentrations of 50-200 mg/L in a concentration-dependent manner. The inhibitory rate of platelet aggregation was significantly increased by apyrase and Ro31-8220 in a concentration-dependent manner, while RBp-induced platelet aggregation was completely inhibited by P2Y1, P2Y12 and the PKC receptor antagonists. However, the aggregation was not sensitive to TXA2. RBp, the active ingredients of Rhizoma Bletillae responsible for its hemostatic effect, could significantly accelerate the platelet aggregation and shape change. The hemostatic mechanism may involve activation of the P2Y1, P2Y12, and PKC receptors in the adenosine diphosphate (ADP) receptor signaling pathway.


Subject(s)
Hemostatics/pharmacology , Platelet-Rich Plasma/drug effects , Polysaccharides/pharmacology , Receptors, Purinergic P2/drug effects , Signal Transduction/drug effects , Animals , Dose-Response Relationship, Drug , In Vitro Techniques , Molecular Weight , Plant Extracts/pharmacology , Plant Tubers/chemistry , Platelet Aggregation/drug effects , Protein Kinase C/drug effects , Rats , Receptors, Purinergic P2Y1/drug effects , Receptors, Purinergic P2Y12/drug effects
5.
J Neurosci ; 40(15): 3052-3062, 2020 04 08.
Article in English | MEDLINE | ID: mdl-32132265

ABSTRACT

Maintenance of cardiorespiratory homeostasis depends on autonomic reflexes controlled by neuronal circuits of the brainstem. The neurophysiology and neuroanatomy of these reflex pathways are well understood, however, the mechanisms and functional significance of autonomic circuit modulation by glial cells remain largely unknown. In the experiments conducted in male laboratory rats we show that astrocytes of the nucleus of the solitary tract (NTS), the brain area that receives and integrates sensory information from the heart and blood vessels, respond to incoming afferent inputs with [Ca2+]i elevations. Astroglial [Ca2+]i responses are triggered by transmitters released by vagal afferents, glutamate acting at AMPA receptors and 5-HT acting at 5-HT2A receptors. In conscious freely behaving animals blockade of Ca2+-dependent vesicular release mechanisms in NTS astrocytes by virally driven expression of a dominant-negative SNARE protein (dnSNARE) increased baroreflex sensitivity by 70% (p < 0.001). This effect of compromised astroglial function was specific to the NTS as expression of dnSNARE in astrocytes of the ventrolateral brainstem had no effect. ATP is considered the principle gliotransmitter and is released by vesicular mechanisms blocked by dnSNARE expression. Consistent with this hypothesis, in anesthetized rats, pharmacological activation of P2Y1 purinoceptors in the NTS decreased baroreflex gain by 40% (p = 0.031), whereas blockade of P2Y1 receptors increased baroreflex gain by 57% (p = 0.018). These results suggest that glutamate and 5-HT, released by NTS afferent terminals, trigger Ca2+-dependent astroglial release of ATP to modulate baroreflex sensitivity via P2Y1 receptors. These data add to the growing body of evidence supporting an active role of astrocytes in brain information processing.SIGNIFICANCE STATEMENT Cardiorespiratory reflexes maintain autonomic balance and ensure cardiovascular health. Impaired baroreflex may contribute to the development of cardiovascular disease and serves as a robust predictor of cardiovascular and all-cause mortality. The data obtained in this study suggest that astrocytes are integral components of the brainstem mechanisms that process afferent information and modulate baroreflex sensitivity via the release of ATP. Any condition associated with higher levels of "ambient" ATP in the NTS would be expected to decrease baroreflex gain by the mechanism described here. As ATP is the primary signaling molecule of glial cells (astrocytes, microglia), responding to metabolic stress and inflammatory stimuli, our study suggests a plausible mechanism of how the central component of the baroreflex is affected in pathological conditions.


Subject(s)
Astrocytes/physiology , Baroreflex/physiology , Solitary Nucleus/physiology , Adenosine Triphosphate/physiology , Animals , Calcium Signaling/physiology , Male , Neurons, Afferent/metabolism , Neurotransmitter Agents/metabolism , Neurotransmitter Agents/physiology , Purinergic P2Y Receptor Agonists/pharmacology , Purinergic P2Y Receptor Antagonists/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, Serotonin, 5-HT2A/drug effects , Receptors, AMPA/drug effects , Receptors, Purinergic P2Y1/drug effects , SNARE Proteins/physiology , Serotonin/pharmacology , Vagus Nerve Stimulation
6.
Neuroscience ; 423: 98-108, 2019 12 15.
Article in English | MEDLINE | ID: mdl-31689490

