Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
1.
J Thromb Haemost ; 18(1): 44-56, 2020 01.
Article in English | MEDLINE | ID: mdl-31448510

ABSTRACT

BACKGROUND: Intestinal inflammation is associated with bleeding and thrombosis, two processes that may involve both platelets and neutrophils. However, the mechanisms and the respective contribution of these cells to intestinal bleeding and extra-intestinal thrombosis remain largely unknown. OBJECTIVE: Our study aimed at investigating the mechanisms underlying the maintenance of vascular integrity and thrombosis in intestinal inflammation. METHODS: We used a mouse model of acute colitis induced by oral administration of dextran sodium sulfate (DSS) for 7 days. Bleeding was assessed after depletion of platelets, neutrophils, or glycoprotein VI (GPVI); treatment with aspirin or clopidogrel; or in P2X1-deficient mice. Extra-intestinal thrombosis was analyzed using a laser-induced injury model of thrombosis in cremaster muscle arterioles. RESULTS: Platelet depletion or P2X1 deficiency led to macrocytic regenerative anemia due to intestinal hemorrhage. In contrast, GPVI, P2Y12, and thromboxane A2 were dispensable. Platelet P-selectin expression and regulated on activation, normal T-cell expressed and secreted (RANTES) plasma levels were lower in DSS-treated P2X1-deficient mice as compared to wild-type mice, indicative of a platelet secretion defect. Circulating neutrophils had a more activated phenotype, and neutrophil infiltration in the colon was increased. P2X1-deficient mice also had elevated plasma granulocyte-colony stimulating factor (G-CSF) levels. Neutrophil depletion limited blood loss in these mice, whereas exogenous administration of G-CSF in colitic wild-type mice caused macrocytic anemia. Anemic colitic P2X1-deficient mice formed atypical neutrophil- and fibrin-rich, and platelet-poor thrombi upon arteriolar endothelial injury. CONCLUSIONS: Platelets and P2X1 ion channels are mandatory to preserve vascular integrity in inflamed intestine. Upon P2X1 deficiency, neutrophils contribute to bleeding and they may also be responsible for enhanced thrombosis.


Subject(s)
Hemorrhage , Intestines/physiopathology , Receptors, Purinergic P2X1 , Thrombosis , Animals , Blood Platelets , Hemorrhage/chemically induced , Mice
2.
Biochem Biophys Res Commun ; 523(1): 190-195, 2020 02 26.
Article in English | MEDLINE | ID: mdl-31843194

ABSTRACT

P2X receptors are trimeric ATP-gated ion channels. In response to ATP binding, conformational changes lead to opening of the channel and ion flow. Current flow can decline during continued ATP binding in a process called desensitisation. The rate and extent of desensitisation is affected by multiple factors, for instance the T18A mutation in P2X2 makes the ion channel fast desensitising. We have used this mutation to investigate whether the gate restricting ion flow is different in the desensitised and the closed state, by combining molecular modelling and cysteine modification using MTSET (2-(Trimethylammonium)ethyl methanethiosulfonate). Homology modelling of the P2X2 receptor and negative space imaging of the channel suggested a movement of the restriction gate with residue T335 being solvent accessible in the desensitised, but not the closed state. This was confirmed experimentally by probing the accessibility of T335C in the P2X2 T18A/T335C (fast desensitisation) and T335C (slow desensitisation) mutants with MTSET which demonstrates that the barrier to ion flow is different in the closed and the desensitised states. To investigate the T18A induced switch in desensitisation we compared molecular dynamics simulations of the wild type and T18A P2X2 receptor which suggest that the differences in time course of desensitisation are due to structural destabilization of a hydrogen bond network of conserved residues in the proximity of T18.


Subject(s)
Adenosine Triphosphate/metabolism , Receptors, Purinergic P2X2/chemistry , Receptors, Purinergic P2X2/metabolism , Humans , Models, Molecular , Mutation , Receptors, Purinergic P2X2/genetics
3.
Mol Pharmacol ; 96(3): 355-363, 2019 09.
Article in English | MEDLINE | ID: mdl-31263019

ABSTRACT

The P2X7 receptor is a trimeric ligand-gated ion channel activated by ATP. It is implicated in the cellular response to trauma/disease and considered to have significant therapeutic potential. Using chimeras and point mutants we have mapped the binding site of the P2X7R-selective antagonist AZ11645373 to the known allosteric binding pocket at the interface between two subunits, in proximity to, but separated from the ATP binding site. Our structural model of AZ11645373 binding is consistent with effects of mutations on antagonist sensitivity, and the proposed binding mode explains variation in antagonist sensitivity between the human and rat P2X7 receptors. We have also determined the site of action for the P2X7R-selective antagonists ZINC58368839, brilliant blue G, KN-62, and calmidazolium. The effect of intersubunit allosteric pocket "signature mutants" F88A, T90V, D92A, F103A, and V312A on antagonist sensitivity suggests that ZINC58368839 comprises a binding mode similar to AZ11645373 and other previously characterized antagonists. For the larger antagonists, brilliant blue G, KN-62, and calmidazolium, our data imply an overlapping but distinct binding mode involving the central upper vestibule of the receptor in addition to the intersubunit allosteric pocket. Our work explains the site of action for a series of P2X7R antagonists and establishes "signature mutants" for P2X7R binding-mode characterization.


Subject(s)
Point Mutation , Purinergic P2X Receptor Antagonists/pharmacology , Receptors, Purinergic P2X7/chemistry , Receptors, Purinergic P2X7/metabolism , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/chemistry , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Adenosine Triphosphate/metabolism , Allosteric Site , Amides/chemistry , Amides/pharmacology , Binding Sites , Humans , Imidazoles/chemistry , Imidazoles/pharmacology , Indoles/chemistry , Indoles/pharmacology , Models, Molecular , Molecular Docking Simulation , Purinergic P2X Receptor Antagonists/chemistry , Receptors, Purinergic P2X7/genetics , Rosaniline Dyes/chemistry , Rosaniline Dyes/pharmacology , Thiazoles/chemistry , Thiazoles/pharmacology
4.
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
5.
J Gen Physiol ; 151(2): 146-155, 2019 02 04.
Article in English | MEDLINE | ID: mdl-30626615

ABSTRACT

The human P2X1 receptor (hP2X1R) is a trimeric ligand-gated ion channel opened by extracellular ATP. The intracellular amino and carboxyl termini play significant roles in determining the time-course and regulation of channel gating-for example, the C terminus regulates recovery from the desensitized state following agonist washout. This suggests that the intracellular regions of the channel have distinct structural features. Studies on the hP2X3R have shown that the intracellular regions associate to form a cytoplasmic cap in the open state of the channel. However, intracellular features could not be resolved in the agonist-free apo and ATP-bound desensitized structures. Here we investigate the organization of the intracellular regions of hP2X1R in the apo and ATP-bound desensitized states following expression in HEK293 cells. We couple cysteine scanning mutagenesis of residues R25-G30 and H355-R360 with the use of bi-functional cysteine reactive cross-linking compounds of different lengths (MTS-2-MTS, BMB, and BM(PEG)2), which we use as molecular calipers. If two cysteine residues come into close proximity, we predict they will be cross-linked and result in ∼66% of the receptor subunits running on a Western blot as dimers. In the control construct (C349A) that removed the free cysteine C349, and some cysteine-containing mutants, cross-linker treatment does not result in dimerization. However, we detect efficient dimerization for R25C, G30C, P358C, K359C, and R360C. This selective pattern indicates that there is structural organization to these regions in the apo and desensitized states in a native membrane environment. The existence of such precap (apo) and postcap (desensitized) organization of the intracellular domains would facilitate efficient gating of the channel.


Subject(s)
Receptors, Purinergic P2X1/chemistry , Amino Acid Substitution , Cross-Linking Reagents/pharmacology , Cysteine/chemistry , Cysteine/genetics , HEK293 Cells , Humans , Molecular Dynamics Simulation , Protein Conformation/drug effects , Protein Multimerization , Purinergic P2X Receptor Agonists/pharmacology , Receptors, Purinergic P2X1/genetics , Receptors, Purinergic P2X1/metabolism
6.
Annu Rev Physiol ; 81: 43-62, 2019 02 10.
Article in English | MEDLINE | ID: mdl-30354932

ABSTRACT

In the nervous system, ATP is co-stored in vesicles with classical transmitters and released in a regulated manner. ATP from the intracellular compartment can also exit the cell through hemichannels and following shear stress or membrane damage. In the past 30 years, the action of ATP as an extracellular transmitter at cell-surface receptors has evolved from somewhat of a novelty that was treated with skepticism to purinergic transmission being accepted as having widespread important functional roles mediated by ATP-gated ionotropic P2X receptors (P2XRs). This review focuses on work published in the last five years and provides an overview of ( a) structural studies, ( b) the molecular basis of channel properties and regulation of P2XRs, and ( c) the physiological and pathophysiological roles of ATP acting at defined P2XR subtypes.


