Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
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
2.
Cell Death Dis ; 11(5): 394, 2020 05 26.
Article in English | MEDLINE | ID: mdl-32457291

ABSTRACT

Nod-like receptor protein 3 (NLRP3)-mediated pyroptosis has a causal role in the pathogenesis of gout. P2Y14 receptor (P2Y14R) distributed in immune cells including macrophages is a Gi-coupled receptor that inhibits the synthesis of cAMP, which has been regarded as a potential regulator of inflammatory response. Nevertheless, the role of P2Y14R in MSU-induced pyroptosis of macrophages involved in acute gouty arthritis is still unclear. In our present study, P2Y14R knockout (P2Y14R-KO) disrupted MSU-induced histopathologic changes in rat synoviums, accompanied with a significant inhibition of pyroptotic cell death characterized by Caspase-1/PI double-positive and blockade of NLRP3 inflammasome activation in synovial tissues, which was consistent with that observed in in vitro studies. Owing to the interaction of NLRP3 inflammasome and cAMP, we then investigated the effect of adenylate cyclase activator (Forskolin) on macrophage pyroptosis and gout flare caused by MSU stimulation. The reversal effect of Forskolin verified the negative regulatory role of cAMP in MSU-induced pyroptosis. More importantly, adenylate cyclase inhibitor (SQ22536) intervention led to a reversal of protection attributed to P2Y14R deficiency. Findings in air pouch animal models also verified aforementioned experimental results. Our study first identified the role of P2Y14R/cAMP/NLRP3 signaling pathway in acute gouty arthritis, which provides a novel insight into the pathological mechanisms of pyroptosis-related diseases.


Subject(s)
Arthritis, Gouty/metabolism , Inflammasomes/metabolism , Macrophages/metabolism , Receptors, Purinergic P2Y/metabolism , Animals , Disease Models, Animal , Gout/metabolism , Gout/pathology , Inflammasomes/drug effects , Male , Pyroptosis/drug effects , Pyroptosis/physiology , Rats, Sprague-Dawley , Receptors, Purinergic P2Y/drug effects , Signal Transduction/drug effects , Signal Transduction/physiology , Uric Acid/pharmacology
3.
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
4.
Brain Res Bull ; 151: 125-131, 2019 09.
Article in English | MEDLINE | ID: mdl-30599217

ABSTRACT

Neuropathic pain is generally resistant to currently available treatments, and it is often a consequence of nerve injury due to surgery, diabetes or infection. Myocardial ischemic nociceptive signaling increases the sympathoexcitatory reflex to aggravate myocardial injury. Elucidation of the pathogenetic factors might provide a target for optimal treatment. Abundant evidence in the literature suggests that P2X and P2Y receptors play important roles in signal transmission. Traditional Chinese medicines, such as emodin, puerarin and resveratrol, antagonize nociceptive transmission mediated by purinergic 2 (P2) receptors in primary afferent neurons. This review summarizes recently published data on P2 receptor-mediated neuropathic pain and myocardial ischemia in dorsal root ganglia (DRG), superior cervical ganglia (SCG) and stellate ganglia (SG), with a special focus on the beneficial role of natural compounds.


Subject(s)
Neuralgia/therapy , Receptors, Purinergic P2/metabolism , Animals , Disease Models, Animal , Ganglia, Spinal/pathology , Humans , Medicine, Chinese Traditional/methods , Myocardial Ischemia/drug therapy , Neuralgia/metabolism , Neurons/physiology , Receptors, Purinergic P2/drug effects , Receptors, Purinergic P2X/drug effects , Receptors, Purinergic P2Y/drug effects , Reflex/physiology , Signal Transduction/physiology , Superior Cervical Ganglion/pathology
5.
Pharmacol Ther ; 190: 24-80, 2018 10.
Article in English | MEDLINE | ID: mdl-29660366

ABSTRACT

P2Y receptors (P2YRs) are a family of G protein-coupled receptors activated by extracellular nucleotides. Physiological P2YR agonists include purine and pyrimidine nucleoside di- and triphosphates, such as ATP, ADP, UTP, UDP, nucleotide sugars, and dinucleotides. Eight subtypes exist, P2Y1, P2Y2, P2Y4, P2Y6, P2Y11, P2Y12, P2Y13, and P2Y14, which represent current or potential future drug targets. Here we provide a comprehensive overview of ligands for the subgroup of the P2YR family that is activated by uracil nucleotides: P2Y2 (UTP, also ATP and dinucleotides), P2Y4 (UTP), P2Y6 (UDP), and P2Y14 (UDP, UDP-glucose, UDP-galactose). The physiological agonists are metabolically unstable due to their fast hydrolysis by ectonucleotidases. A number of agonists with increased potency, subtype-selectivity and/or enzymatic stability have been developed in recent years. Useful P2Y2R agonists include MRS2698 (6-01, highly selective) and PSB-1114 (6-05, increased metabolic stability). A potent and selective P2Y2R antagonist is AR-C118925 (10-01). For studies of the P2Y4R, MRS4062 (3-15) may be used as a selective agonist, while PSB-16133 (10-06) is a selective antagonist. Several potent P2Y6R agonists have been developed including 5-methoxyuridine 5'-O-((Rp)α-boranodiphosphate) (6-12), PSB-0474 (3-11), and MRS2693 (3-26). The isocyanate MRS2578 (10-08) is used as a selective P2Y6R antagonist, although its reactivity and low water-solubility are limiting. With MRS2905 (6-08), a potent and metabolically stable P2Y14R agonist is available, while PPTN (10-14) represents a potent and selective P2Y14R antagonist. The radioligand [3H]UDP can be used to label P2Y14Rs. In addition, several fluorescent probes have been developed. Uracil nucleotide-activated P2YRs show great potential as drug targets, especially in inflammation, cancer, cardiovascular and neurodegenerative diseases.


