Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Purinergic Signal ; 15(3): 397-402, 2019 09.
Article in English | MEDLINE | ID: mdl-31286385

ABSTRACT

A P2X1-eYFP knock-in mouse was generated to study receptor expression and mobility in smooth muscle and blood cells. eYFP was added to the C-terminus of the P2X1R and replaced the native P2X1R. Fluorescence corresponding to P2X1-eYFPR was detected in urinary bladder smooth muscle, platelets and megakaryocytes. ATP-evoked currents from wild type and P2X1-eYFP isolated urinary bladder smooth muscle cells had the same peak current amplitude and time-course showing that the eYFP addition had no obvious effect on properties. Fluorescence recovery after photobleaching (FRAP) in bladder smooth muscle cells demonstrated that surface P2X1Rs are mobile and their movement is reduced following cholesterol depletion. Compared to the platelet and megakaryocyte, P2X1-eYFP fluorescence was negligible in red blood cells and the majority of smaller marrow cells. The spatial pattern of P2X1-eYFP fluorescence in the megakaryocyte along with FRAP assessment of mobility suggested that P2X1Rs are expressed extensively throughout the membrane invagination system of this cell type. The current study highlights that the spatiotemporal properties of P2X1R expression can be monitored in real time in smooth muscle cells and megakaryocytes/platelets using the eYFP knock-in mouse model.


Subject(s)
Gene Knock-In Techniques/methods , Receptors, Purinergic P2X1/analysis , Receptors, Purinergic P2X1/metabolism , Animals , Bacterial Proteins , Luminescent Proteins , Mice , Models, Animal
2.
J Immunol ; 196(12): 4877-84, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27183585

ABSTRACT

Eosinophils play an important role in the pathogenesis of asthma and can be activated by extracellular nucleotides released following cell damage or inflammation. For example, increased ATP concentrations were reported in bronchoalveolar lavage fluids of asthmatic patients. Although eosinophils are known to express several subtypes of P2 receptors for extracellular nucleotides, their function and contribution to asthma remain unclear. In this article, we show that transcripts for P2X1, P2X4, and P2X5 receptors were expressed in healthy and asthmatic eosinophils. The P2X receptor agonist α,ß-methylene ATP (α,ß-meATP; 10 µM) evoked rapidly activating and desensitizing inward currents (peak 18 ± 3 pA/pF at -60 mV) in healthy eosinophils, typical of P2X1 homomeric receptors, which were abolished by the selective P2X1 antagonist NF449 (1 µM) (3 ± 2 pA/pF). α,ß-meATP-evoked currents were smaller in eosinophils from asthmatic patients (8 ± 2 versus 27 ± 5 pA/pF for healthy) but were enhanced following treatment with a high concentration of the nucleotidase apyrase (17 ± 5 pA/pF for 10 IU/ml and 11 ± 3 pA/pF for 0.32 IU/ml), indicating that the channels are partially desensitized by extracellular nucleotides. α,ß-meATP (10 µM) increased the expression of CD11b activated form in eosinophils from healthy, but not asthmatic, donors (143 ± 21% and 108 ± 11% of control response, respectively). Furthermore, α,ß-meATP increased healthy (18 ± 2% compared with control 10 ± 1%) but not asthmatic (13 ± 1% versus 10 ± 0% for control) eosinophil adhesion. Healthy human eosinophils express functional P2X1 receptors whose activation leads to eosinophil αMß2 integrin-dependent adhesion. P2X1 responses are constitutively reduced in asthmatic compared with healthy eosinophils, probably as the result of an increase in extracellular nucleotide concentration.


Subject(s)
Asthma/immunology , Cell Adhesion , Eosinophils/physiology , Receptors, Purinergic P2X1/metabolism , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Apyrase/pharmacology , Asthma/physiopathology , Benzenesulfonates/pharmacology , CD11b Antigen/genetics , CD11b Antigen/metabolism , Eosinophils/drug effects , Eosinophils/immunology , Healthy Volunteers , Humans , Leukocyte Count , Purinergic P2X Receptor Agonists/pharmacology , Real-Time Polymerase Chain Reaction , Receptors, Purinergic P2X1/genetics , Receptors, Purinergic P2X4/genetics , Receptors, Purinergic P2X5/genetics , Reverse Transcriptase Polymerase Chain Reaction
3.
J Biol Chem ; 289(41): 28629-39, 2014 Oct 10.
Article in English | MEDLINE | ID: mdl-25160621

ABSTRACT

In this work, we report that Entpd1(-/-) mice, deficient for the ectonucleotidase nucleoside triphosphate diphosphohydrolase-1 (NTPDase1), produce smaller litters (27% reduction) compared with wild-type C57BL6 animals. This deficit is linked to reduced in vivo oocyte fertilization by Entpd1(-/-) males (61 ± 11% versus 88 ± 7% for Entpd1(+/+)). Normal epididymal sperm count, spermatozoa morphology, capacitation, and motility and reduced ejaculated sperm number (2.4 ± 0.5 versus 3.7 ± 0.4 million for Entpd1(+/+)) pointed to vas deferens dysfunction. NTPDase1 was localized by immunofluorescence in the tunica muscularis of the vas deferens. Its absence resulted in a major ATP hydrolysis deficiency, as observed in situ by histochemistry and in primary smooth muscle cell cultures. In vitro, Entpd1(-/-) vas deferens displayed an exacerbated contraction to ATP, a diminished response to its non-hydrolysable analog αßMeATP, and a reduced contraction to electrical field stimulation, suggesting altered P2X1 receptor function with a propensity to desensitize. This functional alteration was accompanied by a 3-fold decrease in P2X1 protein expression in Entpd1(-/-) vas deferens with no variation in mRNA levels. Accordingly, exogenous nucleotidase activity was required to fully preserve P2X1 receptor activation by ATP in vitro. Our study demonstrates that NTPDase1 is required to maintain normal P2X1 receptor functionality in the vas deferens and that its absence leads to impaired peristalsis, reduced spermatozoa concentration in the semen, and, eventually, reduced fertility. This suggests that alteration of NTPDase1 activity affects ejaculation efficacy and male fertility. This work may contribute to unveil a cause of infertility and open new therapeutic potentials.


Subject(s)
Antigens, CD/genetics , Apyrase/genetics , Infertility, Male/genetics , Oligospermia/genetics , Receptors, Purinergic P2X1/genetics , Spermatozoa/physiology , Vas Deferens/enzymology , Adenosine Triphosphate/metabolism , Animals , Apyrase/deficiency , Ejaculation , Epididymis/enzymology , Epididymis/physiopathology , Female , Gene Expression Regulation , Infertility, Male/enzymology , Infertility, Male/physiopathology , Male , Mice , Mice, Knockout , Muscle Contraction , Muscle, Smooth/enzymology , Muscle, Smooth/physiopathology , Oligospermia/enzymology , Oligospermia/physiopathology , Oocytes/physiology , Receptors, Purinergic P2X1/metabolism , Sperm Capacitation , Vas Deferens/physiopathology
4.
Proc Natl Acad Sci U S A ; 109(12): 4663-7, 2012 Mar 20.
Article in English | MEDLINE | ID: mdl-22393010

ABSTRACT

P2X receptors for ATP have a wide range of physiological roles and comprise a structurally distinct family of ligand-gated trimeric ion channels. The crystal structure of a P2X4 receptor, in combination with mutagenesis studies, has provided a model of the intersubunit ATP-binding sites and identified an extracellular lateral portal, adjacent to the membrane, that funnels ions to the channel pore. However, little is known about the extent of ATP-induced conformational changes in the extracellular domain of the receptor. To address this issue, we have used MTSEA-biotinylation (N-Biotinoylaminoethyl methanethiosulfonate) to show ATP-sensitive accessibility of cysteine mutants at the human P2X1 receptor. Mapping these data to a P2X1 receptor homology model identifies significant conformational rearrangement. Electron microscopy of purified P2X1 receptors showed marked changes in structure on ATP binding, and introducing disulphide bonds between adjacent subunits to restrict intersubunit movements inhibited channel function. These results are consistent with agonist-induced rotation of the propeller-head domain of the receptor, sliding of adjacent subunits leading to restricted access to the upper vestibule, movement in the ion conducting lateral portals, and gating of the channel pore.


Subject(s)
Receptors, Purinergic P2X1/chemistry , Adenosine Triphosphate/chemistry , Animals , Binding Sites , Biotinylation , Disulfides/chemistry , Humans , Ions/chemistry , Microscopy, Electron/methods , Molecular Conformation , Mutagenesis , Oocytes/metabolism , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Xenopus
5.
J Immunol ; 186(2): 1209-17, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21149610

ABSTRACT

We investigated whether adenosine, a potent contributor to the regulation of pulmonary function, can modulate human lung mast cell (HLMC) fibrinolytic activity. Tissue plasminogen activator (tPA) activity and tPA transcript expression levels from a human mast cell line (HMC-1) and HLMC were monitored following adenosine application. Adenosine potentiated mast cell tPA activity and tPA gene expression in a dose-dependent manner. Adenosine effects were abolished in the presence of adenosine deaminase. HMC-1 cells and HLMC predominantly expressed adenosine A(2A) and A(2B) receptor transcripts (A(2B) ≈ A(2A) > A(3) >> A(1)). Pharmacological and signaling studies suggest that the A(2A) receptor is the major subtype accounting for adenosine-induced mast cell tPA activity. Finally, the supernatant from HMC-1 cells and HLMC treated with adenosine (for 24 h) significantly increased fibrin clot lysis, whereas ZM241385, an A(2A) receptor antagonist, abolished this effect. To our knowledge, this study provides the first data to demonstrate the potentiating effect of adenosine on mast cell tPA activity and fibrin clot lysis.