ABSTRACT

Non-synaptic transmission is pervasive throughout the nervous system. It appears especially prevalent in peripheral ganglia, where non-synaptic interactions between neighboring cell bodies have been described in both physiological and pathological conditions, a phenomenon referred to as cross-depolarization (CD) and thought to play a role in sensory processing and chronic pain. CD has been proposed to be mediated by a chemical agent, but its identity has remained elusive. Here, we report that in the rat dorsal root ganglion (DRG), the P2Y1 purinergic receptor (P2RY1) plays an important role in regulating CD. The effect of P2RY1 is cell-type specific: pharmacological blockade of P2RY1 inhibited CD in A-type neurons while enhancing it in C-type neurons. In the nodose ganglion of the vagus, CD requires extracellular calcium in a large percentage of cells. In contrast, we show that in the DRG extracellular calcium appears to play no major role, pointing to a mechanistic difference between the two peripheral ganglia. Furthermore, we show that DRG glial cells also play a cell-type specific role in CD regulation. Fluorocitrate-induced glial inactivation had no effect on A-cells but enhanced CD in C-cells. These findings shed light on the mechanism of CD in the DRG and pave the way for further analysis of non-synaptic neuronal communication in sensory ganglia.


Subject(s)
Cell Communication/physiology , Ganglia, Spinal/physiology , Neurons/physiology , Receptors, Purinergic P2Y1/physiology , 2-Amino-5-phosphonovalerate/pharmacology , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Action Potentials/physiology , Adenosine Diphosphate/analogs & derivatives , Adenosine Diphosphate/pharmacology , Animals , Benzenesulfonates/pharmacology , Calcium/physiology , Citrates/pharmacology , Electric Stimulation , Male , Neural Inhibition/physiology , Neuroglia/drug effects , Neuroglia/physiology , Neurons/drug effects , Nodose Ganglion/physiology , Piperazines/pharmacology , Rats , Receptors, Purinergic P2Y1/drug effects
7.
Am J Physiol Endocrinol Metab ; 317(1): E25-E41, 2019 07 01.
Article in English | MEDLINE | ID: mdl-30912960

ABSTRACT

Fructose is widely used as a sweetener in processed food and is also associated with metabolic disorders, such as obesity. However, the underlying cellular mechanisms remain unclear, in particular, regarding the pancreatic ß-cell. Here, we investigated the effects of chronic exposure to fructose on the function of insulinoma cells and isolated mouse and human pancreatic islets. Although fructose per se did not acutely stimulate insulin exocytosis, our data show that chronic fructose rendered rodent and human ß-cells hyper-responsive to intermediate physiological glucose concentrations. Fructose exposure reduced intracellular ATP levels without affecting mitochondrial function, induced AMP-activated protein kinase activation, and favored ATP release from the ß-cells upon acute glucose stimulation. The resulting increase in extracellular ATP, mediated by pannexin1 (Panx1) channels, activated the calcium-mobilizer P2Y purinergic receptors. Immunodetection revealed the presence of both Panx1 channels and P2Y1 receptors in ß-cells. Addition of an ectonucleotidase inhibitor or P2Y1 agonists to naïve ß-cells potentiated insulin secretion stimulated by intermediate glucose, mimicking the fructose treatment. Conversely, the P2Y1 antagonist and Panx1 inhibitor reversed the effects of fructose, as confirmed using Panx1-null islets and by the clearance of extracellular ATP by apyrase. These results reveal an important function of ATP signaling in pancreatic ß-cells mediating fructose-induced hyper-responsiveness.


Subject(s)
Adenosine Triphosphate/physiology , Fructose/pharmacology , Glucose/pharmacology , Insulin Secretion/drug effects , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Signal Transduction/drug effects , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Adenosine Triphosphate/metabolism , Animals , Apyrase/metabolism , Connexins/genetics , Connexins/metabolism , Humans , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Purinergic P2Y Receptor Agonists/pharmacology , Purinergic P2Y Receptor Antagonists/pharmacology , Receptors, Purinergic P2Y/drug effects , Receptors, Purinergic P2Y/metabolism , Receptors, Purinergic P2Y1/drug effects , Receptors, Purinergic P2Y1/metabolism
8.
Cardiol J ; 26(6): 782-789, 2019.
Article in English | MEDLINE | ID: mdl-29671861

ABSTRACT

BACKGROUND: Activated platelets release platelet extracellular vesicles (PEVs). Adenosine diphosphate (ADP) receptors P2Y1 and P2Y12 both play a role in platelet activation, The present hypothesis herein is that the inhibition of these receptors may affect the release of PEVs. METHODS: Platelet-rich plasma from 10 healthy subjects was incubated with saline, P2Y1 antagonist MRS2179 (100 µM), P2Y12 antagonist ticagrelor (1 µM), and a combination of both antagonists. Platelets were activated by ADP (10 µM) under stirring conditions at 37°C. Platelet reactivity was assessed by impedance aggregometry. Concentrations of PEVs- (positive for CD61 but negative for P-selectin and phosphatidylserine) and PEVs+ (positive for all) were determined by a state-of-the-art flow cytometer. Procoagulant activity of PEVs was measured by a fibrin generation test. RESULTS: ADP-induced aggregation (57 ± 13 area under curve {AUC] units) was inhibited 73% by the P2Y1 antagonist, 86% by the P2Y12 antagonist, and 95% when combined (p < 0.001 for all). The release of PEVs- (2.9 E ± 0.8 × 108/mL) was inhibited 48% in the presence of both antagonists (p = 0.015), whereas antagonists alone were ineffective. The release of PEVs+ (2.4 ± 1.6 × 107/mL) was unaffected by the P2Y1 antagonist, but was 62% inhibited by the P2Y12 antagonist (p = 0.035), and 72% by both antagonists (p = 0.022). PEVs promoted coagulation in presence of tissue factor. CONCLUSIONS: Inhibition of P2Y1 and P2Y12 receptors reduces platelet aggregation and affects the release of distinct subpopulations of PEVs. Ticagrelor decreases the release of procoagulant PEVs from activated platelets, which may contribute to the observed clinical benefits in patients treated with ticagrelor.


