Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Res Pract Thromb Haemost ; 8(1): 102326, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38404940

RESUMEN

Background: Inositol 1,3,4,5-tetrakisphosphate (IP4) is formed from inositol 1,4,5-trisphosphate (IP3) by IP3 3-kinase (ITPK) in most cells. Its function is unknown but has been suggested to be involved in Ca2+ entry, IP3 regulation, and phosphoinositide 3-kinase antagonism. Objectives: To better elucidate a function for IP4, we tested a specific inhibitor of ITPK (GNF362) on platelets, the effects of IP4 directly in permeabilized platelets and its effect on phosphatidylinositol 3,4,5-trisphosphate (PIP3) binding to pleckstrin-homology (PH) domain-containing proteins in platelets. Methods: Human platelets were utilized in whole blood for thrombus formation, in platelet-rich plasma and washed suspensions for aggregation, and for Ca2+ studies, or resuspended in high K+ and low Na+ buffers for permeabilization experiments. Phosphorylation of AKT-Ser473 and Rap1-GTP formation were measured by Western blotting and PIP3 binding using PIP3 beads. Results: GNF362-enhanced platelet aggregation stimulated by low concentrations of ADP, collagen, thrombin, U46619, and thrombus formation in collagen-coated capillaries. GNF362 induced a transient elevation of Ca2+ concentration, elevated basal levels of IP3, and enhanced the peak height of Ca2+ elevated by agonists. In permeabilized platelets, IP4 inhibited GTPγS induced formation of AKT-Ser473 phosphorylation and platelet aggregation. IP4 reduced GTPγS-stimulated Rap1-GTP levels and potently reduced extraction of RASA3 and BTK by PIP3 beads. Conclusion: ITPK and IP4 are negative regulators of platelet function. IP4 regulation of PH domain-containing proteins may represent a pathway by which platelet activation may be controlled during thrombosis.

3.
J Clin Invest ; 110(2): 247-57, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12122117

RESUMEN

Griscelli syndrome (GS) patients and the corresponding mouse model ashen exhibit defects mainly in two types of lysosome-related organelles, melanosomes in melanocytes and lytic granules in CTLs. This disease is caused by loss-of-function mutations in RAB27A, which encodes 1 of the 60 known Rab GTPases, critical regulators of vesicular transport. Here we present evidence that Rab27a function can be compensated by a closely related protein, Rab27b. Rab27b is expressed in platelets and other tissues but not in melanocytes or CTLs. Morphological and functional tests in platelets derived from ashen mice are all within normal limits. Both Rab27a and Rab27b are found associated with the limiting membrane of platelet-dense granules and to a lesser degree with alpha-granules. Ubiquitous transgenic expression of Rab27a or Rab27b rescues ashen coat color, and melanocytes derived from transgenic mice exhibit widespread peripheral distribution of melanosomes instead of the perinuclear clumping observed in ashen melanocytes. Finally, transient expression in ashen melanocytes of Rab27a or Rab27b, but not other Rab's, restores peripheral distribution of melanosomes. Our data suggest that Rab27b is functionally redundant with Rab27a and that the pathogenesis of GS is determined by the relative expression of Rab27a and Rab27b in specialized cell types.


Asunto(s)
Histiocitosis de Células no Langerhans/etiología , Hipopigmentación/etiología , Proteínas de Unión al GTP rab/fisiología , Animales , Plaquetas/patología , Plaquetas/fisiología , Modelos Animales de Enfermedad , Expresión Génica , Histiocitosis de Células no Langerhans/genética , Histiocitosis de Células no Langerhans/patología , Histiocitosis de Células no Langerhans/fisiopatología , Humanos , Hipopigmentación/genética , Hipopigmentación/patología , Hipopigmentación/fisiopatología , Melanocitos/patología , Melanocitos/fisiología , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Mutantes , Ratones Transgénicos , Mutación , Síndrome , Linfocitos T Citotóxicos/fisiología , Proteínas de Unión al GTP rab/genética , Proteínas rab27 de Unión a GTP
4.
Blood ; 113(9): 1872-3, 2009 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-19246564
5.
Cell Signal ; 27(7): 1488-98, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25797048

RESUMEN

Phosphatidylinositol trisphosphate (PIP3) has been implicated in many platelet functions however many of the mechanisms need clarification. We have used cell permeable analogues of PIP3,1-O-(1,2-di-palmitoyl-sn-glyero-3-O-phosphoryl)-D-myo-inositol-3,4,5-trisphosphate (DiC16-PIP3) or 1-O-(1,2-di-octanoyl-sn-glyero-3-O-phosphoryl)-D-myo-inositol-3,4,5-trisphosphate (DiC8-PIP3) to study their effects on activation on washed human platelets. Addition of either DiC8- or DiC16-PIP3 to human platelets induced aggregation in the presence of extracellular Ca(2+). This was reduced by the presence of indomethacin, the phospholipase C inhibitor U73122 and apyrase. DiC8-PIP3 induced the phosphorylation of Akt-Ser(473) which was reduced by the Akt inhibitor IV, wortmannin and EGTA (suggesting a dependence on Ca(2+) entry). In Fura2 loaded platelets DiC8-PIP3 was effective at increasing intracellular Ca(2+) in a distinct and transient manner that was reduced in the presence of indomethacin, U73122 and 2-aminoethyl diphenylborinate (2APB). Ca(2+) elevation was reduced by the non-SOCE inhibitor LOE908 and also by the SOCE inhibitor BTP2. DiC8-PIP3 induced the release of Ca(2+) from stores which was not affected by the proton dissipating agent bafilomycin A1 and was more potent than the two-pore channel agonist DiC8-PI[3,5]P2 suggesting release from an endoplasmic reticulum type store. DiC8-PIP3 weakly induced the tyrosine phosphorylation of Syk but not of PLCγ2. Finally like thrombin DiC8-PIP3 induced the formation of thromboxane B2 that was inhibited by the Akt inhibitor IV. These studies suggest that PIP3 via Ca(2+) elevation and Akt phosphorylation forms a central role in thromboxane A2 formation and the amplification of platelet activation.


Asunto(s)
Plaquetas/efectos de los fármacos , Calcio/metabolismo , Fosfatos de Fosfatidilinositol/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Tromboxano A2/metabolismo , Androstadienos/farmacología , Plaquetas/citología , Plaquetas/metabolismo , Ácido Egtácico/farmacología , Ensayo de Inmunoadsorción Enzimática , Fura-2/química , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Fosfolipasa C gamma/antagonistas & inhibidores , Fosfolipasa C gamma/metabolismo , Fosforilación/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Quinasa Syk , Tromboxano A2/análisis , Wortmanina
6.
Pflugers Arch ; 457(2): 377-87, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18546016

RESUMEN

Changes in [Ca(2+)](i) are a central step in platelet activation. In nonexcitable cells, receptor-mediated depletion of intracellular Ca(2+) stores triggers Ca(2+) entry through store-operated calcium (SOC) channels. Stromal interaction molecule 1 (STIM1) has been identified as an endoplasmic reticulum (ER)-resident Ca(2+) sensor that regulates store-operated calcium entry (SOCE), but the identity of the SOC channel in platelets has been controversially debated. Some investigators proposed transient receptor potential (TRP) C1 to fulfil this function based on the observation that antibodies against the channel impaired SOCE in platelets. However, others could not detect TRPC1 in the plasma membrane of platelets and raised doubts about the specificity of the inhibiting anti-TRPC1 antibodies. To address the role of TRPC1 in SOCE in platelets, we analyzed mice lacking TRPC1. Platelets from these mice display fully intact SOCE and also otherwise unaltered calcium homeostasis compared to wild-type. Furthermore, platelet function in vitro and in vivo is not altered in the absence of TRPC1. Finally, studies on human platelets revealed that the presumably inhibitory anti-TRPC1 antibodies have no specific effect on SOCE and fail to bind to the protein. Together, these results provide evidence that SOCE in platelets is mediated by channels other than TRPC1.


Asunto(s)
Plaquetas/metabolismo , Señalización del Calcio , Calcio/sangre , Canales Catiónicos TRPC/sangre , Animales , Coagulación Sanguínea , Cloruros , Modelos Animales de Enfermedad , Compuestos Férricos , Humanos , Proteínas de la Membrana/sangre , Ratones , Ratones Noqueados , Proteínas de Neoplasias/sangre , Activación Plaquetaria , Molécula de Interacción Estromal 1 , Canales Catiónicos TRPC/deficiencia , Canales Catiónicos TRPC/genética , Trombosis/sangre , Trombosis/inducido químicamente , Factores de Tiempo
7.
Proc Natl Acad Sci U S A ; 104(14): 5872-7, 2007 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-17384153

RESUMEN

The Rab27 GTPase subfamily consists of two closely related homologs, Rab27a and Rab27b. Rab27a has been shown previously to regulate organelle movement and regulated exocytosis in a wide variety of secretory cells. However, the role of the more restrictedly expressed Rab27b remains unclear. Here we describe the creation of Rab27b knockout (KO) strain that was subsequently crossed with the naturally occurring Rab27a KO line, ashen, to produce double KO (Rab27a(ash/ash) Rab27b(-/-)) mice. Rab27b KO (and double KO) exhibit significant hemorrhagic disease in contrast to ashen mice. In vitro assays demonstrated impaired aggregation with collagen and U46619 and reduced secretion of dense granules in both Rab27b and double KO strains. Additionally, we detected a 50% reduction in the number of dense granules per platelet and diminished platelet serotonin content, possibly due to a dense granule packaging defect into proplatelets during megakaryocyte maturation. The presence of Rab27a partially compensated for the secretory defect but not the reduced granule number. The morphology and function of platelet alpha-granules were unaffected. Our data suggest that Rab27b is a key regulator of dense granule secretion in platelets and thus a candidate gene for delta-storage pool deficiency in humans.


Asunto(s)
Plaquetas/ultraestructura , Gránulos Citoplasmáticos/ultraestructura , Proteínas de Unión al GTP rab/fisiología , Animales , Plaquetas/química , Gránulos Citoplasmáticos/química , Gránulos Citoplasmáticos/efectos de los fármacos , Ratones , Ratones Noqueados , Selectina-P/análisis , Agregación Plaquetaria/genética , Agregación Plaquetaria/fisiología , Recuento de Plaquetas , Serotonina/análisis , Serotonina/metabolismo , Proteínas de Unión al GTP rab/genética , Proteínas de Unión al GTP rab/farmacología
8.
Mol Cell Proteomics ; 6(3): 548-64, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17186946

RESUMEN

The platelet surface is poorly characterized due to the low abundance of many membrane proteins and the lack of specialist tools for their investigation. In this study we identified novel human platelet and mouse megakaryocyte membrane proteins using specialist proteomics and genomics approaches. Three separate methods were used to enrich platelet surface proteins prior to identification by liquid chromatography and tandem mass spectrometry: lectin affinity chromatography, biotin/NeutrAvidin affinity chromatography, and free flow electrophoresis. Many known, abundant platelet surface transmembrane proteins and several novel proteins were identified using each receptor enrichment strategy. In total, two or more unique peptides were identified for 46, 68, and 22 surface membrane, intracellular membrane, and membrane proteins of unknown subcellular localization, respectively. The majority of these were single transmembrane proteins. To complement the proteomics studies, we analyzed the transcriptome of a highly purified preparation of mature primary mouse megakaryocytes using serial analysis of gene expression in view of the increasing importance of mutant mouse models in establishing protein function in platelets. This approach identified all of the major classes of platelet transmembrane receptors, including multitransmembrane proteins. Strikingly 17 of the 25 most megakaryocyte-specific genes (relative to 30 other serial analysis of gene expression libraries) were transmembrane proteins, illustrating the unique nature of the megakaryocyte/platelet surface. The list of novel plasma membrane proteins identified using proteomics includes the immunoglobulin superfamily member G6b, which undergoes extensive alternate splicing. Specific antibodies were used to demonstrate expression of the G6b-B isoform, which contains an immunoreceptor tyrosine-based inhibition motif. G6b-B undergoes tyrosine phosphorylation and association with the SH2 domain-containing phosphatase, SHP-1, in stimulated platelets suggesting that it may play a novel role in limiting platelet activation.


Asunto(s)
Plaquetas/química , Megacariocitos/química , Proteínas de la Membrana/análisis , Animales , Membrana Celular/química , Células Cultivadas , Cromatografía de Afinidad , Expresión Génica , Genómica , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Activación Plaquetaria , Proteómica , ARN/metabolismo , Receptores Inmunológicos/análisis , Receptores Inmunológicos/metabolismo , Espectrometría de Masas en Tándem
9.
Blood ; 105(4): 1500-7, 2005 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-15466936

RESUMEN

Formation and rearrangement of disulfide bonds during the correct folding of nascent proteins is modulated by a family of enzymes known as thiol isomerases, which include protein disulfide isomerase (PDI), endoplasmic reticulum protein 5 (ERP5), and ERP57. Recent evidence supports an alternative role for this family of proteins on the surface of cells, where they are involved in receptor remodeling and recognition. In platelets, blocking PDI with inhibitory antibodies inhibits a number of platelet activation pathways, including aggregation, secretion, and fibrinogen binding. Analysis of human platelet membrane fractions identified the presence of the thiol isomerase protein ERP5. Further study showed that ERP5 is resident mainly on platelet intracellular membranes, although it is rapidly recruited to the cell surface in response to a range of platelet agonists. Blocking cell-surface ERP5 using inhibitory antibodies leads to a decrease in platelet aggregation in response to agonists, and a decrease in fibrinogen binding and P-selectin exposure. It is possible that this is based on the disruption of integrin function, as we observed that ERP5 becomes physically associated with the integrin beta(3) subunit during platelet stimulation. These results provide new insights into the involvement of thiol isomerases and regulation of platelet activation.


Asunto(s)
Plaquetas/enzimología , Plaquetas/fisiología , Proteína Disulfuro Isomerasas/fisiología , Secuencia de Aminoácidos , Anticuerpos Bloqueadores/farmacología , Membrana Celular/enzimología , Gránulos Citoplasmáticos/inmunología , Gránulos Citoplasmáticos/metabolismo , Fibrinógeno/metabolismo , Humanos , Integrina beta3/metabolismo , Líquido Intracelular/enzimología , Datos de Secuencia Molecular , Selectina-P/metabolismo , Activación Plaquetaria/fisiología , Inhibidores de Agregación Plaquetaria/farmacología , Unión Proteica/fisiología , Proteína Disulfuro Isomerasas/antagonistas & inhibidores , Proteína Disulfuro Isomerasas/inmunología , Proteína Disulfuro Isomerasas/aislamiento & purificación , Proteína Disulfuro Isomerasas/metabolismo
10.
Blood ; 100(8): 2801-11, 2002 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-12351388

RESUMEN

Store-operated Ca(++) entry (SOCE) is thought to comprise the major pathway for Ca(++) entry in platelets. Recently, a number of transient receptor potential (TRP) proteins, which have been divided into 3 groups (TRPC, TRPM, and TRPV), have been suggested as SOCE channels. We report the expression and function of TRPC proteins in human platelets. TRPC6 is found at high levels and TRPC1 at low levels. Using purified plasma (PM) and intracellular membranes (IM), TRPC6 is found in the PM, but TRPC1 is localized to the IM. Using Fura-2-loaded platelets, we report that, in line with TRPC6 expression, 1-oleoyl-2-acetyl-sn-glycerol (OAG) stimulated the entry of Ca(++) and Ba(2+) independently of protein kinase C. Thrombin also induced the entry of Ca(++) and Ba(2+), but thapsigargin, which depletes the stores, induced the entry of only Ca(++). Thus, thrombin activated TRPC6 via a SOCE-independent mechanism. In phosphorylation studies, we report that neither TRPC6 nor TRPC1 was a substrate for tyrosine kinases. TRPC6 was phosphorylated by cAMP-dependent protein kinase (cAMP-PK) and associated with other cAMP-PK substrates. TRPC1 was not phosphorylated by cAMP-PK but also associated with other substrates. Activation of cAMP-PK inhibited Ca(++) but not Ba(2+) entry induced by thrombin and neither Ca(++) nor Ba(2+) entry stimulated by OAG. These results suggest that TRPC6 is a SOCE-independent, nonselective cation entry channel stimulated by thrombin and OAG. TRPC6 is a substrate for cAMP-PK, although phosphorylation appears to not affect cation permeation. TRPC1 is located in IM, suggesting a role at the level of the stores.


Asunto(s)
Plaquetas/fisiología , Canales de Calcio/sangre , Calcio/sangre , Secuencia de Aminoácidos , Transporte Biológico , Canales de Calcio/química , Canales de Calcio/genética , Células Cultivadas , Proteínas Quinasas Dependientes de AMP Cíclico/sangre , Activación Enzimática , Fura-2 , Humanos , Cinética , Datos de Secuencia Molecular , Fragmentos de Péptidos/química , Fosfatos/sangre , Canales Catiónicos TRPC , Canal Catiónico TRPC6
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA