Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
1.
Platelets ; 33(3): 451-461, 2022 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-34348571

RESUMEN

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.


Asunto(s)
Plaquetas/metabolismo , Colágeno/metabolismo , Integrina alfa2beta1/metabolismo , Adhesividad Plaquetaria/fisiología , Agregación Plaquetaria/fisiología , Canales de Potasio con Entrada de Voltaje/metabolismo , Humanos
2.
Platelets ; 32(7): 872-879, 2021 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-33872124

RESUMEN

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.


Asunto(s)
Plaquetas/metabolismo , Canales de Potasio/metabolismo , Animales , Humanos , Ratones
3.
Purinergic Signal ; 15(3): 397-402, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31286385

RESUMEN

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.


Asunto(s)
Técnicas de Sustitución del Gen/métodos , Receptores Purinérgicos P2X1/análisis , Receptores Purinérgicos P2X1/metabolismo , Animales , Proteínas Bacterianas , Proteínas Luminiscentes , Ratones , Modelos Animales
4.
Platelets ; 30(8): 962-966, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31008669

RESUMEN

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.


Asunto(s)
Anoctaminas/antagonistas & inhibidores , Calcio/metabolismo , Megacariocitos/metabolismo , Proteínas de Transferencia de Fosfolípidos/antagonistas & inhibidores , Animales , Transporte Biológico , Humanos , Ratones , Ratas
5.
J Biol Chem ; 292(22): 9204-9217, 2017 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-28416610

RESUMEN

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.


Asunto(s)
Plaquetas/metabolismo , Señalización del Calcio , Calcio/metabolismo , Canales Iónicos/metabolismo , Megacariocitos/metabolismo , Trombosis/metabolismo , Plaquetas/patología , Línea Celular , Femenino , Humanos , Péptidos y Proteínas de Señalización Intercelular , Canales Iónicos/antagonistas & inhibidores , Masculino , Megacariocitos/patología , Péptidos/farmacología , Receptores Purinérgicos P2X1/metabolismo , Venenos de Araña/farmacología , Estrés Mecánico , Trombosis/patología
6.
J Immunol ; 196(12): 4877-84, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27183585

RESUMEN

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.


Asunto(s)
Asma/inmunología , Adhesión Celular , Eosinófilos/fisiología , Receptores Purinérgicos P2X1/metabolismo , Adenosina Trifosfato/análogos & derivados , Adenosina Trifosfato/farmacología , Apirasa/farmacología , Asma/fisiopatología , Bencenosulfonatos/farmacología , Antígeno CD11b/genética , Antígeno CD11b/metabolismo , Eosinófilos/efectos de los fármacos , Eosinófilos/inmunología , Voluntarios Sanos , Humanos , Recuento de Leucocitos , Agonistas del Receptor Purinérgico P2X/farmacología , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Purinérgicos P2X1/genética , Receptores Purinérgicos P2X4/genética , Receptores Purinérgicos P2X5/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
7.
J Biol Chem ; 291(34): 17907-18, 2016 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-27325704

RESUMEN

The ether à go-go family of voltage-gated potassium channels is structurally distinct. The N terminus contains an eag domain (eagD) that contains a Per-Arnt-Sim (PAS) domain that is preceded by a conserved sequence of 25-27 amino acids known as the PAS-cap. The C terminus contains a region with homology to cyclic nucleotide binding domains (cNBHD), which is directly linked to the channel pore. The human EAG1 (hEAG1) channel is remarkably sensitive to inhibition by intracellular calcium (Ca(2+) i) through binding of Ca(2+)-calmodulin to three sites adjacent to the eagD and cNBHD. Here, we show that the eagD and cNBHD interact to modulate Ca(2+)-calmodulin as well as voltage-dependent gating. Sustained elevation of Ca(2+) i resulted in an initial profound inhibition of hEAG1 currents, which was followed by a phase when current amplitudes partially recovered, but activation gating was slowed and shifted to depolarized potentials. Deletion of either the eagD or cNBHD abolished the inhibition by Ca(2+) i However, deletion of just the PAS-cap resulted in a >15-fold potentiation in response to elevated Ca(2+) i Mutations of residues at the interface between the eagD and cNBHD have been linked to human cancer. Glu-600 on the cNBHD, when substituted with residues with a larger volume, resulted in hEAG1 currents that were profoundly potentiated by Ca(2+) i in a manner similar to the ΔPAS-cap mutant. These findings provide the first evidence that eagD and cNBHD interactions are regulating Ca(2+)-dependent gating and indicate that the binding of the PAS-cap with the cNBHD is required for the closure of the channels upon CaM binding.


Asunto(s)
Calcio/metabolismo , Calmodulina/metabolismo , Canales de Potasio Éter-A-Go-Go/metabolismo , Secuencia de Aminoácidos , Animales , Calmodulina/genética , Canales de Potasio Éter-A-Go-Go/genética , Humanos , Dominios Proteicos , Eliminación de Secuencia , Xenopus laevis
9.
Adv Exp Med Biol ; 898: 305-29, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27161234

RESUMEN

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.


Asunto(s)
Plaquetas/metabolismo , Señalización del Calcio , Activación del Canal Iónico , Canales Iónicos/metabolismo , Receptores Purinérgicos P2X1/metabolismo , Animales , Citosol/metabolismo , Humanos , Canales Iónicos/química , Ligandos , Conformación Proteica , Receptores Purinérgicos P2X1/química
10.
Biochem Soc Trans ; 43(3): 502-7, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26009198

RESUMEN

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.


Asunto(s)
Adenosina Trifosfato/metabolismo , Plaquetas/metabolismo , Conexinas/genética , Proteínas del Tejido Nervioso/genética , Señalización del Calcio/genética , Conexinas/metabolismo , Humanos , Proteínas del Tejido Nervioso/metabolismo , Fosforilación , Activación Plaquetaria/genética
11.
Mol Pharmacol ; 86(3): 243-51, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24923466

RESUMEN

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.


Asunto(s)
Adenosina Difosfato/metabolismo , Plaquetas/metabolismo , Calcio/metabolismo , Agregación Plaquetaria , Receptores Purinérgicos P2X1/metabolismo , Receptores Purinérgicos P2Y1/metabolismo , Señalización del Calcio , Células HEK293 , Humanos , Técnicas In Vitro , Proteínas Recombinantes/metabolismo
12.
Blood ; 119(15): 3613-21, 2012 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-22228626

RESUMEN

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.


Asunto(s)
Plaquetas/metabolismo , Calcio/metabolismo , Glicoproteínas de Membrana Plaquetaria/metabolismo , Receptores Purinérgicos P2X1/metabolismo , Receptores Toll-Like/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/fisiología , Bovinos , Células Cultivadas , Colágeno Tipo I/farmacología , Fenómenos Electrofisiológicos/efectos de los fármacos , Epoprostenol/farmacología , Humanos , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/metabolismo , Espacio Intracelular/fisiología , Agonistas del Receptor Purinérgico P2/metabolismo , Agonistas del Receptor Purinérgico P2/farmacología
13.
J Biol Chem ; 287(39): 32747-54, 2012 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-22851178

RESUMEN

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.


Asunto(s)
Benzoquinonas/farmacología , Plaquetas/metabolismo , Inhibidores Enzimáticos/farmacología , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Lactamas Macrocíclicas/farmacología , Receptores Purinérgicos P2X1/metabolismo , Adenosina Trifosfato/genética , Adenosina Trifosfato/metabolismo , Plaquetas/citología , Colágeno/genética , Colágeno/metabolismo , Colágeno/farmacología , Femenino , Células HEK293 , Proteínas HSP90 de Choque Térmico/genética , Proteínas HSP90 de Choque Térmico/metabolismo , Hemostasis/efectos de los fármacos , Humanos , Masculino , Transporte de Proteínas/efectos de los fármacos , Receptores Purinérgicos P2X1/genética , Receptores Purinérgicos P2Y1/genética , Receptores Purinérgicos P2Y1/metabolismo
14.
Circulation ; 125(20): 2479-91, 2012 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-22528526

RESUMEN

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.


Asunto(s)
Plaquetas/fisiología , Conexinas/genética , Uniones Comunicantes/fisiología , Hemostasis/fisiología , Trombosis/fisiopatología , Animales , Plaquetas/citología , Plaquetas/ultraestructura , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/fisiología , Señalización del Calcio/efectos de la radiación , Carbenoxolona/farmacología , Comunicación Celular/fisiología , Retracción del Coagulo/fisiología , Conexina 43/metabolismo , Conexinas/metabolismo , Recuperación de Fluorescencia tras Fotoblanqueo , Uniones Comunicantes/efectos de los fármacos , Uniones Comunicantes/ultraestructura , Células HeLa , Humanos , Ratones , Ratones Transgénicos , Microscopía Electrónica de Transmisión , Inhibidores de Agregación Plaquetaria/farmacología , Proteína beta1 de Unión Comunicante , Proteína alfa-4 de Unión Comunicante
15.
Br J Haematol ; 153(1): 83-91, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21332705

RESUMEN

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.