Subject(s)
Purinergic P2Y Receptor Agonists/pharmacology , Purinergic P2Y Receptor Antagonists/pharmacology , Receptors, Purinergic P2Y/drug effects , Animals , Drug Development/methods , Humans , Ligands , Purinergic P2Y Receptor Antagonists/chemistry , Radioligand Assay , Receptors, Purinergic P2Y/metabolism , Solubility , Uracil Nucleotides/metabolism
6.
PLoS One ; 13(2): e0190893, 2018.
Article in English | MEDLINE | ID: mdl-29466379

ABSTRACT

Small alterations in extracellular acidity are potentially important modulators of neuronal signaling within the vertebrate retina. Here we report a novel extracellular acidification mechanism mediated by glial cells in the retina. Using self-referencing H+-selective microelectrodes to measure extracellular H+ fluxes, we show that activation of retinal Müller (glial) cells of the tiger salamander by micromolar concentrations of extracellular ATP induces a pronounced extracellular H+ flux independent of bicarbonate transport. ADP, UTP and the non-hydrolyzable analog ATPγs at micromolar concentrations were also potent stimulators of extracellular H+ fluxes, but adenosine was not. The extracellular H+ fluxes induced by ATP were mimicked by the P2Y1 agonist MRS 2365 and were significantly reduced by the P2 receptor blockers suramin and PPADS, suggesting activation of P2Y receptors. Bath-applied ATP induced an intracellular rise in calcium in Müller cells; both the calcium rise and the extracellular H+ fluxes were significantly attenuated when calcium re-loading into the endoplasmic reticulum was inhibited by thapsigargin and when the PLC-IP3 signaling pathway was disrupted with 2-APB and U73122. The anion transport inhibitor DIDS also markedly reduced the ATP-induced increase in H+ flux while SITS had no effect. ATP-induced H+ fluxes were also observed from Müller cells isolated from human, rat, monkey, skate and lamprey retinae, suggesting a highly evolutionarily conserved mechanism of potential general importance. Extracellular ATP also induced significant increases in extracellular H+ flux at the level of both the outer and inner plexiform layers in retinal slices of tiger salamander which was significantly reduced by suramin and PPADS. We suggest that the novel H+ flux mediated by ATP-activation of Müller cells and of other glia as well may be a key mechanism modulating neuronal signaling in the vertebrate retina and throughout the brain.


Subject(s)
Adenosine Triphosphate/metabolism , Ependymoglial Cells/metabolism , Retina/cytology , Retina/metabolism , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , Adenosine Diphosphate/analogs & derivatives , Adenosine Diphosphate/pharmacology , Adenosine Triphosphate/pharmacology , Ambystoma , Animals , Ependymoglial Cells/drug effects , Extracellular Fluid/drug effects , Extracellular Fluid/metabolism , Humans , Hydrogen-Ion Concentration , Ictaluridae , In Vitro Techniques , Ion Transport/drug effects , Lampreys , Macaca fascicularis , Macaca mulatta , Pyridoxal Phosphate/analogs & derivatives , Pyridoxal Phosphate/pharmacology , Rats , Receptors, Purinergic P2Y/drug effects , Signal Transduction , Skates, Fish , Suramin/pharmacology
7.
Am J Physiol Cell Physiol ; 314(5): C627-C639, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29365273

ABSTRACT

The objective of this study was to determine the molecular identity of ion channels involved in K+ secretion by the mammary epithelium and to examine their regulation by purinoceptor agonists. Apical membrane voltage-clamp experiments were performed on human mammary epithelial cells where the basolateral membrane was exposed to the pore-forming antibiotic amphotericin B dissolved in a solution with intracellular-like ionic composition. Addition of the Na+ channel inhibitor benzamil reduced the basal current, consistent with inhibition of Na+ uptake across the apical membrane, whereas the KCa3.1 channel blocker TRAM-34 produced an increase in current resulting from inhibition of basal K+ efflux. Treatment with two-pore potassium (K2P) channel blockers quinidine, bupivacaine and a selective TASK1/TASK3 inhibitor (PK-THPP) all produced concentration-dependent inhibition of apical K+ efflux. qRT-PCR experiments detected mRNA expression for nine K2P channel subtypes. Western blot analysis of biotinylated apical membranes and confocal immunocytochemistry revealed that at least five K2P subtypes (TWIK1, TREK1, TREK2, TASK1, and TASK3) are expressed in the apical membrane. Apical UTP also increased the current, but pretreatment with the PKC inhibitor GF109203X blocked the response. Similarly, direct activation of PKC with phorbol 12-myristate 13-acetate produced a similar increase in current as observed with UTP. These results support the conclusion that the basal level of K+ secretion involves constitutive activity of apical KCa3.1 channels and multiple K2P channel subtypes. Apical UTP evoked a transient increase in KCa3.1 channel activity, but over time caused persistent inhibition of K2P channel function leading to an overall decrease in K+ secretion.


Subject(s)
Epithelial Cells/metabolism , Mammary Glands, Human/metabolism , Potassium Channels, Tandem Pore Domain/metabolism , Potassium/metabolism , Receptors, Purinergic P2Y/metabolism , Cell Line, Transformed , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Sodium Channels/metabolism , Female , Humans , Mammary Glands, Human/cytology , Mammary Glands, Human/drug effects , Membrane Potentials , Potassium Channel Blockers/pharmacology , Potassium Channels, Tandem Pore Domain/antagonists & inhibitors , Potassium Channels, Tandem Pore Domain/genetics , Protein Kinase C/metabolism , Purinergic P2Y Receptor Agonists/pharmacology , Receptors, Purinergic P2Y/drug effects , Secretory Pathway , Sodium/metabolism , Uridine Triphosphate/pharmacology
8.
Am J Physiol Heart Circ Physiol ; 311(1): H299-309, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27233766

ABSTRACT

Uridine adenosine tetraphosphate (Up4A), a dinucleotide, exerts vascular influence via purinergic receptors (PR). We investigated the effects of Up4A on angiogenesis and the putative PR involved. Tubule formation assay was performed in a three-dimensional system, in which human endothelial cells were cocultured with pericytes with various Up4A concentrations for 5 days. Expression of PR subtypes and angiogenic factors was assessed in human endothelial cells with and without P2Y6R antagonist. No difference in initial tubule formation was detected between Up4A stimulation and control conditions at day 2 In contrast, a significant increase in vascular density in response to Up4A was observed at day 5 Up4A at an optimal concentration of 5 µM promoted total tubule length, number of tubules, and number of junctions, all of which were inhibited by the P2Y6R antagonist MRS2578. Higher concentrations of Up4A (10 µM) had no effects on angiogenesis parameters. Up4A increased mRNA level of P2YRs (P2Y2R, P2Y4R, and P2Y6R) but not P2XR (P2X4R and P2X7R) or P1R (A2AR and A2BR), while Up4A upregulated VEGFA and ANGPT1, but not VEGFR2, ANGPT2, Tie1, and Tie2. In addition, Up4A increased VEGFA protein levels. Transcriptional upregulation of P2YRs by Up4A was inhibited by MRS2578. In conclusion, Up4A is functionally capable of promoting tubule formation in an in vitro coculture system, which is likely mediated by pyrimidine-favored P2YRs but not P2XRs or P1Rs, and involves upregulation of angiogenic factors.


Subject(s)
Angiogenesis Inducing Agents/pharmacology , Dinucleoside Phosphates/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Neovascularization, Physiologic/drug effects , Purinergic P2Y Receptor Agonists/pharmacology , Receptors, Purinergic P2Y/drug effects , Angiopoietin-1/genetics , Angiopoietin-1/metabolism , Cells, Cultured , Coculture Techniques , Dose-Response Relationship, Drug , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Pericytes/metabolism , Purinergic P2Y Receptor Antagonists/pharmacology , Receptors, Purinergic P2Y/genetics , Receptors, Purinergic P2Y/metabolism , Signal Transduction/drug effects , Time Factors , Transfection , Up-Regulation , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
9.
Arch Oral Biol ; 60(1): 160-6, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25455130

ABSTRACT

OBJECTIVE: To investigate the relationship between ATP and IL-6 in mechanical stress-induced REX-1 expression in SHEDs. METHODS: Cells were stimulated with mechanical stress (0-2.5 gcm(-2)), IL-6 (0.1-5 ng/ml), or ATP (10-100 µM) for 2h in serum-free media. IL-6 and REX-1 expression was examined by qualitative and quantitative polymerase chain reaction. ATP release was measured using a bioluminescence assay. The molecular mechanisms of the signalling pathways were investigated using chemical inhibitors. RESULTS: Mechanical stress induced IL-6 and REX-1 mRNA expression and ATP release. JAK inhibitor I inhibited the increase in REX-1 expression and ATP release but not IL-6 induction. Furthermore, suramin inhibited the upregulation of REX-1 mRNA expression but not ATP release. Exogenous IL-6 promoted both ATP release and REX-1 expression. The IL-6-induced REX-1 expression was attenuated by a P2Y1-specific receptor antagonist. Moreover, REX-1 expression was upregulated in a dose-dependent manner by the addition of ATP or a P2Y1 agonist. This inductive effect was abolished by the P2Y1-specific receptor antagonist. CONCLUSIONS: ATP-P2Y1 signalling is involved in IL-6-regulated stress-induced REX-1 expression in SHEDs. These results imply the participation of mechanical stress, IL-6, and ATP in regulating the expression of REX-1, a pluripotent stem cell marker.