Subject(s)
Blood Coagulation/drug effects , Blood Platelets/drug effects , Extracellular Vesicles/drug effects , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/drug effects , Purinergic P2Y Receptor Antagonists/pharmacology , Receptors, Purinergic P2Y12/drug effects , Ticagrelor/pharmacology , Adenosine Diphosphate/analogs & derivatives , Adenosine Diphosphate/pharmacology , Blood Platelets/metabolism , Extracellular Vesicles/metabolism , Healthy Volunteers , Humans , Receptors, Purinergic P2Y1/blood , Receptors, Purinergic P2Y1/drug effects , Receptors, Purinergic P2Y12/blood , Signal Transduction
9.
Sci Rep ; 8(1): 8084, 2018 05 24.
Article in English | MEDLINE | ID: mdl-29795391

ABSTRACT

Many hemorheologic Traditional Chinese Medicines (TCMs) that are widely-used clinically lack molecular mechanisms of action. We hypothesized that some of the active components of hemorheologic TCMs may function through targeting prothrombotic P2Y1 and/or P2Y12 receptors. The interactions between 253 antithrombotic compounds from TCM and these two G protein-coupled P2Y receptors were evaluated using virtual screening. Eleven highly ranked hits were further tested in radioligand binding and functional assays. Among these compounds, salvianolic acid A and C antagonized the activity of both P2Y1 and P2Y12 receptors in the low µM range, while salvianolic acid B antagonized the P2Y12 receptor. These three salvianolic acids are the major active components of the broadly-used hemorheologic TCM Danshen (Salvia militorrhiza), the antithrombotic molecular mechanisms of which were largely unknown. Thus, the combination of virtual screening and experimental validation identified potential mechanisms of action of multicomponent drugs that are already employed clinically.


Subject(s)
Alkenes/isolation & purification , Alkenes/pharmacology , Fibrinolytic Agents/isolation & purification , Fibrinolytic Agents/pharmacology , Polyphenols/isolation & purification , Polyphenols/pharmacology , Purinergic P2Y Receptor Antagonists , Salvia miltiorrhiza/chemistry , Alkenes/chemistry , Benzofurans/chemistry , Benzofurans/isolation & purification , Benzofurans/pharmacology , Caffeic Acids/chemistry , Caffeic Acids/isolation & purification , Caffeic Acids/pharmacology , Drugs, Chinese Herbal/chemistry , Fibrinolytic Agents/chemistry , Humans , Lactates/chemistry , Lactates/isolation & purification , Lactates/pharmacology , Medicine, Chinese Traditional , Models, Molecular , Molecular Docking Simulation , Molecular Structure , Polyphenols/chemistry , Purinergic P2Y Receptor Antagonists/chemistry , Purinergic P2Y Receptor Antagonists/isolation & purification , Purinergic P2Y Receptor Antagonists/pharmacology , Receptors, Purinergic P2Y1/chemistry , Receptors, Purinergic P2Y1/drug effects , Receptors, Purinergic P2Y1/metabolism , Receptors, Purinergic P2Y12/chemistry , Receptors, Purinergic P2Y12/drug effects , Receptors, Purinergic P2Y12/metabolism , Tumor Cells, Cultured
10.
J Cell Physiol ; 233(3): 2526-2536, 2018 Mar.
Article in English | MEDLINE | ID: mdl-28777435

ABSTRACT

Extracellular nucleotides can regulate cell proliferation in both normal and tumorigenic tissues. Here, we studied how extracellular nucleotides regulate the proliferation of ZL55 cells, a mesothelioma-derived cell line obtained from bioptic samples of asbestos-exposed patients. ADP and 2-MeS-ADP inhibited ZL55 cell proliferation, whereas ATP, UTP, and UDP were inactive. The nucleotide potency profile and the blockade of the ADP-mediated inhibitory effect by the phospholipase C inhibitor U-73122 suggest that P2Y1 receptor controls ZL55 cell proliferation. The activation of P2Y1 receptor by ADP leads to activation of intracellular transduction pathways involving [Ca2+ ]i , PKC-δ/PKC-α, and MAPKs, ERK1/2 and JNK1/2. Cell treatment with ADP or 2-MeS-ADP also provokes the activation of p53, causing an accumulation of the G1 cyclin-dependent kinase inhibitors p21WAF1 and p27Kip . Inhibition of ZL55 cell proliferation by ADP was completely reversed by inhibiting MEK1/2, or JNK1/2, or PKC-δ, and PKC-α. Through the inhibition of ADP-activated transductional kinases it was found that PKC-δ was responsible for JNK1/2 activation. JNK1/2 has a role in transcriptional up-regulation of p53, p21WAF1/CIP1 , and p27kip1 . Conversely, the ADP-activated PKC-α provoked ERK1/2 phosphorylation. ERK1/2 increased p53 stabilization, required to G1 arrest of ZL55 cells. Concluding, the importance of the study is twofold: first, results shed light on the mechanism of cell cycle inhibition by ADP; second, results suggest that extracellular ADP may inhibit mesothelioma progression.


Subject(s)
Adenosine Diphosphate/pharmacology , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Mesothelioma/drug therapy , Protein Kinase C-alpha/metabolism , Protein Kinase C-delta/metabolism , Purinergic P2Y Receptor Agonists/pharmacology , Receptors, Purinergic P2Y1/drug effects , Signal Transduction/drug effects , Adenosine Diphosphate/analogs & derivatives , Asbestos/adverse effects , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Dose-Response Relationship, Drug , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Mesothelioma/enzymology , Mesothelioma/genetics , Mesothelioma/pathology , Phosphorylation , Protein Kinase C-alpha/genetics , Protein Kinase C-delta/genetics , Protein Stability , RNA Interference , Receptors, Purinergic P2Y1/metabolism , Thionucleotides/pharmacology , Time Factors , Transfection , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
11.
Arterioscler Thromb Vasc Biol ; 37(9): 1646-1656, 2017 09.
Article in English | MEDLINE | ID: mdl-28684613

ABSTRACT

OBJECTIVE: Impaired endothelial cell (EC) autophagy compromises shear stress-induced nitric oxide (NO) generation. We determined the responsible mechanism. APPROACH AND RESULTS: On autophagy compromise in bovine aortic ECs exposed to shear stress, a decrease in glucose uptake and EC glycolysis attenuated ATP production. We hypothesized that decreased glycolysis-dependent purinergic signaling via P2Y1 (P2Y purinoceptor 1) receptors, secondary to impaired autophagy in ECs, prevents shear-induced phosphorylation of eNOS (endothelial nitric oxide synthase) at its positive regulatory site S1117 (p-eNOSS1177) and NO generation. Maneuvers that restore glucose transport and glycolysis (eg, overexpression of GLUT1 [glucose transporter 1]) or purinergic signaling (eg, addition of exogenous ADP) rescue shear-induced p-eNOSS1177 and NO production in ECs with impaired autophagy. Conversely, inhibiting glucose transport via GLUT1 small interfering RNA, blocking purinergic signaling via ectonucleotidase-mediated ATP/ADP degradation (eg, apyrase), or inhibiting P2Y1 receptors using pharmacological (eg, MRS2179 [2'-deoxy-N6-methyladenosine 3',5'-bisphosphate tetrasodium salt]) or genetic (eg, P2Y1-receptor small interfering RNA) procedures inhibit shear-induced p-eNOSS1177 and NO generation in ECs with intact autophagy. Supporting a central role for PKCδT505 (protein kinase C delta T505) in relaying the autophagy-dependent purinergic-mediated signal to eNOS, we find that (1) shear stress-induced activating phosphorylation of PKCδT505 is negated by inhibiting autophagy, (2) shear-induced p-eNOSS1177 and NO generation are restored in autophagy-impaired ECs via pharmacological (eg, bryostatin) or genetic (eg, constitutively active PKCδ) activation of PKCδT505, and (3) pharmacological (eg, rottlerin) and genetic (eg, PKCδ small interfering RNA) PKCδ inhibition prevents shear-induced p-eNOSS1177 and NO generation in ECs with intact autophagy. Key nodes of dysregulation in this pathway on autophagy compromise were revealed in human arterial ECs. CONCLUSIONS: Targeted reactivation of purinergic signaling and PKCδ has strategic potential to restore compromised NO generation in pathologies associated with suppressed EC autophagy.


Subject(s)
Adenosine Triphosphate/metabolism , Autophagy , Endothelial Cells/enzymology , Glycolysis , Mechanotransduction, Cellular , Nitric Oxide Synthase Type III/metabolism , Nitric Oxide/metabolism , Receptors, Purinergic P2Y1/metabolism , Animals , Autophagy/drug effects , Autophagy-Related Proteins/deficiency , Autophagy-Related Proteins/genetics , Cattle , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/pathology , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Glycolysis/drug effects , Humans , Mechanotransduction, Cellular/drug effects , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Protein Kinase C-delta/antagonists & inhibitors , Protein Kinase C-delta/genetics , Protein Kinase C-delta/metabolism , Protein Kinase Inhibitors/pharmacology , Purinergic P2Y Receptor Antagonists/pharmacology , RNA Interference , Reactive Oxygen Species/metabolism , Receptors, Purinergic P2Y1/drug effects , Receptors, Purinergic P2Y1/genetics , Serine , Stress, Mechanical , Transfection , Ubiquitin-Conjugating Enzymes/deficiency , Ubiquitin-Conjugating Enzymes/genetics
12.
Purinergic Signal ; 13(3): 331-338, 2017 09.
Article in English | MEDLINE | ID: mdl-28555330