Asunto(s)
Adenosina Difosfato/farmacología , Calcio/farmacología , Agregación Plaquetaria/efectos de los fármacos , Receptores Purinérgicos P2Y12/fisiología , Adenosina/farmacología , Plaquetas/metabolismo , Calcio/administración & dosificación , Células Cultivadas , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Humanos , Tromboxano A2/biosíntesis
16.
Purinergic Signal ; 7(3): 341-56, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21484087

RESUMEN

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.

17.
Genesis ; 48(2): 127-36, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20049953

RESUMEN

CLIC1 belongs to a family of highly conserved and widely expressed intracellular chloride ion channel proteins existing in both soluble and membrane integrated forms. To study the physiological and biological role of CLIC1 in vivo, we undertook conditional gene targeting to engineer Clic1 gene knock-out mice. This represents creation of the first gene knock-out of a vertebrate CLIC protein family member. We first generated a Clic1 Knock-in (Clic1(FN)) allele, followed by Clic1 knock-out (Clic1(-/-)) mice by crossing Clic1(FN) allele with TNAP-cre mice, resulting in germline gene deletion through Cre-mediated recombination. Mice heterozygous or homozygous for these alleles are viable and fertile and appear normal. However, Clic1(-) (/-) mice show a mild platelet dysfunction characterized by prolonged bleeding times and decreased platelet activation in response to adenosine diphosphate stimulation linked to P2Y(12) receptor signaling.


Asunto(s)
Canales de Cloruro/genética , Eliminación de Gen , Marcación de Gen/métodos , Ingeniería Genética , Modelos Genéticos , Alelos , Animales , Plaquetas/metabolismo , Cruzamientos Genéticos , Hemorragia , Heterocigoto , Homocigoto , Inmunohistoquímica , Integrasas/metabolismo , Ratones , Ratones Noqueados , Recombinación Genética
18.
J Physiol ; 588(Pt 10): 1683-93, 2010 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-20351041

RESUMEN

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.


Asunto(s)
Lisofosfolípidos/fisiología , Receptores Acoplados a Proteínas G/fisiología , Animales , Calcio/metabolismo , Electrofisiología , Inosina Trifosfato/fisiología , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Potenciales de la Membrana/efectos de los fármacos , Microinyecciones , Microscopía Fluorescente , Oocitos , Soluciones , Xenopus
19.
J Physiol ; 588(Pt 9): 1399-406, 2010 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-20308249

RESUMEN

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.


Asunto(s)
Plaquetas/fisiología , Señalización del Calcio/fisiología , Canal de Potasio Kv1.3/sangre , Megacariocitos/fisiología , Potenciales de la Membrana/fisiología , Recuento de Plaquetas , Animales , Plaquetas/efectos de los fármacos , Plaquetas/ultraestructura , Señalización del Calcio/efectos de los fármacos , Tamaño de la Célula , ADN Complementario/biosíntesis , ADN Complementario/genética , Humanos , Técnicas In Vitro , Megacariocitos/efectos de los fármacos , Megacariocitos/ultraestructura , Potenciales de la Membrana/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Técnicas de Placa-Clamp , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Venenos de Escorpión/farmacología , Sistemas de Mensajero Secundario/fisiología
20.
J Neurochem ; 113(5): 1177-87, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20374431

RESUMEN

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.


Asunto(s)
Receptores Purinérgicos P2/genética , Receptores Purinérgicos P2/metabolismo , Adenosina Trifosfato/análogos & derivados , Adenosina Trifosfato/farmacología , Brefeldino A/farmacología , Línea Celular , Cicloheximida/farmacología , Interpretación Estadística de Datos , Dinaminas/antagonistas & inhibidores , Electrofisiología , Recuperación de Fluorescencia tras Fotoblanqueo , Proteínas Fluorescentes Verdes , Humanos , Hidrazonas/farmacología , Microscopía Confocal , Mutagénesis Insercional , Técnicas de Placa-Clamp , Inhibidores de la Síntesis de la Proteína/farmacología , Agonistas del Receptor Purinérgico P2 , Receptores Purinérgicos P2X , Sistemas de Mensajero Secundario/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA