Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
Platelets ; 33(3): 451-461, 2022 Apr 03.
Article in English | MEDLINE | ID: mdl-34348571

ABSTRACT

Kv1.3 is a voltage-gated K+-selective channel with roles in immunity, insulin-sensitivity, neuronal excitability and olfaction. Despite being one of the largest ionic conductances of the platelet surface membrane, its contribution to platelet function is poorly understood. Here we show that Kv1.3-deficient platelets display enhanced ADP-evoked platelet aggregation and secretion, and an increased surface expression of platelet integrin αIIb. In contrast, platelet adhesion and thrombus formation in vitro under arterial shear conditions on surfaces coated with collagen were reduced for samples from Kv1.3-/- compared to wild type mice. Use of collagen-mimetic peptides revealed a specific defect in the engagement with α2ß1. Kv1.3-/- platelets developed significantly fewer, and shorter, filopodia than wild type platelets during adhesion to collagen fibrils. Kv1.3-/- mice displayed no significant difference in thrombus formation within cremaster muscle arterioles using a laser-induced injury model, thus other pro-thrombotic pathways compensate in vivo for the adhesion defect observed in vitro. This may include the increased platelet counts of Kv1.3-/- mice, due in part to a prolonged lifespan. The ability of Kv1.3 to modulate integrin-dependent platelet adhesion has important implications for understanding its contribution to normal physiological platelet function in addition to its reported roles in auto-immune diseases and thromboinflammatory models of stroke.


Subject(s)
Blood Platelets/metabolism , Collagen/metabolism , Integrin alpha2beta1/metabolism , Platelet Adhesiveness/physiology , Platelet Aggregation/physiology , Potassium Channels, Voltage-Gated/metabolism , Humans
2.
Platelets ; 32(7): 872-879, 2021 Oct 03.
Article in English | MEDLINE | ID: mdl-33872124

ABSTRACT

Potassium ions have widespread roles in cellular homeostasis and activation as a consequence of their large outward concentration gradient across the surface membrane and ability to rapidly move through K+-selective ion channels. In platelets, the predominant K+ channels include the voltage-gated K+ channel Kv1.3, and the intermediate conductance Ca2+-activated K+ channel KCa3.1, also known as the Gardos channel. Inwardly rectifying potassium GIRK channels and KCa1.1 large conductance Ca2+-activated K+ channels have also been reported in the platelet, although they remain to be demonstrated using electrophysiological techniques. Whole-cell patch clamp and fluorescent indicator measurements in the platelet or their precursor cell reveal that Kv1.3 sets the resting membrane potential and KCa3.1 can further hyperpolarize the cell during activation, thereby controlling Ca2+ influx. Kv1.3-/- mice exhibit an increased platelet count, which may result from an increased splenic megakaryocyte development and longer platelet lifespan. This review discusses the evidence in the literature that Kv1.3, KCa3.1. GIRK and KCa1.1 channels contribute to a number of platelet functional responses, particularly collagen-evoked adhesion, procoagulant activity and GPCR function. Putative roles for other K+ channels and known accessory proteins which to date have only been detected in transcriptomic or proteomic studies, are also discussed.


Subject(s)
Blood Platelets/metabolism , Potassium Channels/metabolism , Animals , Humans , Mice
3.
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
4.
Platelets ; 30(8): 962-966, 2019.
Article in English | MEDLINE | ID: mdl-31008669

ABSTRACT

TMEM16F is a surface membrane protein critical for platelet procoagulant activity, which exhibits both phospholipid scramblase and ion channel activities following sustained elevation of cytosolic Ca2+. The extent to which the ionic permeability of TMEM16F is important for platelet scramblase responses remains controversial. To date, only one study has reported the electrophysiological properties of TMEM16F in cells of platelet/megakaryocyte lineage, which observed cation-selectivity within excised patch recordings from murine marrow-derived megakaryocytes. This contrasts with reports using whole-cell recordings that describe this channel as displaying either selectivity for anions or being relatively non-selective amongst the major physiological monovalent ions. We have studied TMEM16F expression and channel activity in primary rat and mouse megakaryocytes and the human erythroleukemic (HEL) cell line that exhibits megakaryocytic surface markers. Immunocytochemical analysis was consistent with surface TMEM16F expression in cells from all three species. Whole-cell recordings in the absence of K+-selective currents revealed an outwardly rectifying conductance activated by a high intracellular Ca2+ concentration in all three species. These currents appeared after 5-6 minutes and were blocked by CaCCinh-A01, properties typical of TMEM16F. Ion substitution experiments showed that the underlying conductance was predominantly Cl--permeable in rat megakaryocytes and HEL cells, yet non-selective between monovalent anions and cations in mouse megakaryocytes. In conclusion, the present study further highlights the difference in ionic selectivity of TMEM16F in platelet lineage cells of the mouse compared to other mammalian species. This provides additional support for the ionic "leak" hypothesis that the scramblase activity of TMEM16F does not rely upon its ability to conduct ions of a specific type.