Subject(s)
Adenosine/pharmacology , Lung/enzymology , Mast Cells/enzymology , Tissue Plasminogen Activator/metabolism , Blood Coagulation Tests , Cell Line , Drug Synergism , Enzyme Activation/drug effects , Enzyme Activation/physiology , Fibrinolysis/physiology , Humans , Lung/cytology , Lung/drug effects , Mast Cells/cytology , Mast Cells/drug effects , Receptor, Adenosine A2A/metabolism , Tissue Plasminogen Activator/biosynthesis , Up-Regulation/drug effects , Up-Regulation/immunology
6.
J Physiol ; 587(Pt 21): 5275-88, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19736301

ABSTRACT

Contraction of urinary bladder smooth muscle (UBSM) is caused by the release of ATP and ACh from parasympathetic nerves. Although both purinergic and muscarinic pathways are important to contraction, their relative contributions and signalling mechanisms are not well understood. Here, the contributions of each pathway to urinary bladder contraction and the underlying electrical and Ca(2+) signalling events were examined in UBSM strips from wild type mice and mice deficient in P2X1 receptors (P2X1(-/-)) before and after pharmacological inhibition of purinergic and muscarinic receptors. Electrical field stimulation was used to excite parasympathetic nerves to increase action potentials, Ca(2+) flash frequency, and force. Loss of P2X1 function not only eliminated action potentials and Ca(2+) flashes during stimulation, but it also led to a significant increase in Ca(2+) flashes following stimulation and a corresponding increase in the force transient. Block of muscarinic receptors did not affect action potentials or Ca(2+) flashes during stimulation, but prevented them following stimulation. These findings indicate that nerve excitation leads to rapid engagement of smooth muscle P2X1 receptors to increase action potentials (Ca(2+) flashes) during stimulation, and a delayed increase in excitability in response to muscarinic receptor activation. Together, purinergic and muscarinic stimulation shape the time course of force transients. Furthermore, this study reveals a novel inhibitory effect of P2X1 receptor activation on subsequent increases in muscarinic-driven excitability and force generation.


Subject(s)
Action Potentials/physiology , Calcium Signaling/physiology , Muscle Contraction/physiology , Muscle, Smooth/innervation , Muscle, Smooth/physiology , Receptors, Purinergic P2/metabolism , Urinary Bladder/innervation , Urinary Bladder/physiology , Animals , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Inhibition/physiology , Receptors, Purinergic P2X
7.
Biochem Biophys Res Commun ; 350(1): 244-8, 2006 Nov 10.
Article in English | MEDLINE | ID: mdl-16997281

ABSTRACT

The intracellular amino and carboxy termini of P2X receptors have been shown to contribute to the regulation of ATP evoked currents. In this study we produced, and expressed in Xenopus oocytes, individual alanine point mutants of positively charged amino acids (eight lysine, seven arginine and one histidine) in the intracellular domains of the human P2X1 receptor. The majority of these mutations had no effect on the amplitude, time-course or rectification of ATP evoked currents. In contrast the mutant K367A was expressed at normal levels at the cell surface however ATP evoked currents were reduced by >99% and desensitised more rapidly demonstrating a role of K367 in channel regulation. This is similar to that previously described for T18A mutant channels. Co-expression of T18A and K367A mutant P2X1 receptors produced larger ATP evoked responses than either mutant alone and suggests that these amino and carboxy terminal regions interact to regulate channel function.


Subject(s)
Ion Channels/chemistry , Ion Channels/metabolism , Receptors, Purinergic P2/chemistry , Receptors, Purinergic P2/metabolism , Animals , Electrophysiology , Humans , Ion Channel Gating , Ion Channels/genetics , Lysine/genetics , Lysine/metabolism , Mutation/genetics , Oocytes , Patch-Clamp Techniques , Protein Subunits/genetics , Protein Subunits/metabolism , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2X , Xenopus laevis
8.
Pflugers Arch ; 452(5): 486-500, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16607539

ABSTRACT

P2X receptors for adenosine tri-phosphate (ATP) are a distinct family of ligand-gated cation channels with two transmembrane domains, intracellular amino and carboxy termini and a large extracellular ligand binding loop. Seven genes (P2X(1-7)) have been cloned and the channels form as either homo or heterotrimeric channels giving rise to a wide range of phenotypes. This review aims to give an account of recent work on the molecular basis of the properties of P2X receptors. In particular, to consider emerging information on the assembly of P2X receptor subunits, channel regulation and desensitisation, targeting, the molecular basis of drug action and the functional contribution of P2X receptors to physiological processes.


Subject(s)
Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Ion Channel Gating/physiology , Receptors, Purinergic P2/chemistry , Receptors, Purinergic P2/metabolism , Amino Acid Sequence , Animals , Binding Sites , Humans , Mice , Models, Chemical , Models, Molecular , Molecular Sequence Data , Protein Binding , Receptors, Purinergic P2X2 , Structure-Activity Relationship
9.
Biochem Biophys Res Commun ; 343(2): 415-9, 2006 May 05.
Article in English | MEDLINE | ID: mdl-16546137

ABSTRACT

ATP-stimulated P2X1 and ADP-stimulated P2Y1 receptors play important roles in platelet activation. An increase in intracellular Ca2+ represents a key signalling event coupled to both of these receptors, mediated via direct gating of Ca2+-permeable channels in the case of P2X1 and phospholipase-C-dependent Ca2+ mobilisation for P2Y1. We show that disruption of cholesterol-rich membrane lipid rafts reduces P2X1 receptor-mediated calcium increases by approximately 80%, while P2Y1 receptor-dependent Ca2+ release is unaffected. In contrast to artery, vas deferens, bladder smooth muscle, and recombinant expression in cell lines, where P2X1 receptors show almost exclusive association with lipid rafts, only approximately 20% of platelet P2X1 receptors are co-expressed with the lipid raft marker flotillin-2. We conclude that lipid rafts play a significant role in the regulation of P2X1 but not P2Y1 receptors in human platelets and that a reserve of non-functional P2X1 receptors may exist.


Subject(s)
Blood Platelets/metabolism , Calcium/metabolism , Differential Threshold/physiology , Membrane Fluidity/physiology , Membrane Microdomains/metabolism , Receptors, Purinergic P2/metabolism , Blood Platelets/chemistry , Calcium/chemistry , Cells, Cultured , Humans , Membrane Microdomains/chemistry , Receptors, Purinergic P2/chemistry , Receptors, Purinergic P2X , Receptors, Purinergic P2Y1
10.
J Neurosci ; 25(34): 7734-42, 2005 Aug 24.
Article in English | MEDLINE | ID: mdl-16120774

ABSTRACT

The whole-cell patch-clamp technique was used to record current responses to nucleotides and nucleosides in human embryonic kidney HEK293 cells transfected with the human purinergic P2X3 receptor. When guanosine 5'-O-(3-thiodiphosphate) was included into the pipette solution, UTP at concentrations that did not alter the holding current facilitated the alpha,beta-methylene ATP (alpha,beta-meATP)-induced current. ATP and GTP, but not UDP or uridine, had an effect similar to that of UTP. Compounds known to activate protein kinase C (PKC) acted like the nucleoside triphosphates investigated, whereas various PKC inhibitors invariably reduced the effects of both PKC activators and UTP. The substitution by Ala of Ser/Thr residues situated within PKC consensus sites of the P2X3 receptor ectodomain either abolished (PKC2 and PKC3; T134A, S178A) or did not alter (PKC4 and PKC6; T196A, S269A) the UTP-induced potentiation of the alpha,beta-meATP current. Both the blockade of ecto-protein kinase C activity and the substitution of Thr-134 or Ser-178 by Ala depressed the maximum of the concentration-response curve for alpha,beta-meATP without altering the EC50 values. Molecular simulation of the P2X3 receptor structure indicated no overlap between assumed nucleotide binding domains and the relevant phosphorylation sites PKC2 and PKC3. alpha,beta-meATP-induced currents through native homomeric P2X3 receptors of rat dorsal root ganglia were also facilitated by UTP. In conclusion, it is suggested that low concentrations of endogenous nucleotides in the extracellular space may prime the sensitivity of P2X3 receptors toward the effect of subsequently applied (released) higher agonistic concentrations. The priming effect of nucleotides might be attributable to a phosphorylation of PKC sites at the ectodomain of P2X3 receptors.


Subject(s)
Protein Kinase C/physiology , Protein Kinases/physiology , Receptors, Purinergic P2/metabolism , Animals , Cell Line , Cells, Cultured , Dose-Response Relationship, Drug , Humans , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Purinergic P2 Receptor Agonists , Rats , Rats, Wistar , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2X3 , Recombinant Proteins/agonists , Recombinant Proteins/metabolism , Uridine Triphosphate/pharmacology
11.
J Biol Chem ; 280(35): 30705-11, 2005 Sep 02.
Article in English | MEDLINE | ID: mdl-16006561

ABSTRACT

P2X1 receptors for ATP are ligand-gated cation channels expressed on a range of smooth muscle preparations and blood platelets. The receptors appear to be clustered close to sympathetic nerve varicosities and mediate the underlying membrane potential changes and constriction following nerve stimulation in a range of arteries and resistance arterioles. In this study we have used discontinuous sucrose density gradients, Western blot analysis, and cholesterol measurements to show that recombinant and smooth muscle (rat tail artery, vas deferens, and bladder) P2X1 receptors are present in cholesterol-rich lipid rafts and co-localize with the lipid raft markers flotillin-1 and -2. Lipid rafts are specialized lipid membrane microdomains involved in signaling and trafficking. To determine whether lipid raft association was essential for P2X1 receptor channel function we used the cholesterol-depleting agent methyl-beta-cyclodextrin (10 mm for 1 h). This led to a redistribution of the P2X1 receptor throughout the sucrose gradient and reduced P2X1 receptor-mediated (alpha,beta-methylene ATP, 10 microm) currents in HEK293 cells by >90% and contractions of the rat tail artery by approximately 50%. However contractions evoked by potassium chloride (60 mm) were unaffected by methyl-beta-cyclodextrin and the inactive analogue alpha-cyclodextrin had no effect on P2X1 receptor-mediated currents or contractions. P2X1 receptors are subject to ongoing regulation by receptors and kinases, and the present results suggest that lipid rafts are an essential component in the maintenance of these localized signaling domains and play an important role in P2X1 receptor-mediated control of arteries.


Subject(s)
Arteries/metabolism , Membrane Microdomains/metabolism , Receptors, Purinergic P2/metabolism , Vasoconstriction/physiology , Animals , Cell Fractionation , Cell Line , Cholesterol/metabolism , Humans , Male , Membrane Microdomains/chemistry , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Rats , Rats, Wistar , Receptors, Purinergic P2X , Signal Transduction/physiology , alpha-Cyclodextrins/metabolism , beta-Cyclodextrins/metabolism
12.
Blood ; 106(5): 1644-51, 2005 Sep 01.
Article in English | MEDLINE | ID: mdl-15914557

ABSTRACT

The difficulty of conducting electrophysiologic recordings from the platelet has restricted investigations into the role of ion channels in thrombosis and hemostasis. We now demonstrate that the well-established synergy between P2Y(1) and P2Y(12) receptors during adenosine diphosphate (ADP)-dependent activation of the platelet alpha(IIb)beta(3) integrin also exists in murine marrow megakaryocytes, further supporting the progenitor cell as a bona fide model of platelet P2 receptor signaling. In patch clamp recordings, ADP (30 microM) stimulated a transient inward current at -70 mV, which was carried by Na(+) and Ca(2+) and was amplified by phenylarsine oxide, a potentiator of certain transient receptor potential (TRP) ion channels by phosphatidylinositol 4,5-bisphosphate depletion. This initial current decayed to a sustained phase, upon which repetitive transient inward cation currents with pre-dominantly P2X(1)-like kinetics were super-imposed. Abolishing P2X(1)-receptor activity prevented most of the repetitive currents, consistent with their activation by secreted adenosine triphosphate (ATP). Recordings in P2Y(1)-receptor-deficient megakaryocytes demonstrated an essential requirement of this receptor for activation of all ADP-evoked inward currents. However, P2Y(12) receptors, through the activation of PI3-kinase, played a synergistic role in both P2Y(1) and P2X(1)-receptor-dependent currents. Thus, direct stimulation of P2Y(1) and P2Y(12) receptors, together with autocrine P2X(1) activation, is responsible for the activation of nonselective cation currents by the platelet agonist ADP.


Subject(s)
Adenosine Diphosphate Ribose/pharmacology , Megakaryocytes/metabolism , Membrane Proteins/metabolism , Receptors, Purinergic P2/physiology , Animals , Arsenicals/pharmacology , Blood Platelets/chemistry , Blood Platelets/physiology , Calcium/metabolism , Cations/metabolism , Cell Membrane/drug effects , Cell Membrane/metabolism , Electric Conductivity , Megakaryocytes/drug effects , Membrane Proteins/drug effects , Mice , Mice, Inbred C57BL , Models, Animal , Receptors, Purinergic P2/drug effects , Receptors, Purinergic P2/metabolism , Receptors, Purinergic P2X , Receptors, Purinergic P2Y1 , Receptors, Purinergic P2Y12 , Signal Transduction/drug effects , Signal Transduction/physiology
13.
J Biol Chem ; 280(2): 1490-8, 2005 Jan 14.
Article in English | MEDLINE | ID: mdl-15528188

ABSTRACT

Emerging evidence suggests that Ca2+ release evoked by certain G-protein-coupled receptors can be voltage-dependent; however, the relative contribution of different components of the signaling cascade to this response remains unclear. Using the electrically inexcitable megakaryocyte as a model system, we demonstrate that inositol 1,4,5-trisphosphate-dependent Ca2+ mobilization stimulated by several agonists acting via Galphaq-coupled receptors is potentiated by depolarization and that this effect is most pronounced for ADP. Voltage-dependent Ca2+ release was not induced by direct elevation of inositol 1,4,5-trisphosphate, by agents mimicking diacylglycerol actions, or by activation of phospholipase Cgamma-coupled receptors. The response to voltage did not require voltage-gated Ca2+ channels as it persisted in the presence of nifedipine and was only weakly affected by the holding potential. Strong predepolarizations failed to affect the voltage-dependent Ca2+ increase; thus, an alteration of G-protein betagamma subunit binding is also not involved. Megakaryocytes from P2Y1(-/-) mice lacked voltage-dependent Ca2+ release during the application of ADP but retained this response after stimulation of other Galphaq-coupled receptors. Although depolarization enhanced Ca2+ mobilization resulting from GTPgammaS dialysis and to a lesser extent during AlF4- or thimerosal, these effects all required the presence of P2Y1 receptors. Taken together, the voltage dependence to Ca2+ release via Galphaq-coupled receptors is not due to control of G-proteins or down-stream signals but, rather, can be explained by a voltage sensitivity at the level of the receptor itself. This effect, which is particularly robust for P2Y1 receptors, has wide-spread implications for cell signaling.


Subject(s)
GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Inositol 1,4,5-Trisphosphate/metabolism , Ion Channel Gating , Receptors, G-Protein-Coupled/metabolism , Receptors, Purinergic P2/metabolism , Signal Transduction , Adenosine Diphosphate/metabolism , Adenosine Diphosphate/pharmacology , Aluminum Compounds/pharmacology , Animals , Calcium/metabolism , Calcium Channels, L-Type/metabolism , Calcium Signaling/drug effects , Diglycerides/metabolism , Enzyme Activation/drug effects , Fluorides/pharmacology , Ion Channel Gating/drug effects , Male , Megakaryocytes/drug effects , Megakaryocytes/metabolism , Membrane Potentials/drug effects , Mice , Mice, Inbred C57BL , Nifedipine/pharmacology , Phospholipase C gamma , Rats , Rats, Wistar , Receptors, G-Protein-Coupled/agonists , Receptors, Purinergic P2/deficiency , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2Y1 , Signal Transduction/drug effects , Thimerosal/pharmacology , Type C Phospholipases/metabolism
14.
Trends Pharmacol Sci ; 25(9): 487-93, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15559251

ABSTRACT

P2X receptors for ATP are expressed throughout the body and mediate a multitude of functions, including muscle contraction, neuronal excitability and bone formation. In the mid-1990s seven genes encoding P2X receptors (P2X(1-7)) were identified. These receptors comprised a novel family of ligand-gated ion channels with subunits that possessed intracellular N- and C-termini, two transmembrane domains and an extracellular ligand-binding loop. No crystal structures are available for these channels. Furthermore, they are distinct from the nicotinic acetylcholine (Cys-loop) and glutamate families of ion channels and have no similarity to other ATP-binding proteins, thus precluding homology modelling-based studies of their structural properties. However, molecular techniques have provided insight into the properties of P2X receptors: mutagenesis and biochemical studies have identified regions associated with ATP binding, ionic conduction, channel gating and regulation. In addition, transgenic approaches have helped to characterize the role of defined receptor subunits in native systems.


Subject(s)
Adenosine Triphosphate/physiology , Ion Channels/physiology , Receptors, Purinergic P2/physiology , Animals , Humans , Ion Channel Gating , Ion Channels/chemistry , Purinergic P2 Receptor Agonists , Receptors, Purinergic P2/chemistry
15.
J Physiol ; 558(Pt 3): 745-57, 2004 Aug 01.
Article in English | MEDLINE | ID: mdl-15181160

ABSTRACT

The role of P2 receptors in synaptic transmission to the rat medial nucleus of the trapezoid body (MNTB) was studied in an in vitro brain slice preparation. Whole-cell patch recordings were made and spontaneous synaptic responses studied under voltage clamp during application of P2X receptor agonists. ATPgammaS (100 microm) had no effect on holding current, but facilitated spontaneous excitatory postsynaptic current (sEPSC) frequency in 41% of recordings and facilitated spontaneous inhibitory postsynaptic currents (sIPSCs) in 20% of recordings. These were blocked by the P2 receptor antagonist suramin (100 microm). alpha,beta-meATP also facilitated sEPSC and sIPSC frequency, while l-beta,gamma-meATP facilitated only sIPSCs. The sEPSC facilitation by ATPgammaS was blocked by TTX (but did not block facilitation of sIPSCs). sEPSC facilitation was blocked by PPADS (30 microm) and the selective P2X(3) receptor antagonist A-317491 (3 microm), suggesting that modulation of sEPSCs involves P2X(3) receptor subunits. alpha,beta-meATP-facilitated sIPSCs were also recorded in wild-type mouse MNTB neurones, but were absent in the MNTB from P2X(1) receptor-deficient mice demonstrating a functional role for P2X(1) receptors in the CNS.


Subject(s)
Adenosine Triphosphate/analogs & derivatives , Brain Stem/physiology , Excitatory Postsynaptic Potentials/physiology , Neural Inhibition/physiology , Receptors, Purinergic P2/physiology , Synapses/physiology , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Adenosine Triphosphate/pharmacology , Animals , Brain Stem/drug effects , Cell Line , Excitatory Postsynaptic Potentials/drug effects , Humans , In Vitro Techniques , Mice , Mice, Knockout , Neural Inhibition/drug effects , Protein Subunits/agonists , Protein Subunits/antagonists & inhibitors , Protein Subunits/deficiency , Protein Subunits/physiology , Purinergic P2 Receptor Agonists , Purinergic P2 Receptor Antagonists , Rats , Receptors, Purinergic P2/deficiency , Synapses/drug effects
16.
Biochem J ; 382(Pt 1): 101-10, 2004 Aug 15.
Article in English | MEDLINE | ID: mdl-15144237

ABSTRACT

P2X1 receptors for ATP are ligand-gated cation channels, which mediate smooth muscle contraction, contribute to blood clotting and are co-expressed with a range of GPCRs (G-protein-coupled receptors). Stimulation of Galpha(q)-coupled mGluR1alpha (metabotropic glutamate receptor 1alpha), P2Y1 or P2Y2 receptors co-expressed with P2X(1) receptors in Xenopus oocytes evoked calcium-activated chloride currents (I(ClCa)) and potentiated subsequent P2X1-receptor-mediated currents by up to 250%. The mGluR1alpha-receptor-mediated effects were blocked by the phospholipase C inhibitor U-73122. Potentiation was mimicked by treatment with the phor-bol ester PMA. P2X receptors have a conserved intracellular PKC (protein kinase C) site; however, GPCR- and PMA-mediated potentiation was still observed with point mutants in which this site was disrupted. Similarly, the potentiation by GPCRs or PMA was unaffected by chelating the intracellular calcium rise with BAPTA/AM [bis(o-aminophenoxy)ethane-N,N,N',N'-tetra-acetic acid tetrakis-(acetoxymethyl ester)] or the PKC inhibitors Ro-32-0432 and bisindolylmaleimide I, suggesting that the regulation does not involve a calcium-sensitive form of PKC. However, both GPCR and PMA potentiation were blocked by the kinase inhibitor staurosporine. Potentiation by phorbol esters was recorded in HEK-293 cells expressing P2X1 receptors, and radiolabelling of phosphorylated proteins in these cells demonstrated that P2X1 receptors are basally phosphorylated and that this level of phosphorylation is unaffected by phorbol ester treatment. This demonstrates that P2X1 regulation does not result directly from phosphorylation of the channel, but more likely by a staurosporine-sensitive phosphorylation of an accessory protein in the P2X1 receptor complex and suggests that in vivo fine-tuning of P2X1 receptors by GPCRs may contribute to cardiovascular control and haemostasis.


Subject(s)
Receptors, G-Protein-Coupled/metabolism , Receptors, Purinergic P2/metabolism , Animals , Calcium/metabolism , Cations , Cell Line , Cloning, Molecular/methods , Enzyme Activation/physiology , Humans , Ion Channel Gating , Kidney/chemistry , Kidney/embryology , Kidney/enzymology , Kidney/metabolism , Oocytes/chemistry , Oocytes/enzymology , Oocytes/metabolism , Patch-Clamp Techniques/methods , Phosphorylation , Protein-Tyrosine Kinases/metabolism , Receptor, Insulin/metabolism , Receptors, Metabotropic Glutamate/metabolism , Receptors, Purinergic P2X , Receptors, Purinergic P2Y1 , Receptors, Purinergic P2Y2 , Time Factors , Type C Phospholipases/metabolism , Xenopus
17.
Mol Pharmacol ; 65(4): 979-85, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15044628

ABSTRACT

Investigation of rat recombinant P2X(6) receptors has been limited because of the difficulty in obtaining functional expression in heterologous systems. In this study, we demonstrate glycosylation-dependent regulation of recombinant P2X(6) receptor function and associated conferral of a novel phenotype that is sensitive to the P2X(1) and P2X(3) receptor agonist, alphabeta-methylene ATP. In cells functionally expressing P2X(6) receptors, ATP and alphabeta-methylene ATP evoked slowly desensitizing inward currents (EC(50) values, 0.5 and 0.6 microM, respectively) with slow kinetics of current decay on agonist washout. 2',3'-O-(2,4,6-trinitrophenyl ATP) and iso-pyridoxalphosphate-6-azophenyl-2'-5'-disulfonate were effective antagonists (IC(50) values, 0.8 and 22 microM, respectively); however, suramin was relatively ineffective. Reverse transcription-polymerase chain reaction analysis confirmed the absence of other P2X receptor subunits. Western analysis of membrane fractions from functional and nonfunctional clones confirmed the presence of P2X(6) at the cell membrane but revealed a difference in apparent molecular mass of immunoreactive products ( approximately 70 and approximately 60 kDa, respectively). N-glycosidase F treatment of both functional and nonfunctional receptor cell membranes increased the electrophoretic mobilities of immunoreactive products, with both proteins migrating at approximately 55 kDa, demonstrating an increased level of glycosylation of the P2X(6) receptor in functional compared with nonfunctional cells. This study demonstrates that nonfunctional rat recombinant P2X(6) receptors 1) are expressed on the membrane surface of human embryonic kidney cells and 2) are glycosylated. Expression of the novel functional receptor phenotype is associated with further glycosylation, resulting in an apparently larger molecular mass. These results suggest that P2X(6) receptor subunits contribute to alphabeta-methylene ATP sensitivity.


Subject(s)
Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Glycosylation , Receptors, Purinergic P2/metabolism , Animals , Cell Membrane/metabolism , Cells, Cultured , Humans , Phenotype , Protein Subunits/metabolism , Rats , Receptors, Purinergic P2/drug effects
18.
J Clin Invest ; 112(12): 1895-905, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14679185

ABSTRACT

This study tests the hypothesis that P2X1 receptors mediate pressure-induced afferent arteriolar autoregulatory responses. Afferent arterioles from rats and P2X1 KO mice were examined using the juxtamedullary nephron technique. Arteriolar diameter was measured in response to step increases in renal perfusion pressure (RPP). Autoregulatory adjustments in diameter were measured before and during P2X receptor blockade with NF279 or A1 receptor blockade with 1,3-dipropyl-8-cyclopentylxanthine (DPCPX). Acute papillectomy or furosemide perfusion was performed to interrupt distal tubular fluid flow past the macula densa, thus minimizing tubuloglomerular feedback-dependent influences on afferent arteriolar function. Under control conditions, arteriolar diameter decreased by 17% and 29% at RPP of 130 and 160 mmHg, respectively. Blockade of P2X1 receptors with NF279 blocked pressure-mediated vasoconstriction, reflecting an attenuated autoregulatory response. The A1 receptor blocker DPCPX did not alter autoregulatory behavior or the response to ATP. Deletion of P2X1 receptors in KO mice significantly blunted autoregulatory responses induced by an increase in RPP, and this response was not further impaired by papillectomy or furosemide. WT control mice exhibited typical RPP-dependent vasoconstriction that was significantly attenuated by papillectomy. These data provide compelling new evidence indicating that tubuloglomerular feedback signals are coupled to autoregulatory preglomerular vasoconstriction through ATP-mediated activation of P2X1 receptors.


Subject(s)
Kidney/blood supply , Microcirculation , Receptors, Purinergic P2/physiology , Renal Circulation , Suramin/analogs & derivatives , Adenosine Triphosphate/metabolism , Animals , Diuretics/pharmacology , Dose-Response Relationship, Drug , Furosemide/pharmacology , Gene Deletion , Male , Mice , Mice, Knockout , Perfusion , Pressure , Rats , Rats, Sprague-Dawley , Receptors, Purinergic P2X , Suramin/pharmacology , Time Factors , Xanthines/pharmacology
19.
Blood ; 102(10): 3646-51, 2003 Nov 15.
Article in English | MEDLINE | ID: mdl-12907444

ABSTRACT

Purine nucleotides acting through P2 receptors play key roles in platelet signaling. The P2X1 receptor is an adenosine triphosphate (ATP)-gated ion channel that mediates a rapid calcium influx signal, but can also synergize with subsequent adenosine diphosphate (ADP)-evoked P2Y1 receptor-mediated responses and thus may contribute to platelet activation during hemostasis. Recent studies have shown that P2X1 receptors contribute to the formation of platelet thrombi, particularly under conditions of high shear stress. Based on intracellular Ca2+ measurements a previous report has suggested that a splice variant of the P2X1 receptor, P2X1del, is expressed in platelets and, in contrast to the full-length P2X1WT receptor, is activated by ADP. In the present study we show that the P2X1del receptor fails to form functional ion channels and is below the limit of detection in human platelets. Furthermore, ADP does not contribute to the rapid ionotropic P2X receptor-mediated response in platelets. These results support the notion that ATP is the principal physiologic agonist at P2X1 receptors and that it plays a role in the activation of platelets.


Subject(s)
Adenosine Diphosphate/pharmacology , Adenosine Triphosphate/physiology , Hemostasis , Membrane Proteins , Receptors, Purinergic P2/metabolism , Thrombosis/etiology , Adenosine Triphosphate/pharmacology , Alternative Splicing , Blood Platelets/metabolism , Calcium Signaling , Cell Line , Electrophysiology , Humans , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2/physiology , Receptors, Purinergic P2X , Receptors, Purinergic P2Y12 , Transfection
20.
J Exp Med ; 198(4): 661-7, 2003 Aug 18.
Article in English | MEDLINE | ID: mdl-12913094

ABSTRACT

The P2X1 receptor is a fast ATP-gated cation channel expressed in blood platelets, where its role has been difficult to assess due to its rapid desensitization and the lack of pharmacological tools. In this paper, we have used P2X1-/- and wild-type mouse platelets, treated with apyrase to prevent desensitization, to demonstrate the function of P2X1 in the response to thrombogenic stimuli. In vitro, the collagen-induced aggregation and secretion of P2X1-deficient platelets was decreased, as was adhesion and thrombus growth on a collagen-coated surface, particularly when the wall shear rate was elevated. In vivo, the functional role of P2X1 could be demonstrated using two models of platelet-dependent thrombotic occlusion of small arteries, in which blood flow is characterized by a high shear rate. The mortality of P2X1-/- mice in a model of systemic thromboembolism was reduced and the size of mural thrombi formed after a laser-induced vessel wall injury was decreased as compared with normal mice, whereas the time for complete thrombus removal was shortened. Overall, the P2X1 receptor appears to contribute to the formation of platelet thrombi, particularly in arteries in which shear forces are high.


Subject(s)
Adenosine Triphosphate/metabolism , Arteries/pathology , Receptors, Purinergic P2/metabolism , Thrombosis/metabolism , Animals , Apyrase/pharmacology , Arteries/cytology , Blood Platelets/drug effects , Blood Platelets/metabolism , Calcium/metabolism , Collagen/metabolism , Lung/anatomy & histology , Lung/metabolism , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Platelet Aggregation/physiology , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2X , Shear Strength , Thrombosis/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...