ABSTRACT

Extracellular ATP (released by endothelial and immune cells) and its metabolite ADP are important pro-inflammatory mediators via the activation of purinergic P2 receptors (P2Y and P2X), which represent potential new targets for anti-inflammatory therapy. Endothelial P2Y1 receptor (P2Y1R) induces endothelial cell activation triggering leukocyte adhesion. A number of data have implicated melatonin as a modulator of immunity, inflammation, and endothelial cell function, but to date no studies have investigated whether melatonin modulates endothelial P2YR signaling. Here, we evaluated the putative effect of melatonin on P2Y1R-mediated leukocyte adhesion to endothelial cells and TNF-α production, using mesenteric endothelial cells and fresh peripheral blood mononuclear cells isolated from rats. Endothelial cells were treated with the P2Y1R agonist 2MeSATP, alone or in combination with melatonin, and then exposed to mononuclear cells. 2MeSATP increased leukocyte adhesion to endothelial cells and TNF-α production in vitro, and melatonin inhibited both effects without altering P2Y1R protein expression. In addition, assays with the Ca2+ chelator BAPTA-AM indicate that the effect of melatonin on 2MeSATP-stimulated leukocyte adhesion depends on intracellular Ca2+ modulation. P2Y1R is considered a potential target to control chronic inflammation. Therefore, our data unveiled a new endothelial cell modulator of purinergic P2Y1 receptor signaling.


Subject(s)
Cell Adhesion/drug effects , Endothelial Cells/drug effects , Leukocytes, Mononuclear/metabolism , Melatonin/pharmacology , Receptors, Purinergic P2Y1/drug effects , Adenosine Diphosphate/pharmacology , Animals , Calcium/metabolism , Endothelial Cells/metabolism , Male , Rats, Wistar , Receptors, Leukocyte-Adhesion/drug effects , Receptors, Leukocyte-Adhesion/metabolism , Receptors, Purinergic P2Y1/metabolism , Signal Transduction/physiology
13.
Oncotarget ; 7(51): 85381-85392, 2016 Dec 20.
Article in English | MEDLINE | ID: mdl-27863418

ABSTRACT

PIN1, which belongs to a family of prolyl isomerases, specifically binds to phosphorylated Ser/Thr-pro motifs to catalytically regulate the post-phosphorylation conformation of its substrates. This study aimed to investigate the importance of Pin1 expression in human dental pulp cells (hDPCs) to understand the involvement of Pin1 in the regulation of P2Y1 and the activation of ADP-mediated P2Y1 signaling. This study found that the protein levels of P2Y1 gradually decreased after the onset of cell recovery following heat stress. Interestedly, hDPC migration significantly decreased during the recovery period. An in vitro study revealed that the silencing of PIN1 by siRNA or the pharmacologic inhibition of its activity decreased the migration of P2Y1 and P2Y1 expression in these cells. In addition, we found that Pin1 directly interacts with S252 of P2Y1 and that its binding stabilizes the P2Y1 protein to increase migration activity. These results strongly suggest that Pin1 mediates cell migration by stabilizing P2Y1 and that the Pin1/P2Y1 signaling pathways might serve as a novel mechanism of cell migration progression in hDPCs.


Subject(s)
Adenosine Diphosphate/pharmacology , Cell Movement/drug effects , Dental Pulp/drug effects , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , Purinergic P2Y Receptor Agonists/pharmacology , Receptors, Purinergic P2Y1/drug effects , Animals , Cell Proliferation/drug effects , Cells, Cultured , Dental Pulp/cytology , Dental Pulp/enzymology , Dose-Response Relationship, Drug , Fibroblasts/drug effects , Fibroblasts/enzymology , Heat-Shock Response , Humans , Mice , Mitogen-Activated Protein Kinases/metabolism , NIMA-Interacting Peptidylprolyl Isomerase/genetics , Protein Stability , RNA Interference , Receptors, Purinergic P2Y1/genetics , Receptors, Purinergic P2Y1/metabolism , Signal Transduction/drug effects , Time Factors , Transfection
14.
Vascul Pharmacol ; 82: 66-72, 2016 07.
Article in English | MEDLINE | ID: mdl-26924460

ABSTRACT

Schistosomiasis is caused by an intravascular parasite and linked to phenotypic changes in endothelial cells that favor inflammation. Endothelial cells express P2Y1 receptors (P2Y1R), and their activation by ADP favors leukocyte adhesion to the endothelial monolayer. We aimed to evaluate the influence of schistosomiasis upon endothelial purinergic signaling-mediated leukocyte adhesion. Mesenteric endothelial cells and mononuclear cells from control and Schistosoma mansoni-infected mice were used in co-culture. P2Y1R levels were similar in both groups. Basal leukocyte adhesion was higher in the infected than in the control group; leukocyte adhesion increased after treatment with the P2Y1R agonist 2-MeSATP in both groups, though it only marginally increased in the infected group. Pre-incubation with the selective P2Y1R antagonist MRS2179 (0.3µM) prevented the agonist effect. However, in the infected group it also reduced the basal leukocyte adhesion, suggesting endothelial cell pre-activation. The endothelial expressions of NTPDases 2 and 3 were significantly increased in the infected group, increasing extracellular ATP hydrolysis and ADP formation by endothelial cells. Therefore, mesenteric endothelial cells are primed by schistosomiasis to a pro-inflammatory phenotype characterized by an increased expression of NTPDases 2 and 3, favoring ADP accumulation and mononuclear cell adhesion, possibly contributing to mesenteric inflammation and schistosomiasis morbidity.


Subject(s)
Adenosine Triphosphatases/metabolism , Cell Adhesion , Endothelial Cells/enzymology , Leukocytes/metabolism , Mesentery/blood supply , Receptors, Purinergic P2Y1/metabolism , Schistosoma mansoni/pathogenicity , Schistosomiasis/enzymology , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Animals , Cells, Cultured , Coculture Techniques , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/parasitology , Host-Pathogen Interactions , Humans , Hydrolysis , Leukocytes/drug effects , Leukocytes/parasitology , Male , Mice , Phenotype , Purinergic P2Y Receptor Agonists/pharmacology , Receptors, Purinergic P2Y1/drug effects , Schistosomiasis/parasitology , Signal Transduction , Up-Regulation
16.
Arterioscler Thromb Vasc Biol ; 36(3): 501-9, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26743169

ABSTRACT

OBJECTIVE: Unlike currently approved adenosine diphosphate receptor antagonists, the new diadenosine tetraphosphate derivative GLS-409 targets not only P2Y12 but also the second human platelet adenosine diphosphate receptor P2Y1 and may, therefore, be a promising antiplatelet drug candidate. The current study is the first to investigate the in vivo antithrombotic effects of GLS-409. APPROACH AND RESULTS: We studied (1) the in vivo effects of GLS-409 on agonist-stimulated platelet aggregation in anesthetized rats, (2) the antithrombotic activity of GLS-409 and the associated effect on the bleeding time in a canine model of platelet-mediated coronary artery thrombosis, and (3) the inhibition of agonist-stimulated platelet aggregation by GLS-409 versus selective P2Y1 and P2Y12 inhibition in vitro in samples from healthy human subjects before and 2 hours after aspirin intake. In vivo treatment with GLS-409 significantly inhibited adenosine diphosphate- and collagen-stimulated platelet aggregation in rats. Further, GLS-409 attenuated cyclic flow variation, that is, platelet-mediated thrombosis, in vivo in our canine model of unstable angina. The improvement in coronary patency was accompanied by a nonsignificant 30% increase in bleeding time. Of note, GLS-409 exerted its effects without affecting rat and canine hemodynamics. Finally, in vitro treatment with GLS-409 showed effects similar to that of cangrelor and the combination of cangrelor with the selective P2Y1 inhibitor MRS 2179 on agonist-stimulated platelet aggregation in human platelet-rich plasma and whole blood before and 2 hours after aspirin intake. CONCLUSIONS: Synergistic inhibition of both P2Y1 and P2Y12 adenosine diphosphate receptors by GLS-409 immediately attenuates platelet-mediated thrombosis and effectively blocks agonist-stimulated platelet aggregation irrespective of concomitant aspirin therapy.


Subject(s)
Blood Platelets/drug effects , Coronary Thrombosis/drug therapy , Dinucleoside Phosphates/pharmacology , Fibrinolytic Agents/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/drug effects , Purinergic P2Y Receptor Antagonists/pharmacology , Receptors, Purinergic P2Y12/drug effects , Receptors, Purinergic P2Y1/drug effects , Adenosine Diphosphate/analogs & derivatives , Adenosine Diphosphate/pharmacology , Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/pharmacology , Adult , Animals , Aspirin/pharmacology , Blood Coagulation/drug effects , Blood Platelets/metabolism , Coronary Thrombosis/blood , Disease Models, Animal , Dogs , Dose-Response Relationship, Drug , Female , Humans , Male , Platelet Function Tests , Rats, Sprague-Dawley , Receptors, Purinergic P2Y1/blood , Receptors, Purinergic P2Y12/blood , Time Factors , Young Adult
17.
Purinergic Signal ; 12(1): 69-78, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26538146

ABSTRACT

Peripheral purinergic signaling plays an important role in nociception. Increasing evidence suggests that metabotropic P2Y receptors are also involved, but little is known about the underlying mechanism. Herein, we report that selective P2Y receptor agonist uridine 5'-triphosphate (UTP) can exert an enhancing effect on the functional activity of acid-sensing ion channels (ASICs), key sensors for extracellular protons, in rat dorsal root ganglia (DRG) neurons. First, UTP dose-dependently increased the amplitude of ASIC currents. UTP also shifted the concentration-response curve for proton upwards, with a 56.6 ± 6.4% increase of the maximal current response to proton. Second, UTP potentiation of proton-gated currents can be mimicked by adenosine 5'-triphosphate (ATP), but not by P2Y1 receptor agonist ADP. Potentiation of UTP was blocked by P2Y receptor antagonist suramin and by inhibition of intracellular G protein, phospholipase C (PLC), protein kinase C (PKC), or protein interacting with C-kinase 1 (PICK1) signaling. Third, UTP altered acidosis-evoked membrane excitability of DRG neurons and caused a significant increase in the amplitude of the depolarization and the number of spikes induced by acid stimuli. Finally, UTP dose-dependently exacerbated nociceptive responses to injection of acetic acid in rats. These results suggest that UTP enhanced ASIC-mediated currents and nociceptive responses, which reveal a novel peripheral mechanism underlying UTP-sensitive P2Y2 receptor involvement in hyperalgesia by sensitizing ASICs in primary sensory neurons.


Subject(s)
Acid Sensing Ion Channels/drug effects , Purinergic P2Y Receptor Agonists/pharmacology , Sensory Receptor Cells/drug effects , Uridine Triphosphate/pharmacology , Acid Sensing Ion Channels/metabolism , Acidosis/physiopathology , Adenosine Diphosphate/pharmacology , Adenosine Triphosphate/pharmacology , Animals , Dose-Response Relationship, Drug , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Male , Membrane Potentials/drug effects , Pain/psychology , Pain Measurement/drug effects , Protons , Purinergic P2Y Receptor Antagonists/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Purinergic P2Y1/drug effects , Signal Transduction/drug effects , Suramin/pharmacology , Uridine Triphosphate/antagonists & inhibitors
18.
Am J Physiol Heart Circ Physiol ; 309(11): H1837-45, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26453330

ABSTRACT

Impairment of moment-to-moment adjustment of cerebral blood flow (CBF) via neurovascular coupling is thought to play a critical role in the genesis of cognitive impairment associated with aging and pathological conditions associated with accelerated cerebromicrovascular aging (e.g., hypertension, obesity). Although previous studies demonstrate that endothelial dysfunction plays a critical role in neurovascular uncoupling in these conditions, the role of endothelial NO mediation in neurovascular coupling responses is not well understood. To establish the link between endothelial function and functional hyperemia, neurovascular coupling responses were studied in mutant mice overexpressing or deficient in endothelial NO synthase (eNOS), and the role of P2Y1 receptors in purinergic glioendothelial coupling was assessed. We found that genetic depletion of eNOS (eNOS(-/-)) and pharmacological inhibition of NO synthesis significantly decreased the CBF responses in the somatosensory cortex evoked by whisker stimulation and by administration of ATP. Overexpression of eNOS enhanced NO mediation of functional hyperemia. In control mice, the selective and potent P2Y1 receptor antagonist MRS2179 attenuated both whisker stimulation-induced and ATP-mediated CBF responses, whereas, in eNOS(-/-) mice, the inhibitory effects of MRS2179 were blunted. Collectively, our findings provide additional evidence for purinergic glio-endothelial coupling during neuronal activity, highlighting the role of ATP-mediated activation of eNOS via P2Y1 receptors in functional hyperemia.


Subject(s)
Astrocytes/enzymology , Cell Communication , Endothelial Cells/enzymology , Hyperemia/enzymology , Microcirculation , Neurovascular Coupling , Nitric Oxide Synthase Type III/metabolism , Receptors, Purinergic P2Y1/metabolism , Somatosensory Cortex/enzymology , Animals , Cell Communication/drug effects , Endothelial Cells/drug effects , Enzyme Inhibitors/pharmacology , Hemodynamics , Homeostasis , Hyperemia/genetics , Hyperemia/physiopathology , Mechanotransduction, Cellular , Mice, Inbred C57BL , Mice, Knockout , Microcirculation/drug effects , Neurovascular Coupling/drug effects , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/antagonists & inhibitors , Nitric Oxide Synthase Type III/deficiency , Nitric Oxide Synthase Type III/genetics , Purinergic P2Y Receptor Agonists/pharmacology , Purinergic P2Y Receptor Antagonists/pharmacology , Receptors, Purinergic P2Y1/drug effects , Somatosensory Cortex/blood supply , Somatosensory Cortex/drug effects , Somatosensory Cortex/physiopathology , Vibrissae/innervation
19.
Purinergic Signal ; 11(4): 491-506, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26395809

ABSTRACT

Purinergic signaling may be involved in embryonic development of the heart. In the present study, the effects of purinergic receptor stimulation on cardiomyogenesis of mouse embryonic stem (ES) cells were investigated. ADP or ATP increased the number of cardiac clusters and cardiac cells, as well as beating frequency. Cardiac-specific genes showed enhanced expression of α-MHC, MLC2v, α-actinin, connexin 45 (Cx45), and HCN4, on both gene and protein levels upon ADP/ATP treatment, indicating increased cardiomyogenesis and pacemaker cell differentiation. Real-time RT-PCR analysis of purinergic receptor expression demonstrated presence of P2X1, P2X4, P2X6, P2X7, P2Y1, P2Y2, P2Y4, and P2Y6 on differentiating ES cells. ATP and ADP as well as the P2X agonists ß,γ-methylenadenosine 5'-triphosphate (ß,γ-MetATP) and 8-bromoadenosine 5'-triphosphate (8-Br-ATP) but not UTP or UDP transiently increased the intracellular calcium concentration ([Ca(2+)](i)) as evaluated by the calcium indicator Fluo-4, whereas no changes in membrane potential were observed. [Ca(2+)](i) transients induced by ADP/ATP were abolished by the phospholipase C-ß (PLC-ß) inhibitor U-73122, suggesting involvement of metabotropic P2Y receptors. Furthermore, partial inhibition of [Ca(2+)](i) transients was achieved in presence of MRS2179, a selective P2Y1 receptor antagonist, whereas PPADS, a non-selective P2 receptor inhibitor, completely abolished the [Ca(2+)](i) response. Consequently, cardiomyocyte differentiation was decreased upon long term co-incubation of cells with ADP and P2 receptor antagonists. In summary, activation of purinoceptors and the subsequent [Ca(2+)](i) transients enhance the differentiation of ES cells toward cardiomyocytes. Purinergic receptor stimulation may be a promising strategy to drive the fate of pluripotent ES cells into a particular population of cardiomyocytes.


Subject(s)
Adenosine Diphosphate/pharmacology , Adenosine Triphosphate/pharmacology , Embryonic Stem Cells/drug effects , Muscle Development/drug effects , Myocytes, Cardiac/drug effects , Adenosine Triphosphate/antagonists & inhibitors , Animals , Calcium/metabolism , Calcium Signaling/drug effects , Cell Differentiation/drug effects , Female , Gene Expression/drug effects , Membrane Potentials/drug effects , Mice , Myocardial Contraction/drug effects , Pregnancy , Purinergic P2X Receptor Agonists/pharmacology , Purinergic P2Y Receptor Antagonists/pharmacology , Receptors, Purinergic P2X/biosynthesis , Receptors, Purinergic P2X/drug effects , Receptors, Purinergic P2X/genetics , Receptors, Purinergic P2Y1/drug effects , Uridine Diphosphate/pharmacology , Uridine Triphosphate/pharmacology
20.
Br J Pharmacol ; 172(21): 5199-210, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26282717

ABSTRACT

BACKGROUND AND PURPOSE: Stimulation of soluble guanylyl cyclase (sGC) is a valuable therapeutic strategy for the treatment of several cardiovascular diseases. The sGC stimulator riociguat has been approved for the treatment of two forms of pulmonary hypertension. Platelets contain large amounts of sGC and play a key role in the regulation of haemostasis. Therefore, we investigated the effects of riociguat on platelet function. EXPERIMENTAL APPROACH: The effect of riociguat treatment on human platelet activation and aggregation was investigated. The sGC-specific effects of riociguat were determined by comparing wild-type and platelet-specific sGC-knockout mice. KEY RESULTS: Riociguat induced cGMP synthesis and subsequent PKG activation in human platelets, suggesting that the inhibitory effects are mediated by cGMP signalling. This finding was confirmed when sGC-knockout platelets were not inhibited by riociguat. In washed human platelets, 100 nM riociguat reduced ADP-induced GPIIb/IIIa activation, while a 10-fold higher concentration was required to reduce convulxin-stimulated GPIIb/IIIa activation. Riociguat inhibited ADP-induced platelet shape change and aggregation, while ATP-induced shape change remained unaffected. However, in PRP and whole blood, 50-100 µM riociguat was required to inhibit platelet activation and aggregation. Riociguat in combination with iloprost significantly inhibited platelet aggregation, even in whole blood. CONCLUSIONS AND IMPLICATIONS: Riociguat inhibits platelet activation in whole blood only at concentrations above 50 µM, while the plasma concentrations in riociguat-treated patients are 150 to 500 nM. This finding indicates that riociguat treatment does not affect platelet function in patients. Nevertheless, the possibility that riociguat acts synergistically with iloprost to inhibit platelet activation should be considered.


Subject(s)
Blood , Guanylate Cyclase/metabolism , Platelet Activation/drug effects , Platelet Aggregation/drug effects , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Enzyme Activation , Humans , Iloprost/pharmacology , Mice , Mice, Knockout , Platelet Aggregation/physiology , Receptors, Purinergic P2Y1/drug effects , Receptors, Purinergic P2Y1/physiology , Receptors, Purinergic P2Y12/drug effects , Receptors, Purinergic P2Y12/physiology , Soluble Guanylyl Cyclase
SELECTION OF CITATIONS
SEARCH DETAIL
...