Subject(s)
Anoctamins/antagonists & inhibitors , Calcium/metabolism , Megakaryocytes/metabolism , Phospholipid Transfer Proteins/antagonists & inhibitors , Animals , Biological Transport , Humans , Mice , Rats
5.
J Biol Chem ; 292(22): 9204-9217, 2017 06 02.
Article in English | MEDLINE | ID: mdl-28416610

ABSTRACT

The role of mechanosensitive (MS) Ca2+-permeable ion channels in platelets is unclear, despite the importance of shear stress in platelet function and life-threatening thrombus formation. We therefore sought to investigate the expression and functional relevance of MS channels in human platelets. The effect of shear stress on Ca2+ entry in human platelets and Meg-01 megakaryocytic cells loaded with Fluo-3 was examined by confocal microscopy. Cells were attached to glass coverslips within flow chambers that allowed applications of physiological and pathological shear stress. Arterial shear (1002.6 s-1) induced a sustained increase in [Ca2+] i in Meg-01 cells and enhanced the frequency of repetitive Ca2+ transients by 80% in platelets. These Ca2+ increases were abrogated by the MS channel inhibitor Grammostola spatulata mechanotoxin 4 (GsMTx-4) or by chelation of extracellular Ca2+ Thrombus formation was studied on collagen-coated surfaces using DiOC6-stained platelets. In addition, [Ca2+] i and functional responses of washed platelet suspensions were studied with Fura-2 and light transmission aggregometry, respectively. Thrombus size was reduced 50% by GsMTx-4, independently of P2X1 receptors. In contrast, GsMTx-4 had no effect on collagen-induced aggregation or on Ca2+ influx via TRPC6 or Orai1 channels and caused only a minor inhibition of P2X1-dependent Ca2+ entry. The Piezo1 agonist, Yoda1, potentiated shear-dependent platelet Ca2+ transients by 170%. Piezo1 mRNA transcripts and protein were detected with quantitative RT-PCR and Western blotting, respectively, in both platelets and Meg-01 cells. We conclude that platelets and Meg-01 cells express the MS cation channel Piezo1, which may contribute to Ca2+ entry and thrombus formation under arterial shear.


Subject(s)
Blood Platelets/metabolism , Calcium Signaling , Calcium/metabolism , Ion Channels/metabolism , Megakaryocytes/metabolism , Thrombosis/metabolism , Blood Platelets/pathology , Cell Line , Female , Humans , Intercellular Signaling Peptides and Proteins , Ion Channels/antagonists & inhibitors , Male , Megakaryocytes/pathology , Peptides/pharmacology , Receptors, Purinergic P2X1/metabolism , Spider Venoms/pharmacology , Stress, Mechanical , Thrombosis/pathology
7.
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
8.
Biochem Soc Trans ; 43(3): 502-7, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26009198

ABSTRACT

Pannexin-1 (Panx1) forms anion-selective channels with a permeability up to 1 kDa and represents a pathway for the release of cytosolic ATP. Several structurally similar connexin (Cx) proteins have been identified in platelets and shown to play roles in haemostasis and thrombosis. More recently, functional Panx1 channels have been demonstrated on the surface of human platelets [Taylor et al. (2014) J. Thromb. Haemost. 12, 987-998]. Since their identification in the year 2000, several mechanisms have been reported to activate Panx1 channels, including mechanical stimulation, oxygen-glucose deprivation, a rise of [Ca2+]i, caspase cleavage and phosphorylation. Within this review, the regulation of Panx1 channels is discussed, with a focus on how they may contribute to platelet function.


Subject(s)
Adenosine Triphosphate/metabolism , Blood Platelets/metabolism , Connexins/genetics , Nerve Tissue Proteins/genetics , Calcium Signaling/genetics , Connexins/metabolism , Humans , Nerve Tissue Proteins/metabolism , Phosphorylation , Platelet Activation/genetics
9.
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
10.
Blood ; 119(15): 3613-21, 2012 Apr 12.
Article in English | MEDLINE | ID: mdl-22228626

ABSTRACT

Inhibition of Ca(2+) mobilization by cyclic nucleotides is central to the mechanism whereby endothelial-derived prostacyclin and nitric oxide limit platelet activation in the intact circulation. However, we show that ∼ 50% of the Ca(2+) response after stimulation of glycoprotein VI (GPVI) by collagen, or of Toll-like 2/1 receptors by Pam(3)Cys-Ser-(Lys)(4) (Pam(3)CSK(4)), is resistant to prostacyclin. At low agonist concentrations, the prostacyclin-resistant Ca(2+) response was predominantly because of P2X1 receptors activated by ATP release via a phospholipase-C-coupled secretory pathway requiring both protein kinase C and cytosolic Ca(2+) elevation. At higher agonist concentrations, an additional pathway was observed because of intracellular Ca(2+) release that also depended on activation of phospholipase C and, for TLR 2/1, PI3-kinase. Secondary activation of P2X1-dependent Ca(2+) influx also persisted in the presence of nitric oxide, delivered from spermine NONOate, or increased ectonucleotidase levels (apyrase). Surprisingly, apyrase was more effective than prostacyclin and NO at limiting secondary P2X1 activation. Dilution of platelets reduced the average extracellular ATP level without affecting the percentage contribution of P2X1 receptors to collagen-evoked Ca(2+) responses, indicating a highly efficient activation mechanism by local ATP. In conclusion, platelets possess inhibitor-resistant Ca(2+) mobilization pathways, including P2X1 receptors, that may be particularly important during early thrombotic or immune-dependent platelet activation.


Subject(s)
Blood Platelets/metabolism , Calcium/metabolism , Platelet Membrane Glycoproteins/metabolism , Receptors, Purinergic P2X1/metabolism , Toll-Like Receptors/metabolism , Adenosine Triphosphate/metabolism , Animals , Calcium Signaling/drug effects , Calcium Signaling/physiology , Cattle , Cells, Cultured , Collagen Type I/pharmacology , Electrophysiological Phenomena/drug effects , Epoprostenol/pharmacology , Humans , Intracellular Space/drug effects , Intracellular Space/metabolism , Intracellular Space/physiology , Purinergic P2 Receptor Agonists/metabolism , Purinergic P2 Receptor Agonists/pharmacology
11.
J Biol Chem ; 287(39): 32747-54, 2012 Sep 21.
Article in English | MEDLINE | ID: mdl-22851178

ABSTRACT

We have used selective inhibitors to determine whether the molecular chaperone heat shock protein 90 (HSP90) has an effect on both recombinant and native human P2X1 receptors. P2X1 receptor currents in HEK293 cells were reduced by ∼70-85% by the selective HSP90 inhibitor geldanamycin (2 µM, 20 min). This was associated with a speeding in the time course of desensitization as well as a reduction in cell surface expression. Imaging in real time of photoactivatable GFP-tagged P2X receptors showed that they are highly mobile. Geldanamycin almost abolished this movement for P2X1 receptors but had no effect on P2X2 receptor trafficking. P2X1/2 receptor chimeras showed that the intracellular N and C termini were involved in geldanamycin sensitivity. Geldanamycin also inhibited native P2X1 receptor-mediated responses. Platelet P2X1 receptors play an important role in hemostasis, contribute to amplification of signaling to a range of stimuli including collagen, and are novel targets for antithrombotic therapies. Platelet P2X1 receptor-, but not P2Y1 receptor-, mediated increases in intracellular calcium were reduced by 40-45% following HSP90 inhibition with geldanamycin or radicicol. Collagen stimulation leads to ATP release from platelets, and calcium increases to low doses of collagen were also reduced by ∼40% by the HSP90 inhibitors consistent with an effect on P2X1 receptors. These studies suggest that HSP90 inhibitors may be as effective as selective antagonists in regulating platelet P2X1 receptors, and their potential effects on hemostasis should be considered in clinical studies.


Subject(s)
Benzoquinones/pharmacology , Blood Platelets/metabolism , Enzyme Inhibitors/pharmacology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Lactams, Macrocyclic/pharmacology , Receptors, Purinergic P2X1/metabolism , Adenosine Triphosphate/genetics , Adenosine Triphosphate/metabolism , Blood Platelets/cytology , Collagen/genetics , Collagen/metabolism , Collagen/pharmacology , Female , HEK293 Cells , HSP90 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/metabolism , Hemostasis/drug effects , Humans , Male , Protein Transport/drug effects , Receptors, Purinergic P2X1/genetics , Receptors, Purinergic P2Y1/genetics , Receptors, Purinergic P2Y1/metabolism
12.
Circulation ; 125(20): 2479-91, 2012 May 22.
Article in English | MEDLINE | ID: mdl-22528526

ABSTRACT

BACKGROUND: Connexins are a widespread family of membrane proteins that assemble into hexameric hemichannels, also known as connexons. Connexons regulate membrane permeability in individual cells or couple between adjacent cells to form gap junctions and thereby provide a pathway for regulated intercellular communication. We have examined the role of connexins in platelets, blood cells that circulate in isolation but on tissue injury adhere to each other and the vessel wall to prevent blood loss and to facilitate wound repair. METHODS AND RESULTS: We report the presence of connexins in platelets, notably connexin37, and that the formation of gap junctions within platelet thrombi is required for the control of clot retraction. Inhibition of connexin function modulated a range of platelet functional responses before platelet-platelet contact and reduced laser-induced thrombosis in vivo in mice. Deletion of the Cx37 gene (Gja4) in transgenic mice reduced platelet aggregation, fibrinogen binding, granule secretion, and clot retraction, indicating an important role for connexin37 hemichannels and gap junctions in platelet thrombus function. CONCLUSIONS: Together, these data demonstrate that platelet gap junctions and hemichannels underpin the control of hemostasis and thrombosis and represent potential therapeutic targets.


Subject(s)
Blood Platelets/physiology , Connexins/genetics , Gap Junctions/physiology , Hemostasis/physiology , Thrombosis/physiopathology , Animals , Blood Platelets/cytology , Blood Platelets/ultrastructure , Calcium Signaling/drug effects , Calcium Signaling/physiology , Calcium Signaling/radiation effects , Carbenoxolone/pharmacology , Cell Communication/physiology , Clot Retraction/physiology , Connexin 43/metabolism , Connexins/metabolism , Fluorescence Recovery After Photobleaching , Gap Junctions/drug effects , Gap Junctions/ultrastructure , HeLa Cells , Humans , Mice , Mice, Transgenic , Microscopy, Electron, Transmission , Platelet Aggregation Inhibitors/pharmacology , Gap Junction beta-1 Protein , Gap Junction alpha-4 Protein
13.
Br J Haematol ; 153(1): 83-91, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21332705

ABSTRACT

ADP is considered a weak platelet agonist due to the limited aggregation responses it induces in vitro at physiological concentrations of extracellular Ca(2+) [(Ca(2+) )(o) ]. Lowering [Ca(2+) ](o) paradoxically enhances ADP-evoked aggregation, an effect that has been attributed to enhanced thromboxane A(2) production. This study examined the role of ectonucleotidases in the [Ca(2+) ](o) -dependence of platelet activation. Reducing [Ca(2+) ](o) from millimolar to micromolar levels converted ADP (10 µmol/l)-evoked platelet aggregation from a transient to a sustained response in both platelet-rich plasma and washed suspensions. Blocking thromboxane A(2) production with aspirin had no effect on this [Ca(2+) ](o) -dependence. Prevention of ADP degradation abolished the differences between low and physiological [Ca(2+) ](o) resulting in a robust and sustained aggregation in both conditions. Measurements of extracellular ADP revealed reduced degradation in both plasma and apyrase-containing saline at micromolar compared to millimolar [Ca(2+) ](o) . As reported previously, thromboxane A(2) generation was enhanced at low [Ca(2+) ](o) , however this was independent of ectonucleotidase activity(.) P2Y receptor antagonists cangrelor and MRS2179 demonstrated the necessity of P2Y(12) receptors for sustained ADP-evoked aggregation, with a minor role for P2Y(1) . In conclusion, Ca(2+) -dependent ectonucleotidase activity is a major factor determining the extent of platelet aggregation to ADP and must be controlled for in studies of P2Y receptor activation.


Subject(s)
Adenosine Diphosphate/pharmacology , Calcium/pharmacology , Platelet Aggregation/drug effects , Receptors, Purinergic P2Y12/physiology , Adenosine/pharmacology , Blood Platelets/metabolism , Calcium/administration & dosage , Cells, Cultured , Dose-Response Relationship, Drug , Drug Interactions , Humans , Thromboxane A2/biosynthesis
14.
Purinergic Signal ; 7(3): 341-56, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21484087

ABSTRACT

Extracellular nucleotides are ubiquitous signalling molecules, acting via the P2 class of surface receptors. Platelets express three P2 receptor subtypes, ADP-dependent P2Y1 and P2Y12 G-protein-coupled receptors and the ATP-gated P2X1 non-selective cation channel. Platelet P2X1 receptors can generate significant increases in intracellular Ca(2+), leading to shape change, movement of secretory granules and low levels of α(IIb)ß(3) integrin activation. P2X1 can also synergise with several other receptors to amplify signalling and functional events in the platelet. In particular, activation of P2X1 receptors by ATP released from dense granules amplifies the aggregation responses to low levels of the major agonists, collagen and thrombin. In vivo studies using transgenic murine models show that P2X1 receptors amplify localised thrombosis following damage of small arteries and arterioles and also contribute to thromboembolism induced by intravenous co-injection of collagen and adrenaline. In vitro, under flow conditions, P2X1 receptors contribute more to aggregate formation on collagen-coated surfaces as the shear rate is increased, which may explain their greater contribution to localised thrombosis in arterioles compared to venules within in vivo models. Since shear increases substantially near sites of stenosis, anti-P2X1 therapy represents a potential means of reducing thrombotic events at atherosclerotic plaques.

15.
J Physiol ; 588(Pt 10): 1683-93, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20351041

ABSTRACT

Lysophosphatidic acid (LPA) G-protein-coupled receptors (GPCRs) play important roles in a variety of physiological and pathophysiological processes, including cell proliferation, angiogenesis, central nervous system development and carcinogenesis. Whilst many ion channels and transporters are recognized to be controlled by a change in cell membrane potential, little is known about the voltage dependence of other proteins involved in cell signalling. Here, we show that the InsP(3)-mediated Ca(2+) response stimulated by the endogenous LPA GPCR in Xenopus oocytes is potentiated by membrane depolarization. Depolarization was able to repetitively stimulate transient [Ca(2+)](i) increases after the initial agonist-evoked response. In addition, the initial rate and amplitude of the LPA-dependent Ca(2+) response were significantly modulated by the steady holding potential over the physiological range, such that the response to LPA was potentiated at depolarized potentials and inhibited at hyperpolarized potentials. Enhancement of LPA receptor-evoked Ca(2+) mobilization by membrane depolarization was observed over a wide range of agonist concentrations. Importantly, the amplitude of the depolarization-evoked intracellular Ca(2+) increase displayed an inverse relationship with agonist concentration such that the greatest effect of voltage was observed at near-threshold levels of agonist. Voltage-dependent Ca(2+) release was not induced by direct elevation of InsP(3) or by activation of heterotrimeric G-proteins in the absence of agonist, indicating that the LPA GPCR itself represents the primary site of action of membrane voltage. This novel modulation of LPA signalling by membrane potential may have important consequences for control of Ca(2+) signals both in excitable and non-excitable tissues.


Subject(s)
Lysophospholipids/physiology , Receptors, G-Protein-Coupled/physiology , Animals , Calcium/metabolism , Electrophysiology , Inosine Triphosphate/physiology , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Membrane Potentials/drug effects , Microinjections , Microscopy, Fluorescence , Oocytes , Solutions , Xenopus
16.
J Physiol ; 588(Pt 9): 1399-406, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20308249