Subject(s)
Adenosine Triphosphate/physiology , Interleukin-6/physiology , Kruppel-Like Transcription Factors/metabolism , Receptors, Purinergic P2Y/physiology , Tooth, Deciduous/cytology , Biomechanical Phenomena , Humans , Luminescent Measurements , Mechanotransduction, Cellular/physiology , Mesenchymal Stem Cells/physiology , Purinergic P2Y Receptor Agonists/pharmacology , Purinergic P2Y Receptor Antagonists/pharmacology , Receptors, Purinergic P2Y/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Stress, Mechanical , Up-Regulation
10.
Invest Ophthalmol Vis Sci ; 55(10): 6565-74, 2014 Sep 11.
Article in English | MEDLINE | ID: mdl-25212776

ABSTRACT

PURPOSE: We evaluated whether DA-6034 is involved in mucin secretion via P2Y receptor activation and/or intracellular Ca2+ concentration ([Ca2+]i) change. Also, we investigated the effect of P2Y receptor inhibitors or Ca2+ chelators on the DA-6034-induced mucin secretion and [Ca2+]i increases. METHODS: Effects of DA-6034 on mucin expression in primary, cultured, conjunctival epithelial cells was studied using RT-PCR, Western blot analysis, and periodic acid-schiff (PAS) staining. To evaluate thin film layer thickness generated by mucin and fluid secretion, cells were incubated in DA-6034 with/without P2Y antagonists or extracellular/intracellular Ca2+ chelators, and were imaged with confocal microscope using Texas Red-dextran dye. In addition, DA-6034-induced Ca2+-dependent Cl- channels opening was evaluated using perforated patch clamp. Fluo-4/AM was used to measure changes in [Ca2+]i induced by DA-6034 in Ca2+-free or Ca2+-containing buffered condition, as well as P2Y antagonists. RESULTS: DA-6034 induced the expression of mucin genes, production of mucin protein, and increase of number of mucin-secreting cells. P2Y antagonists inhibited DA-6034-induced mucin and fluid secretion, which was also affected by extracellular/intracellular Ca2+ chelators. DA-6034 stimulated Cl- channel opening and [Ca2+]i elevation. Further, [Ca2+]i increases induced by DA-6034 were lacking in either P2Y antagonists or Ca2+-free buffered condition, and diminished when endoplasmic reticulum Ca2+ was depleted by cyclopiazonic acid in Ca2+-free buffered condition. CONCLUSIONS: This study demonstrated that DA-6034 has a potential to induce mucin secretion via Ca2+-dependent pathways through P2Y receptors in multilayer, cultured, human conjunctival epithelial cells.


Subject(s)
Calcium/metabolism , Flavonoids/pharmacology , Intracellular Fluid/metabolism , Mucins/metabolism , Receptors, Purinergic P2Y/drug effects , Adult , Blotting, Western , Calcium Signaling , Cell Proliferation , Cells, Cultured , Conjunctiva/cytology , Conjunctiva/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gene Expression Regulation/drug effects , Humans , Membrane Potentials , Middle Aged , Mucins/drug effects , Mucins/genetics , Patch-Clamp Techniques , RNA/genetics , Receptors, Purinergic P2Y/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
11.
Am J Physiol Renal Physiol ; 306(9): F1088-97, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24553432

ABSTRACT

Extracellular ATP may contribute to Ca(2+) signaling in podocytes during tubuloglomerular feedback (TGF) and possibly as a result of local tissue damage. TRPC6 channels are Ca(2+)-permeable cationic channels that have been implicated in the pathophysiology of podocyte diseases. Here we show using whole cell recordings that ATP evokes robust activation of TRPC6 channels in mouse podocyte cell lines and in rat podocytes attached to glomerular capillaries in ex vivo glomerular explants. The EC50 for ATP is ~10 µM and is maximal at 100 µM, and currents were blocked by the P2 antagonist suramin. In terms of maximal currents that can be evoked, ATP is the strongest activator of podocyte TRPC6 that we have characterized to date. Smaller currents were observed in response to ADP, UTP, and UDP. ATP-evoked currents in podocytes were abolished by TRPC6 knockdown and by pretreatment with 10 µM SKF-96365 or 50 µM La(3+). ATP effects were also abolished by inhibiting G protein signaling and by the PLC/PLA2 inhibitor D-609. ATP effects on TRPC6 were also suppressed by knockdown of the slit diaphragm scaffolding protein podocin, and also by tempol, a membrane-permeable quencher of reactive oxygen species. Modulation of podocyte TRPC6 channels, especially in foot processes, could provide a mechanism for regulation of glomerular function by extracellular nucleotides, possibly leading to changes in permeation through slit diaphragms. These results raise the possibility that sustained ATP signaling could contribute to foot process effacement, Ca(2+)-dependent changes in gene expression, and/or detachment of podocytes.


Subject(s)
Adenosine Triphosphate/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Ion Channel Gating , Membrane Proteins/metabolism , Podocytes/metabolism , Reactive Oxygen Species/metabolism , Receptors, Purinergic P2Y/metabolism , TRPC Cation Channels/metabolism , Animals , Antioxidants/pharmacology , Calcium Signaling , Cell Line , Dose-Response Relationship, Drug , Gene Knockdown Techniques , Intracellular Signaling Peptides and Proteins/genetics , Ion Channel Gating/drug effects , Male , Membrane Potentials , Membrane Proteins/genetics , Mice , Phospholipase A2 Inhibitors/pharmacology , Phospholipases A2/metabolism , Podocytes/drug effects , Purinergic P2Y Receptor Antagonists/pharmacology , RNA Interference , Rats , Rats, Sprague-Dawley , Receptors, Purinergic P2Y/drug effects , TRPC Cation Channels/genetics , TRPC6 Cation Channel , Transfection , Type C Phospholipases/antagonists & inhibitors , Type C Phospholipases/metabolism
12.
J Assoc Res Otolaryngol ; 15(1): 13-30, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24263968

ABSTRACT

Phalloidin, a toxin isolated from the death cap mushroom, Amanita phalloides, binds to filamentous actin with high affinity, and this has made fluorophore-conjugated phalloidin a useful tool in cellular imaging. Hepatocytes take up phalloidin via the liver-specific organic anion transporting polypeptide 1b2, but phalloidin does not permeate most living cells. Rapid entry of styryl dyes into live hair cells has been used to evaluate function, but the usefulness of those fluorescence dyes is limited by broad and fixed absorption spectra. Since phalloidin can be conjugated to fluorophores with various spectra, we investigated whether it would permeate living hair cells. When we incubated mouse utricles in 66 nM phalloidin-CF488A and followed that by washes in phalloidin-free medium, we observed that it entered a subset of hair cells and labeled entire hair bundles fluorescently after 20 min. Incubations of 90 min labeled nearly all the hair bundles. When phalloidin-treated utricles were cultured for 24 h after washout, the label disappeared from the hair cells and progressively but heterogeneously labeled filamentous actin in the supporting cells. We investigated how phalloidin may enter hair cells and found that P2 receptor antagonists, pyridoxalphosphate-6-azophenyl-2', 4'-disulfonic acid and suramin, blocked phalloidin entry, while the P2Y receptor ligands, uridine-5'-diphosphate and uridine-5'-triphosphaste, stimulated uptake. Consistent with that, the P2Y6 receptor antagonist, MRS 2578, decreased phalloidin uptake. The results show that phalloidin permeates live hair cells through a pathway that requires metabotropic P2Y receptor signaling and suggest that phalloidin can be transferred from hair cells to supporting cells in culture.


Subject(s)
Amanita , Cell Membrane Permeability/physiology , Chromophore-Assisted Light Inactivation , Hair Cells, Auditory, Inner/metabolism , Phalloidine/pharmacokinetics , Plant Extracts/pharmacokinetics , Receptors, Purinergic P2Y/metabolism , Actins/metabolism , Animals , Cell Membrane Permeability/drug effects , Cells, Cultured , Fluorescent Dyes , Hair Cells, Auditory, Inner/cytology , Isothiocyanates/pharmacology , Mice , Models, Animal , Purinergic P2Y Receptor Antagonists/pharmacology , Receptors, Purinergic P2Y/drug effects , Second Messenger Systems/drug effects , Second Messenger Systems/physiology , Signal Transduction/drug effects , Signal Transduction/physiology , Thiourea/analogs & derivatives , Thiourea/pharmacology
13.
J Med Chem ; 56(10): 3749-67, 2013 May 23.
Article in English | MEDLINE | ID: mdl-23597047

ABSTRACT

Adenosine receptor (ARs) and P2Y receptors (P2YRs) that respond to extracellular nucleosides/nucleotides are associated with new directions for therapeutics. The X-ray structures of the A2AAR complexes with agonists and antagonists are examined in relationship to the G-protein-coupled receptor (GPCR) superfamily and applied to drug discovery. Much of the data on AR ligand structure from early SAR studies now are explainable from the A2AAR X-ray crystallography. The ligand-receptor interactions in related GPCR complexes can be identified by means of modeling approaches, e.g., molecular docking. Thus, molecular recognition in binding and activation processes has been studied effectively using homology modeling and applied to ligand design. Virtual screening has yielded new nonnucleoside AR antagonists, and existing ligands have been improved with knowledge of the receptor interactions. New agonists are being explored for central nervous system and peripheral therapeutics based on in vivo activity, such as chronic neuropathic pain. Ligands for receptors more distantly related to the X-ray template, i.e., P2YRs, have been introduced and are mainly used as pharmacological tools for elucidating the physiological role of extracellular nucleotides. Other ligand tools for drug discovery include fluorescent probes, radioactive probes, multivalent probes, and functionalized nanoparticles.


Subject(s)
Receptors, G-Protein-Coupled/drug effects , Receptors, Purinergic P1/drug effects , Receptors, Purinergic P2Y/drug effects , Adenosine A2 Receptor Agonists/chemical synthesis , Adenosine A2 Receptor Agonists/pharmacology , Adenosine A2 Receptor Antagonists/chemical synthesis , Adenosine A2 Receptor Antagonists/pharmacology , Animals , Drug Design , Drug Discovery , Fluorescent Dyes , High-Throughput Screening Assays , Humans , Indicators and Reagents , Ligands , Nanoconjugates , Radiopharmaceuticals , Receptor, Adenosine A2A/drug effects , Small Molecule Libraries , Structure-Activity Relationship , X-Ray Diffraction
14.
Curr Diabetes Rev ; 9(3): 228-36, 2013 May.
Article in English | MEDLINE | ID: mdl-23506378

ABSTRACT

Morbidity and mortality from diabetes mellitus (DM) are serious worldwide concerns. By the year 2030, the estimated number of diabetic patients will reach a staggering 439 million worldwide. Diabetes mellitus type 2 (DM2), which involves disturbances in both insulin secretion and resistance, is the most common form of diabetes and affects approximately 5 to 7% of the world's population. When a patient with DM2 cannot regulate his or her blood glucose levels through diet, weight loss, or exercise, oral medications, such as hypoglycemic agents (i.e., sulphonylureas, biguanides, alpha glucosidase inhibitors and thiazolidinediones), are crucial. Here, we discuss some physiological aspects of P2 receptors on pancreatic ß-cells, which express a variety of P2 receptor isoforms. These receptors enhance glucose-dependent insulin release. In addition, we speculate on the potential of purinergic compounds as novel or additional treatments for Type 2 Diabetes mellitus.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/pharmacology , Insulin-Secreting Cells/drug effects , Receptors, Purinergic P2X/drug effects , Receptors, Purinergic P2Y/drug effects , Receptors, Purinergic P2/drug effects , Animals , Cell Line , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/metabolism , Humans , Immunohistochemistry , Insulin-Secreting Cells/metabolism , Mice , Phosphorylation , Purinergic P2 Receptor Agonists/pharmacology , Purinergic P2Y Receptor Antagonists/pharmacology , Rats , Receptors, Purinergic P2/metabolism , Receptors, Purinergic P2X/metabolism , Receptors, Purinergic P2Y/metabolism , Reverse Transcriptase Polymerase Chain Reaction
15.
CNS Neurol Disord Drug Targets ; 11(6): 722-38, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22963441

ABSTRACT

P2Y receptors for extracellular nucleotides are coupled to activation of a variety of G proteins and stimulate diverse intracellular signaling pathways that regulate functions of cell types that comprise the central nervous system (CNS). There are 8 different subtypes of P2Y receptor expressed in cells of the CNS that are activated by a select group of nucleotide agonists. Here, the agonist selectivity of these 8 P2Y receptor subtypes is reviewed with an emphasis on synthetic agonists with high potency and resistance to degradation by extracellular nucleotidases that have potential applications as therapeutic agents. In addition, the recent identification of a wide variety of subtype-selective antagonists is discussed, since these compounds are critical for discerning cellular responses mediated by activation of individual P2Y receptor subtypes. The functional expression of P2Y receptor subtypes in cells that comprise the CNS is also reviewed and the role of each subtype in the regulation of physiological and pathophysiological responses is considered. Other topics include the role of P2Y receptors in the regulation of blood-brain barrier integrity and potential interactions between different P2Y receptor subtypes that likely impact tissue responses to extracellular nucleotides in the CNS. Overall, current research suggests that P2Y receptors in the CNS regulate repair mechanisms that are triggered by tissue damage, inflammation and disease and thus P2Y receptors represent promising targets for the treatment of neurodegenerative diseases.


Subject(s)
Blood-Brain Barrier/metabolism , Central Nervous System/metabolism , Inflammation/metabolism , Neurodegenerative Diseases/metabolism , Purinergic P2Y Receptor Agonists/pharmacology , Purinergic P2Y Receptor Antagonists/pharmacology , Receptors, Purinergic P2Y/physiology , Animals , Central Nervous System/drug effects , Humans , Receptors, Purinergic P2Y/drug effects , Receptors, Purinergic P2Y/metabolism
16.
Curr Pharm Des ; 18(37): 6169-80, 2012.
Article in English | MEDLINE | ID: mdl-23004340

ABSTRACT

In the last two decades a tremendous development has been noticed in our understanding of the purinergic system, consisting of heterogeneously expressed purinoceptor subtypes and its classical agonists: e.g., adenosine triphosphate, uridine triphosphate or complex dinucleoside polyphosphates. These agonists exert multiple effects on the vascular system: they regulate the relaxation and constriction of arterial blood vessels, lead to proliferation and migration in endothelial cells and vascular smooth muscle cells, and mediate potent proinflammatory responses or phenotypic cell changes. This review focuses on the P2 purinoceptor subtype P2Y and its pleiotropic effects in the vascular wall under physiological and pathophysiological condition. Various experimental and clinical studies provide evidence that pharmacological targeting of P2Y might be effective in reducing vascular alterations under disease conditions.


Subject(s)
Cardiovascular Agents/pharmacology , Purinergic P2Y Receptor Antagonists/pharmacology , Receptors, Purinergic P2Y/drug effects , Vascular Diseases/drug therapy , Animals , Cardiovascular Agents/chemistry , Drug Design , Humans , Ligands , Purinergic P2Y Receptor Antagonists/chemistry , Receptors, Purinergic P2Y/metabolism , Signal Transduction/drug effects , Vascular Diseases/metabolism
17.
Curr Pharm Des ; 18(33): 5294-308, 2012.
Article in English | MEDLINE | ID: mdl-22724416

ABSTRACT

The concept of "pharmacogenetics" addresses genetically determined variation in how individuals respond to drugs. Accordingly, specific genetic variants have been suggested as contributors to a reduced response to various antiplatelet drugs. Aspirin is a cornerstone in secondary cardiovascular prevention and has been thoroughly investigated. The efficacy of aspirin is well documented, although with considerable interindividual variation. According to meta-analyses, a reduced antiplatelet effect of aspirin confers an increased risk of cardiovascular events. The platelet response to aspirin is assessed by in vitro evaluation of thromboxane-dependent platelet function. A reduced antiplatelet effect of aspirin can be explained by several mechanisms, which are largely determined by clinical, pharmacodynamic, biological and genetic factors. During the past decade, numerous studies have identified genetic polymorphisms significantly associated with cardiovascular events and modulating the antiplatelet effect of aspirin. However, results have been contradictory allowing only few firm conclusions to be drawn. Polymorphisms in genes encoding glycoproteins (IIb/IIIa, Ia/IIa, VI and Ibα), cyclooxygenases (1 and 2), adenosine diphosphate receptors (P2Y1 and P2Y12) and proteins of importance for haemostasis (thromboxane A2 receptor, coagulation factor XIII, etc.) have been investigated. In particular, a polymorphism in the gene encoding glycoprotein IIb/IIIa has been associated with a reduced antiplatelet effect of aspirin. The additive value of an individual's genetic makeup in predicting the antiplatelet effect of aspirin and the risk of cardiovascular events remains controversial. The present review outlines the pharmacology of aspirin and provides an overview of specific genetic variations considered to influence the antiplatelet effect of aspirin.


Subject(s)
Aspirin/therapeutic use , Blood Platelets/drug effects , Cyclooxygenase Inhibitors/therapeutic use , Drug Resistance/genetics , Pharmacogenetics , Platelet Aggregation Inhibitors/therapeutic use , Polymorphism, Genetic , Purinergic P2Y Receptor Antagonists/therapeutic use , Animals , Aspirin/adverse effects , Blood Platelets/metabolism , Cyclooxygenase Inhibitors/adverse effects , Genotype , Hemorrhage/chemically induced , Hemorrhage/genetics , Humans , Phenotype , Platelet Aggregation Inhibitors/adverse effects , Platelet Function Tests , Platelet Membrane Glycoproteins/drug effects , Platelet Membrane Glycoproteins/genetics , Platelet Membrane Glycoproteins/metabolism , Prostaglandin-Endoperoxide Synthases/blood , Prostaglandin-Endoperoxide Synthases/genetics , Purinergic P2Y Receptor Antagonists/adverse effects , Receptors, Purinergic P2Y/blood , Receptors, Purinergic P2Y/drug effects , Receptors, Purinergic P2Y/genetics , Risk Factors , Treatment Outcome
18.
Pflugers Arch ; 463(2): 319-26, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22057585

ABSTRACT

Muscle activity is associated with an increase in extracellular purines (ATP, ADP), which are involved in signalling mechanisms. The present study investigates the effect of purines on the function of Na,K-ATPase (Na,K-pump) in rat skeletal muscle. Na,K-ATPase activity was quantified by measuring the release of inorganic phosphate in the presence of ATP and variable Na(+) concentrations. In membranes purified from glycolytic muscle fibres, purinergic stimulation increases V (max) and decreases the K (m) (higher Na(+) affinity) of the Na,K-ATPase. Stimulatory effects were obtained using ATP, ADP, 2-methylthio-ADP and UPT, but not UDP and adenosine. The effect of ADP on V (max) can be inhibited by the non-specific P2Y receptor antagonists, suramin and PPADS. Moreover, the P2Y(13) receptor antagonist MRS 2211 strongly inhibited the response to ADP, whereas the specific P2Y(1) receptor antagonist MRS 2500 had less effect. Based on results from these agonists and antagonists, we conclude that P2Y(13) receptors mediate the main effects observed, that P2Y1 receptors are also involved and that some P2Y(2)/P2Y(4) receptors also appear to be involved. Receptor antagonists had no effect on ADP-induced subunit (phospholemman and α1) phosphorylation and changes in K (m) (Na(+) affinity). Thus, the stimulatory effects of purines are mediated by two independent mechanisms: P2Y receptor-mediated increase in Na,K-ATPase capacity (increased V (max)) and P2Y receptor-independent phosphorylation of Na,K-ATPase phospholemman and α1 subunits, which induce changes in ion affinity. These mechanisms may contribute to up-regulation of Na,K-ATPase during muscle activity.


Subject(s)
Membrane Proteins/metabolism , Muscle, Skeletal/metabolism , Phosphoproteins/metabolism , Protein Subunits/metabolism , Receptors, Purinergic P2Y/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Sodium/metabolism , Adenosine Diphosphate/pharmacology , Adenosine Triphosphate/pharmacology , Animals , Male , Models, Animal , Muscle, Skeletal/drug effects , Phosphorylation , Purinergic P2Y Receptor Antagonists/pharmacology , Purines/pharmacology , Rats , Rats, Wistar , Receptors, Purinergic P2Y/drug effects , Sodium/pharmacology
19.
Neurosci Lett ; 504(3): 311-5, 2011 Oct 31.
Article in English | MEDLINE | ID: mdl-21982803

ABSTRACT

Defect of hypoxanthine phosphoribosyl transferase (HPRT) causes Lesch-Nyhan disease (LND), but the link between HPRT deficiency and the self-injurious behavior of LND is unknown. In a previous study (Pinto et al., J. Neurochem. 72 (2005) 1579-1586) we reported on a decrease in nucleotidase activity in membranes of several HPRT(-) cell lines and fibroblasts from LND patients. Since nucleotidases are involved in ATP-induced signal transduction, in the present study, we tested the hypothesis that P2X and P2Y receptor-mediated signal transduction is impaired in HPRT deficiency. As model we studied rat B103 neuroblastoma cells. Compared to control cells, in HPRT(-) cells, NTP and NDP-induced Ca(2+) influx across the membrane and Ca(2+) mobilization from intracellular stores were impaired. Both P2X and P2Y receptors were involved in the responses. Quantitative real-time PCR revealed reduced expression of receptors P2X(3), P2X(5), P2Y(2), P2Y(4), P2Y(12), P2Y(13) and P2Y(14) in HPRT deficiency. Collectively, HPRT deficiency is associated with abnormal purinergic signaling, encompassing P2X and P2Y receptors and nucleotidases.


Subject(s)
Hypoxanthine Phosphoribosyltransferase/deficiency , Neurons/physiology , Receptors, Purinergic P2X/physiology , Receptors, Purinergic P2Y/physiology , Signal Transduction/physiology , Animals , Calcium Signaling/drug effects , Calcium Signaling/physiology , Cell Line, Tumor/physiology , Lesch-Nyhan Syndrome , Neuroblastoma/pathology , Neurons/drug effects , Nucleotides/pharmacology , Rats , Real-Time Polymerase Chain Reaction , Receptors, Purinergic P2X/biosynthesis , Receptors, Purinergic P2X/drug effects , Receptors, Purinergic P2X/genetics , Receptors, Purinergic P2Y/biosynthesis , Receptors, Purinergic P2Y/drug effects , Receptors, Purinergic P2Y/genetics , Self-Injurious Behavior , Signal Transduction/drug effects
20.
Am J Physiol Heart Circ Physiol ; 301(2): H409-17, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21551273

ABSTRACT

Uridine adenosine tetraphosphate (Up(4)A) was reported as a novel endothelium-derived contracting factor. Up(4)A contains both purine and pyrimidine moieties, which activate purinergic (P2)X and P2Y receptors. However, alterations in the vasoconstrictor responses to Up(4)A in hypertensive states remain unclear. The present study examined the effects of Up(4)A on contraction of isolated renal arteries (RA) and pulmonary arteries (PA) from DOCA-salt rats using isometric tension recording. RA from DOCA-salt rats exhibited increased contraction to Up(4)A versus arteries from control uninephrectomized rats in the absence and presence of N(G)-nitro-l-arginine (nitric oxide synthase inhibitor). On the other hand, the Up(4)A-induced contraction in PA was similar between the two groups. Up(4)A-induced contraction was inhibited by suramin (nonselective P2 antagonist) but not by diinosine pentaphosphate pentasodium salt hydrate (Ip(5)I; P2X(1) antagonist) in RA from both groups. Furthermore, 2-thiouridine 5'-triphosphate tetrasodium salt (2-ThioUTP; P2Y(2) agonist)-, uridine-5'-(γ-thio)-triphosphate trisodium salt (UTPγS; P2Y(2)/P2Y(4) agonist)-, and 5-iodouridine-5'-O-diphosphate trisodium salt (MRS 2693; P2Y(6) agonist)-induced contractions were all increased in RA from DOCA-salt rats. Protein expression of P2Y(2)-, P2Y(4)-, and P2Y(6) receptors in RA was similar between the two groups. In DOCA-salt RA, the enhanced Up(4)A-induced contraction was reduced by PD98059, an ERK pathway inhibitor, and Up(4)A-stimulated ERK activation was increased. These data are the first to indicate that Up(4)A-induced contraction is enhanced in RA from DOCA-salt rats. Enhanced P2Y receptor signaling and activation of the ERK pathway together represent a likely mechanism mediating the enhanced Up(4)A-induced contraction. Up(4)A might be of relevance in the pathophysiology of vascular tone regulation and renal dysfunction in arterial hypertension.


Subject(s)
Dinucleoside Phosphates/pharmacology , Hypertension/physiopathology , Pulmonary Artery/drug effects , Renal Artery/drug effects , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology , Analysis of Variance , Animals , Blood Pressure , Body Weight , Desoxycorticosterone , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Hypertension/chemically induced , Hypertension/metabolism , Male , Myography , Nephrectomy , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Pulmonary Artery/metabolism , Pulmonary Artery/physiopathology , Purinergic Antagonists/pharmacology , Purinergic P2Y Receptor Agonists/pharmacology , Rats , Rats, Wistar , Receptors, Purinergic P2Y/drug effects , Receptors, Purinergic P2Y/metabolism , Renal Artery/metabolism , Renal Artery/physiopathology , Sodium Chloride, Dietary
SELECTION OF CITATIONS
SEARCH DETAIL
...