Subject(s)
Adenosine Triphosphate/metabolism , Ion Channels/metabolism , Receptors, Purinergic P2X/metabolism , Animals , Humans
7.
J Biol Chem ; 293(33): 12820-12831, 2018 08 17.
Article in English | MEDLINE | ID: mdl-29997254

ABSTRACT

ATP is the native agonist for cell-surface ligand-gated P2X receptor (P2XR) cation channels. The seven mammalian subunits (P2X1-7) form homo- and heterotrimeric P2XRs having significant physiological and pathophysiological roles. Pyridoxalphosphate-6-azophenyl-2',4'-disulfonic acid (PPADS) is an effective antagonist at most mammalian P2XRs. Lys-249 in the extracellular domain of P2XR has previously been shown to contribute to PPADS action. To map this antagonist site, we generated human P2X1R cysteine substitutions within a circle centered at Lys-249 (with a radius of 13 Å equal to the length of PPADS). We hypothesized that cysteine substitutions of residues involved in PPADS binding would (i) reduce cysteine accessibility (measured by MTSEA-biotinylation), (ii) exhibit altered PPADS affinity, and (iii) quench the fluorescence of cysteine residues modified with MTS-TAMRA. Of the 26 residues tested, these criteria were met by only four (Lys-70, Asp-170, Lys-190, and Lys-249), defining the antagonist site, validating molecular docking results, and thereby providing the first experimentally supported model of PPADS binding. This binding site overlapped with the ATP-binding site, indicating that PPADS sterically blocks agonist access. Moreover, PPADS induced a conformational change at the cysteine-rich head (CRH) region adjacent to the orthosteric ATP-binding pocket. The importance of this movement was confirmed by demonstrating that substitution introducing positive charge present in the CRH of the hP2X1R causes PPADS sensitivity at the normally insensitive rat P2X4R. This study provides a template for developing P2XR subtype selectivity based on the differences among the mammalian subunits around the orthosteric P2XR-binding site and the CRH.


Subject(s)
Models, Molecular , Purinergic P2X Receptor Antagonists/chemistry , Pyridoxal Phosphate/analogs & derivatives , Receptors, Purinergic P2X1/chemistry , Animals , Binding Sites , Humans , Pyridoxal Phosphate/chemistry , Rats , Receptors, Purinergic P2X1/genetics , Receptors, Purinergic P2X4/chemistry , Receptors, Purinergic P2X4/genetics , Xenopus laevis
8.
Mol Pharmacol ; 93(5): 553-562, 2018 05.
Article in English | MEDLINE | ID: mdl-29535152

ABSTRACT

P2X7 receptor (P2X7R) activation requires ∼100-fold higher concentrations of ATP than other P2X receptor (P2XR) subtypes. Such high levels are found during cellular stress, and P2X7Rs consequently contribute to a range of pathophysiological conditions. We have used chimeric and mutant P2X7Rs, coupled with molecular modeling, to produce a validated model of the binding mode of the subtype-selective antagonist A438079 at an intersubunit allosteric site. Within the allosteric site large effects on antagonist action were found for point mutants of residues F88A, D92A, F95A, and F103A that were conserved or similar between sensitive/insensitive P2XR subtypes, suggesting that these side-chain interactions were not solely responsible for high-affinity antagonist binding. Antagonist sensitivity was increased with mutations that remove the bulk of side chains around the center of the binding pocket, suggesting that the dimensions of the pocket make a significant contribution to selectivity. Chimeric receptors swapping the left flipper (around the orthosteric site) reduced both ATP and antagonist sensitivity. Point mutations within this region highlighted the contribution of a P2X7R-specific aspartic acid residue (D280) that modeling suggests forms a salt bridge with the lower body region of the receptor. The D280A mutant removing this charge increased ATP potency 15-fold providing a new insight into the low ATP sensitivity of the P2X7R. The ortho- and allosteric binding sites form either side of the ß-strand Y291-E301 adjacent to the left flipper. This structural linking may explain the contribution of the left flipper to both agonist and antagonist action.


Subject(s)
Acetamides/pharmacology , Purinergic P2X Receptor Antagonists/pharmacology , Pyridines/pharmacology , Quinolines/pharmacology , Receptors, Purinergic P2X7/drug effects , Tetrazoles/pharmacology , Allosteric Regulation , Allosteric Site , Amino Acid Sequence , Binding Sites , Humans , Ligands , Molecular Docking Simulation , Point Mutation , Receptors, Purinergic P2X7/chemistry , Receptors, Purinergic P2X7/genetics , Sequence Homology, Amino Acid
9.
Article in English | MEDLINE | ID: mdl-28428949

ABSTRACT

α-haemolysin (HlyA)-producing Escherichia coli commonly inflict severe urinary tract infections, including pyelonephritis, which comprises substantial risk for sepsis. In vitro, the cytolytic effect of HlyA is mainly mediated by ATP release through the HlyA pore and subsequent P2X1/P2X7 receptor activation. This amplification of the lytic process is not unique to HlyA but is observed by many other pore-forming proteins including complement-induced haemolysis. Since free hemoglobin in the blood is known to be associated with a worse outcome in sepsis one could speculate that inhibition of P2X receptors would ameliorate the course of sepsis. Surprisingly, this study demonstrates that [Formula: see text] and [Formula: see text] mice are exceedingly sensitive to sepsis with uropathogenic E. coli. These mice have markedly lower survival, higher cytokine levels and activated intravascular coagulation. Quite the reverse is seen in [Formula: see text] mice, which had markedly lower cytokine levels and less coagulation activation compared to controls after exposure to uropathogenic E. coli. The high cytokine levels in the [Formula: see text] mouse are unexpected, since P2X7 is implicated in caspase-1-dependent IL-1ß production. Here, we demonstrate that IL-1ß production during sepsis with uropathogenic E. coli is mediated by caspase-8, since caspase-8 and RIPK3 double knock out mice show substantially lower cytokine during sepsis and increased survival after injection of TNFα. These data support that P2X7 and P2X4 receptor activation has a protective effect during severe E. coli infection.


Subject(s)
Disease Susceptibility , Escherichia coli Infections/pathology , Receptors, Purinergic P2X1/deficiency , Receptors, Purinergic P2X4/deficiency , Receptors, Purinergic P2X7/deficiency , Sepsis/pathology , Animals , Disease Models, Animal , Escherichia coli Infections/genetics , Mice , Mice, Knockout , Survival Analysis , Treatment Outcome
10.
Sci Rep ; 7(1): 725, 2017 04 07.
Article in English | MEDLINE | ID: mdl-28389651

ABSTRACT

The P2X7 receptor (P2X7R) for ATP is a therapeutic target for pathophysiological states including inflammation, pain management and epilepsy. This is facilitated by the predicted low side effect profile as the high concentrations of ATP required to activate the receptor are usually only found following cell damage/disease and so P2X7Rs respond to a "danger" signal and are not normally active. AZ10606120 is a selective antagonist for P2X7Rs (IC50 of ~10 nM) and ineffective at the P2X1R (at 10 µM). To determine the molecular basis of selectivity we generated a series of P2X7/1R chimeras and mutants. Two regions that are unique to the P2X7R, a loop insertion (residues 73-79) and threonine residues T90 and T94, are required for high affinity antagonist action. Point mutations ruled out an orthosteric antagonist site. Mutations and molecular modelling identified an allosteric binding site that forms at the subunit interface at the apex of the receptor. Molecular dynamics simulations indicated that unique P2X7R features regulate access of AZ10606120 to the allosteric site. The characterisation of the allosteric pocket provides a new and novel target for rational P2X7R drug development.


Subject(s)
Adamantane/analogs & derivatives , Adenosine Triphosphate/metabolism , Allosteric Site , Aminoquinolines/chemistry , Binding Sites , Ion Channel Gating , Purinergic P2X Receptor Antagonists/chemistry , Receptors, Purinergic P2X7/chemistry , Receptors, Purinergic P2X7/metabolism , Adamantane/chemistry , Adamantane/pharmacology , Alleles , Allosteric Regulation , Amino Acid Sequence , Amino Acid Substitution , Aminoquinolines/pharmacology , Humans , Lignans , Models, Molecular , Molecular Conformation , Mutation , Protein Binding , Protein Interaction Domains and Motifs , Purinergic P2X Receptor Antagonists/pharmacology , Receptors, Purinergic P2X7/genetics , Structure-Activity Relationship
11.
Sci Rep ; 6: 32918, 2016 09 12.
Article in English | MEDLINE | ID: mdl-27616669

ABSTRACT

Structural studies of P2X receptors show a novel U shaped ATP orientation following binding. We used voltage clamp fluorometry (VCF) and molecular dynamics (MD) simulations to investigate agonist action. For VCF the P2X1 receptor (P2X1R) K190C mutant (adjacent to the agonist binding pocket) was labelled with the fluorophore MTS-TAMRA and changes in fluorescence on agonist treatment provided a real time measure of conformational changes. Studies with heteromeric channels incorporating a key lysine mutation (K68A) in the ATP binding site demonstrate that normally three molecules of ATP activate the receptor. The time-course of VCF responses to ATP, 2'-deoxy ATP, 3'-deoxy ATP, Ap5A and αßmeATP were agonist dependent. Comparing the properties of the deoxy forms of ATP demonstrated the importance of the 2' hydroxyl group on the ribose ring in determining agonist efficacy consistent with MD simulations showing that it forms a hydrogen bond with the γ-phosphate oxygen stabilizing the U-shaped conformation. Comparison of the recovery of fluorescence on agonist washout, with channel activation to a second agonist application for the partial agonists Ap5A and αßmeATP, showed a complex relationship between conformational change and desensitization. These results highlight that different agonists induce distinct conformational changes, kinetics and recovery from desensitization at P2X1Rs.