ABSTRACT

A delayed rectifier voltage-gated K(+) channel (Kv) represents the largest ionic conductance of platelets and megakaryocytes, but is undefined at the molecular level. Quantitative RT-PCR of all known Kv alpha and ancillary subunits showed that only Kv1.3 (KCNA3) is substantially expressed in human platelets. Furthermore, megakaryocytes from Kv1.3(/) mice or from wild-type mice exposed to the Kv1.3 blocker margatoxin completely lacked Kv currents and displayed substantially depolarised resting membrane potentials. In human platelets, margatoxin reduced the P2X(1)- and thromboxaneA(2) receptor-evoked [Ca(2+)](i) increases and delayed the onset of store-operated Ca(2+) influx. Megakaryocyte development was normal in Kv1.3(/) mice, but the platelet count was increased, consistent with a role of Kv1.3 in apoptosis or decreased platelet activation. We conclude that Kv1.3 forms the Kv channel of the platelet and megakaryocyte, which sets the resting membrane potential, regulates agonist-evoked Ca(2+) increases and influences circulating platelet numbers.


Subject(s)
Blood Platelets/physiology , Calcium Signaling/physiology , Kv1.3 Potassium Channel/blood , Megakaryocytes/physiology , Membrane Potentials/physiology , Platelet Count , Animals , Blood Platelets/drug effects , Blood Platelets/ultrastructure , Calcium Signaling/drug effects , Cell Size , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Humans , In Vitro Techniques , Megakaryocytes/drug effects , Megakaryocytes/ultrastructure , Membrane Potentials/drug effects , Mice , Mice, Inbred C57BL , Patch-Clamp Techniques , Reverse Transcriptase Polymerase Chain Reaction , Scorpion Venoms/pharmacology , Second Messenger Systems/physiology
17.
J Neurochem ; 113(5): 1177-87, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20374431

ABSTRACT

P2X1 receptors for ATP contribute to signalling in a variety of cell types and following stimulation undergo rapid desensitisation (within 1 s), and require approximately 5 min to recover. In HEK293 cells P2X1 receptors C-terminally tagged with enhanced green fluorescent protein (P2X1-eGFP) were predominantly expressed at the cell surface. Following > 90% photo-bleaching of P2X1-eGFP within a 6 microm(2) circle at the cell surface fluorescence recovery after photo-bleaching (FRAP) was fit with a time constant of approximately 60 s and recovered to approximately 75% of pre-bleach levels. Following activation of the P2X1 receptor with alpha,beta-methylene ATP the associated calcium influx doubled the FRAP recovery rate. The protein synthesis inhibitor cycloheximide had only a small effect on repeated FRAP and indicated a limited contribution of new P2X1 receptors to the FRAP. Inhibition of trafficking with brefeldin A reduced recovery and this effect could be reversed following receptor activation. In contrast, the dynamin inhibitor dynasore had no effect on FRAP under unstimulated conditions but reduced the level of recovery following agonist stimulation. In functional studies both brefeldin A and dynasore increased the recovery time from desensitisation. Taken together these studies demonstrate for the first time an important role of receptor recycling on P2X1 receptor responsiveness.


Subject(s)
Receptors, Purinergic P2/genetics , Receptors, Purinergic P2/metabolism , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Brefeldin A/pharmacology , Cell Line , Cycloheximide/pharmacology , Data Interpretation, Statistical , Dynamins/antagonists & inhibitors , Electrophysiology , Fluorescence Recovery After Photobleaching , Green Fluorescent Proteins , Humans , Hydrazones/pharmacology , Microscopy, Confocal , Mutagenesis, Insertional , Patch-Clamp Techniques , Protein Synthesis Inhibitors/pharmacology , Purinergic P2 Receptor Agonists , Receptors, Purinergic P2X , Second Messenger Systems/drug effects
18.
Sci Signal ; 13(615)2020 01 21.
Article in English | MEDLINE | ID: mdl-31964805

ABSTRACT

Cells sense extracellular nucleotides through the P2Y class of purinergic G protein-coupled receptors (GPCRs), which stimulate integrin activation through signaling events, including intracellular Ca2+ mobilization. We investigated the relationship between P2Y-stimulated repetitive Ca2+ waves and fibrinogen binding to the platelet integrin αIIbß3 (GPIIb/IIIa) through confocal fluorescence imaging of primary rat megakaryocytes. Costimulation of the receptors P2Y1 and P2Y12 generated a series of Ca2+ transients that each induced a rapid, discrete increase in fibrinogen binding. The peak and net increase of individual fibrinogen binding events correlated with the Ca2+ transient amplitude and frequency, respectively. Using BAPTA loading and selective receptor antagonists, we found that Ca2+ mobilization downstream of P2Y1 was essential for ADP-evoked fibrinogen binding, whereas P2Y12 and the kinase PI3K were also required for αIIbß3 activation and enhanced the number of Ca2+ transients. ADP-evoked fibrinogen binding was initially uniform over the cell periphery but subsequently redistributed with a polarity that correlated with the direction of the Ca2+ waves. Polarization of αIIbß3 may be mediated by the actin cytoskeleton, because surface-bound fibrinogen is highly immobile, and its motility was enhanced by cytoskeletal disruption. In conclusion, spatial and temporal patterns of Ca2+ increase enable fine control of αIIbß3 activation after cellular stimulation. P2Y1-stimulated Ca2+ transients coupled to αIIbß3 activation only in the context of P2Y12 coactivation, thereby providing an additional temporal mechanism of synergy between these Gq- and Gi-coupled GPCRs.


Subject(s)
Calcium/metabolism , Megakaryocytes/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Receptors, Purinergic/metabolism , Signal Transduction/physiology , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Adenosine Diphosphate/pharmacology , Animals , Cells, Cultured , Fibrinogen/metabolism , Humans , Male , Megakaryocytes/cytology , Megakaryocytes/drug effects , Microscopy, Confocal , Rats, Wistar , Signal Transduction/drug effects
19.
Trends Pharmacol Sci ; 29(8): 421-9, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18621424

ABSTRACT

G-protein-coupled receptors (GPCRs) have ubiquitous roles in transducing extracellular signals into cellular responses. Therefore, the concept that members of this superfamily of surface proteins are directly modulated by changes in membrane voltage could have widespread consequences for cell signalling. Although several studies have indicated that GPCRs can be voltage dependent, particularly P2Y(1) receptors in the non-excitable megakaryocyte, the evidence has been mostly indirect. Recent work on muscarinic receptors has stimulated substantial interest in this field by reporting the first voltage-dependent charge movements for a GPCR. An underlying mechanism is proposed whereby a voltage-induced conformational change in the receptor alters its ability to couple to the G protein and thereby influences its affinity for an agonist. We discuss the strength of the evidence behind this hypothesis and include suggestions for future work. We also describe other examples in which direct voltage control of GPCRs can account for effects of membrane potential on downstream signals and highlight the possible physiological consequences of this phenomenon.


Subject(s)
Membrane Potentials , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Animals , Humans , Megakaryocytes/metabolism , Potassium Channels, Voltage-Gated/metabolism , Protein Conformation , Receptors, G-Protein-Coupled/agonists
20.
Platelets ; 19(4): 308-13, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18569867

ABSTRACT

Store-operated Ca2+ influx represents a major route by which cytosolic Ca2+ can be elevated during platelet activation, yet its molecular identity in this cell type remains highly controversial. Using quantitative RT-PCR analysis of candidate receptor-operated cation entry pathways in human platelets, we show a >30-fold higher expression of message for the recently discovered Orai1 store-operated Ca2+ channel, and also the store Ca2+ sensor STIM1, when compared to the non-selective cation channels TRPC1, TRPC6 and TRPM2. Orai1 and STIM1 gene transcripts were also detected at higher levels than TRPC1, TRPC6 and TRPM2 in primary murine megakaryocytes and human megakaryocytic cell lines. In direct electrophysiological recordings from murine megakaryocytes, Ca2+ ionophore-induced store depletion stimulated CRAC currents, which are known to require Orai1, and these overlapped with TRPC6-like currents following P2Y receptor activation. Together with recent transgenic studies, these data provide evidence for STIM1:Orai1 as a primary pathway for agonist-evoked Ca2+ influx in the platelet and megakaryocyte.


Subject(s)
Blood Platelets/metabolism , Calcium Channels/physiology , Calcium/metabolism , Megakaryocytes/metabolism , Animals , Calcium Channels/genetics , Cell Line , Electrophysiology , Gene Expression Profiling , Humans , Membrane Proteins/genetics , Mice , Neoplasm Proteins/genetics , ORAI1 Protein , RNA, Messenger/analysis , Stromal Interaction Molecule 1
SELECTION OF CITATIONS
SEARCH DETAIL