Subject(s)
Adenosine Triphosphate/agonists , Adenosine Triphosphate/pharmacology , Receptors, Purinergic P2X1/chemistry , Receptors, Purinergic P2X1/metabolism , Xenopus laevis/genetics , Adenosine Triphosphate/chemistry , Animals , Binding Sites , Dinucleoside Phosphates/pharmacology , Humans , Kinetics , Ligands , Models, Molecular , Molecular Dynamics Simulation , Mutation , Patch-Clamp Techniques , Protein Binding , Protein Conformation , Receptors, Purinergic P2X1/genetics
12.
Adv Exp Med Biol ; 898: 305-29, 2016.
Article in English | MEDLINE | ID: mdl-27161234

ABSTRACT

Ligand-gated ion channels on the cell surface are directly activated by the binding of an agonist to their extracellular domain and often referred to as ionotropic receptors. P2X receptors are ligand-gated non-selective cation channels with significant permeability to Ca(2+) whose principal physiological agonist is ATP. This chapter focuses on the mechanisms by which P2X1 receptors, a ubiquitously expressed member of the family of ATP-gated channels, can contribute to cellular responses in non-excitable cells. Much of the detailed information on the contribution of P2X1 to Ca(2+) signalling and downstream functional events has been derived from the platelet. The underlying primary P2X1-generated signalling event in non-excitable cells is principally due to Ca(2+) influx, although Na(+) entry will also occur along with membrane depolarization. P2X1 receptor stimulation can lead to additional Ca(2+) mobilization via a range of routes such as amplification of G-protein-coupled receptor-dependent Ca(2+) responses. This chapter also considers the mechanism by which cells generate extracellular ATP for autocrine or paracrine activation of P2X1 receptors. For example cytosolic ATP efflux can result from opening of pannexin anion-permeable channels or following damage to the cell membrane. Alternatively, ATP stored in specialised secretory vesicles can undergo quantal release via the process of exocytosis. Examples of physiological or pathophysiological roles of P2X1-dependent signalling in non-excitable cells are also discussed, such as thrombosis and immune responses.


Subject(s)
Blood Platelets/metabolism , Calcium Signaling , Ion Channel Gating , Ion Channels/metabolism , Receptors, Purinergic P2X1/metabolism , Animals , Cytosol/metabolism , Humans , Ion Channels/chemistry , Ligands , Protein Conformation , Receptors, Purinergic P2X1/chemistry
13.
J Biol Chem ; 290(23): 14556-66, 2015 Jun 05.
Article in English | MEDLINE | ID: mdl-25903136

ABSTRACT

P2X7 receptors are ATP-gated ion channels that contribute to inflammation and cell death. They have the novel property of showing marked facilitation to repeated applications of agonist, and the intrinsic channel pore dilates to allow the passage of fluorescent dyes. A 60-s application of ATP to hP2X7 receptors expressed in Xenopus oocytes gave rise to a current that had a biphasic time course with initial and secondary slowly developing components. A second application of ATP evoked a response with a more rapid time to peak. This facilitation was reversed to initial levels following a 10-min agonist-free interval. A chimeric approach showed that replacement of the pre-TM1 amino-terminal region with the corresponding P2X2 receptor section (P2X7-2Nß) gave responses that quickly reached a steady state and did not show facilitation. Subsequent point mutations of variant residues identified Asn-16 and Ser-23 as important contributors to the time course/facilitation. The P2X7 receptor is unique in having an intracellular carboxyl-terminal cysteine-rich region (Ccys). Deletion of this region removed the secondary slowly developing current, and, when expressed in HEK293 cells, ethidium bromide uptake was only ∼5% that of WT levels, indicating reduced large pore formation. Dye uptake was also reduced for the P2X7-2Nß chimera. Surprisingly, combination of the chimera and the Ccys deletion (P2X7-2NßdelCcys) restored the current rise time and ethidium uptake to WT levels. These findings suggest that there is a coevolved interaction between the juxtatransmembrane amino and carboxyl termini in the regulation of P2X7 receptor gating.


Subject(s)
Adenosine Triphosphate/metabolism , Receptors, Purinergic P2X7/metabolism , Animals , HEK293 Cells , Humans , Mutagenesis , Receptors, Purinergic P2X7/chemistry , Receptors, Purinergic P2X7/genetics , Xenopus laevis
14.
Neurourol Urodyn ; 34(3): 292-8, 2015 Mar.
Article in English | MEDLINE | ID: mdl-24249481

ABSTRACT

AIMS: An age-related increase in prostatic smooth muscle tone is partly responsible for the lower urinary tract symptoms associated with benign prostatic hyperplasia (BPH). Changes in the effectors of prostatic smooth muscle contraction with age may play a role in the development of these symptoms. Using a mouse model of prostate contractility, this study investigated the effect of age on the different components of contractility in the prostate gland. METHODS: The isometric force developed in response to electrical field stimulation or exogenously applied agonists by mouse prostates mounted in organ baths, was evaluated to determine the effect of age on contractile mechanisms. Changes with age in the rate of ATP breakdown and levels of the P2rx1 gene and P2X1-purinoceptor expression in mouse prostate were measured by a modified luciferin-luciferase assay, RT-PCR and western blot, respectively. RESULTS: Nerve mediated contractile responses containing a component elicited by P2X1-purinoceptors were observed in prostates taken from aged mice, but not in prostates taken from young adult mice. Furthermore, the potency of the endogenous purinoceptor agonist ATP was 50-fold greater in aged mice, whereas the potency of its stable analogue α,ß-metATP was unchanged. An age-related decrease in ATP metabolism was also observed. CONCLUSIONS: With age, a purinergic contractile response to nerve stimulation develops in the mouse prostate gland due to a decrease in the rate of ATP breakdown. This may contribute to the increase in muscular tone observed in BPH and suggests that P2X1-purinoceptors are an additional target for the treatment of BPH.


Subject(s)
Adenosine Triphosphate/metabolism , Muscle Contraction , Prostate/physiology , Receptors, Purinergic P2X1/physiology , Age Factors , Animals , Male , Mice , Prostate/metabolism , Time Factors
15.
J Biol Chem ; 290(3): 1559-69, 2015 Jan 16.
Article in English | MEDLINE | ID: mdl-25425641

ABSTRACT

P2X receptor subtype-selective antagonists are promising candidates for treatment of a range of pathophysiological conditions. However, in contrast to high resolution structural understanding of agonist action in the receptors, comparatively little is known about the molecular basis of antagonist binding. We have generated chimeras and point mutations in the extracellular ligand-binding loop of the human P2X1 receptor, which is inhibited by NF449, suramin, and pyridoxal-phosphate-6-azophenyl-2,4-disulfonate, with residues from the rat P2X4 receptor, which is insensitive to these antagonists. There was little or no effect on sensitivity to suramin and pyridoxal-phosphate-6-azophenyl-2,4-disulfonate in chimeric P2X1/4 receptors, indicating that a significant number of residues required for binding of these antagonists are present in the P2X4 receptor. Sensitivity to the P2X1 receptor-selective antagonist NF449 was reduced by ∼60- and ∼135-fold in chimeras replacing the cysteine-rich head, and the dorsal fin region below it in the adjacent subunit, respectively. Point mutants identified the importance of four positively charged residues at the base of the cysteine-rich head and two variant residues in the dorsal fin for high affinity NF449 binding. These six residues were used as the starting area for molecular docking. The four best potential NF449-binding poses were then discriminated by correspondence with the mutagenesis data and an additional mutant to validate the binding of one lobe of NF449 within the core conserved ATP-binding pocket and the other lobes coordinated by positive charge on the cysteine-rich head region and residues in the adjacent dorsal fin.


Subject(s)
Adenosine Triphosphate/chemistry , Benzenesulfonates/chemistry , Point Mutation , Purinergic P2X Receptor Antagonists/chemistry , Animals , Binding Sites , Cysteine/chemistry , Drug Design , Humans , Ligands , Lysine/chemistry , Mice , Models, Molecular , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Oocytes/metabolism , Protein Binding , Rats , Suramin/chemistry , Xenopus laevis
16.
J Immunol ; 194(2): 739-49, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25480563

ABSTRACT

Extracellular ATP is becoming increasingly recognized as an important regulator of inflammation. However, the known repertoire of P2 receptor subtypes responsible for the proinflammatory effects of ATP is sparse. We looked at whether the P2X1 receptor, an ATP-gated cation channel present on platelets, neutrophils, and macrophages, participates in the acute systemic inflammation provoked by LPS. Compared with wild-type (WT) mice, P2X1(-/-) mice displayed strongly diminished pathological responses, with dampened neutrophil accumulation in the lungs, less tissue damage, reduced activation of coagulation, and resistance to LPS-induced death. P2X1 receptor deficiency also was associated with a marked reduction in plasma levels of the main proinflammatory cytokines and chemokines induced by LPS. Interestingly, macrophages and neutrophils isolated from WT and P2X1(-/-) mice produced similar levels of proinflammatory cytokines when stimulated with LPS in vitro. Intravital microscopy revealed a defect in LPS-induced neutrophil emigration from cremaster venules into the tissues of P2X1(-/-) mice. Using adoptive transfer of immunofluorescently labeled neutrophils from WT and P2X1(-/-) mice into WT mice, we demonstrate that the absence of the P2X1 receptor on neutrophils was responsible for this defect. This study reveals a major role for the P2X1 receptor in LPS-induced lethal endotoxemia through its critical involvement in neutrophil emigration from venules.


Subject(s)
Endotoxemia/immunology , Lipopolysaccharides/toxicity , Lung/immunology , Neutrophil Infiltration/immunology , Neutrophils/immunology , Receptors, Purinergic P2X1/immunology , Animals , Blood Coagulation/drug effects , Blood Coagulation/genetics , Blood Coagulation/immunology , Endotoxemia/chemically induced , Endotoxemia/genetics , Endotoxemia/pathology , Lung/pathology , Macrophages/immunology , Macrophages/pathology , Mice , Mice, Knockout , Neutrophil Infiltration/drug effects , Neutrophil Infiltration/genetics , Neutrophils/pathology , Receptors, Purinergic P2X1/genetics
17.
Mol Pharmacol ; 86(6): 707-15, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25296688

ABSTRACT

ATP acts as an extracellular signaling molecule at cell-surface P2X receptors, mediating a variety of important physiologic and pathophysiologic roles. Homomeric P2X1 receptors open on binding ATP and then transition to an ATP-bound closed, desensitized state that requires an agonist-free washout period to recover. Voltage-clamp fluorometry was used to record ion channel activity and conformational changes simultaneously at defined positions in the extracellular loop of the human P2X1 receptor during not only agonist binding and desensitization but also during recovery. ATP evoked distinct conformational changes adjacent to the agonist binding pocket in response to channel activation and desensitization. The speed of recovery of the conformational change on agonist washout was state-dependent, with a faster time constant from the open (5 seconds) compared with the desensitized (75 seconds) form of the channel. The ability of ATP to evoke channel activity on washout after desensitization was not dependent on the degree of conformational rearrangement in the extracellular loop, and desensitization was faster from the partially recovered state. An intracellular mutation in the carboxyl terminus that slowed recovery of P2X1 receptor currents (7-fold less recovery at 30 seconds) had no effect on the time course of the extracellular conformational rearrangements. This study highlights that the intracellular portion of the receptor can regulate recovery and shows for the first time that this is by a mechanism independent of changes in the extracellular domain, suggesting the existence of a distinct desensitization gate in this novel class of ligand gated ion channels.


Subject(s)
Adenosine Triphosphate/pharmacology , Receptors, Purinergic P2X1/chemistry , Fluorometry , Kinetics , Patch-Clamp Techniques , Protein Conformation , Receptors, Purinergic P2X1/physiology
18.
Blood ; 124(16): 2575-85, 2014 Oct 16.
Article in English | MEDLINE | ID: mdl-25150292

ABSTRACT

Adenosine triphosphate (ATP) and its metabolite, adenosine, are key regulators of polymorphonuclear neutrophil (PMN) functions. PMNs have recently been implicated in the initiation of thrombosis. We investigated the role of ATP and adenosine in PMN activation and recruitment at the site of endothelial injury. Following binding to the injured vessel wall, PMNs are activated and release elastase. The recruitment of PMNs and the subsequent fibrin generation and thrombus formation are strongly affected in mice deficient in the P2X1-ATP receptor and in wild-type (WT) mice treated with CGS 21680, an agonist of the A2A adenosine receptor or NF449, a P2X1 antagonist. Infusion of WT PMNs into P2X1-deficient mice increases fibrin generation but not thrombus formation. Restoration of thrombosis requires infusion of both platelets and PMNs from WT mice. In vitro, ATP activates PMNs, whereas CGS 21680 prevents their binding to activated endothelial cells. These data indicate that adenosine triphosphate (ATP) contributes to polymorphonuclear neutrophil (PMN) activation leading to their adhesion at the site of laser-induced endothelial injury, a necessary step leading to the generation of fibrin, and subsequent platelet-dependent thrombus formation. Altogether, our study identifies previously unknown mechanisms by which ATP and adenosine are key molecules involved in thrombosis by regulating the activation state of PMNs.


Subject(s)
Blood Platelets/metabolism , Neutrophils/metabolism , Receptors, Purinergic P2X1/genetics , Thrombosis/genetics , Animals , Blood Platelets/pathology , Fibrin/metabolism , Gene Deletion , Gene Expression , Humans , Mice , Mice, Inbred C57BL , Neutrophils/pathology , Purinergic P2X Receptor Antagonists/pharmacology , Receptors, Purinergic P2X1/metabolism , Thrombosis/metabolism , Thrombosis/pathology
19.
Mol Pharmacol ; 86(3): 243-51, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24923466

ABSTRACT

Many cells express both P2X cation channels and P2Y G-protein-coupled receptors that are costimulated by nucleotides released during physiologic or pathophysiologic responses. For example, during hemostasis and thrombosis, ATP-gated P2X1 channels and ADP-stimulated P2Y1 and P2Y12 G-protein coupled receptors play important roles in platelet activation. It has previously been reported that P2X1 receptors amplify P2Y1-evoked Ca(2+) responses in platelets, but the underlying mechanism and influence on function is unknown. In human platelets, we show that maximally activated P2X1 receptors failed to stimulate significant aggregation but could amplify the aggregation response to a submaximal concentration of ADP. Costimulation of P2X1 and P2Y1 receptors generated a superadditive Ca(2+) increase in both human platelets and human embryonic kidney 293 (HEK293) cells via a mechanism dependent on Ca(2+) influx rather than Na(+) influx or membrane depolarization. The potentiation, due to an enhanced P2Y1 response, was observed if ADP was added up to 60 seconds after P2X1 activation. P2X1 receptors also enhanced Ca(2+) responses when costimulated with type 1 protease activated and M1 muscarinic acetylcholine receptors. The P2X1-dependent amplification of Gq-coupled [Ca(2+)]i increase was mimicked by ionomycin and was not affected by inhibition of protein kinase C, Rho-kinase, or extracellular signal-regulated protein kinase 1/2, which suggests that it results from potentiation of inositol 1,4,5-trisphosphate receptors and/or phospholipase C. We conclude that Ca(2+) influx through P2X1 receptors amplifies Ca(2+) signaling through P2Y1 and other Gq-coupled receptors. This represents a general form of co-incidence detection of ATP and coreleased agonists, such as ADP at sites of vascular injury or synaptic transmitters acting at metabotropic Gq-coupled receptors.


Subject(s)
Adenosine Diphosphate/metabolism , Blood Platelets/metabolism , Calcium/metabolism , Platelet Aggregation , Receptors, Purinergic P2X1/metabolism , Receptors, Purinergic P2Y1/metabolism , Calcium Signaling , HEK293 Cells , Humans , In Vitro Techniques , Recombinant Proteins/metabolism
20.
Proc Natl Acad Sci U S A ; 110(51): 20825-30, 2013 Dec 17.
Article in English | MEDLINE | ID: mdl-24297884

ABSTRACT

Therapeutic targets for male contraception are associated with numerous problems due to their focus on disrupting spermatogenesis or hormonal mechanisms to produce dysfunctional sperm. Here we describe the dual genetic deletion of α1A-adrenergic G protein-coupled receptors (adrenoceptors) and P2X1-purinoceptor ligand gated ion channels in male mice, thereby blocking sympathetically mediated sperm transport through the vas deferens during the emission phase of ejaculation. This modification produced 100% infertility without effects on sexual behavior or function. Sperm taken from the cauda epididymides of double knockout mice were microscopically normal and motile. Furthermore, double knockout sperm were capable of producing normal offspring following intracytoplasmic sperm injection into wild-type ova and implantation of the fertilized eggs into foster mothers. Blood pressure and baroreflex function was reduced in double knockout mice, but no more than single knockout of α1A-adrenoceptors alone. These results suggest that this autonomic method of male contraception appears free of major physiological and behavioral side effects. In addition, they provide conclusive proof of concept that pharmacological antagonism of the P2X1-purinoceptor and α1A-adrenoceptor provides a safe and effective therapeutic target for a nonhormonal, readily reversible male contraceptive.


Subject(s)
Contraception , Infertility, Male/genetics , Receptors, Adrenergic, alpha-1 , Receptors, Purinergic P2X1 , Spermatozoa/metabolism , Adrenergic alpha-1 Receptor Antagonists/pharmacology , Animals , Baroreflex , Gene Knockdown Techniques , Male , Mice , Mice, Knockout , Purinergic P2X Receptor Antagonists/pharmacology , Sperm Motility/physiology , Spermatozoa/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...