Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Br J Haematol ; 204(3): 988-1004, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38062782

RESUMEN

Neonatal platelets present a reduced response to the platelet agonist, thrombin (Thr), thus resulting in a deficient Thr-induced aggregation. These alterations are more pronounced in premature newborns. Here, our aim was to uncover the causes underneath the impaired Ca2+ homeostasis described in neonatal platelets. Both Ca2+ mobilization and Ca2+ influx in response to Thr are decreased in neonatal platelets compared to maternal and control woman platelets. In neonatal platelets, we observed impaired Ca2+ mobilization in response to the PAR-1 agonist (SFLLRN) or by blocking SERCA3 function with tert-butylhydroquinone. Regarding SOCE, the STIM1 regulatory protein, SARAF, was found overexpressed in neonatal platelets, promoting an increase in STIM1/SARAF interaction even under resting conditions. Additionally, higher interaction between SARAF and PDCD61/ALG2 was also observed, reducing SARAF ubiquitination and prolonging its half-life. These results were reproduced by overexpressing SARAF in MEG01 and DAMI cells. Finally, we also observed that pannexin 1 permeability is enhanced in response to Thr in control woman and maternal platelets, but not in neonatal platelets, hence, leading to the deregulation of the Ca2+ entry found in neonatal platelets. Summarizing, we show that in neonatal platelets both Ca2+ accumulation in the intracellular stores and Thr-evoked Ca2+ entry through either capacitative channels or non-selective channels are altered in neonatal platelets, contributing to deregulated Ca2+ homeostasis in neonatal platelets and leading to the altered aggregation observed in these subjects.


Asunto(s)
Proteínas de la Membrana , Trombina , Recién Nacido , Humanos , Trombina/metabolismo , Proteínas de la Membrana/metabolismo , Plaquetas/metabolismo , Homeostasis , Calcio/metabolismo , Señalización del Calcio
2.
Arterioscler Thromb Vasc Biol ; 43(5): e151-e170, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36924231

RESUMEN

BACKGROUND: Altered intracellular Ca2+ homeostasis in neonatal platelets has been previously reported. This study aims to examine the changes in the Ca2+ entry through the store-operated calcium entry (SOCE) mechanism in neonatal platelets. METHODS: Human platelets from either control women, mothers, and neonates were isolated and, following, were fixed after being treated as required. Platelet samples were analyzed by Western blotting, qRT-PCR, and MALDITOF/TOF. Ca2+ homeostasis was also determined. Culture cells were used as surrogated of platelets to overexpress the proteins of interest to reproduce the alterations observed in platelets. RESULTS: Altered TG (thapsigargin)-evoked SOCE, alternative molecular weight form of STIM1 (stromal interaction molecule 1; s-STIM1 [short STIM1 isoform (478 aa)], around 60 kDa) and overexpression of SARAF (SOCE-associated regulatory factor) were found in neonatal platelets as compared to maternal and control women platelets. s-STIM1 may result due to CAPN1 (calpain1)-dependent processing, as confirmed in platelets and MEG01 cells by using calpeptin and overexpressing CAPN1, respectively. In HEK293 (STIM1 and STIM2 [stromal interaction molecule 2] double knockout) cells transfected either with c-STIM1 (canonical STIM1 [685 aa]), s-STIM1 (478), STIM1B (540), and CAPN1 overexpression plasmids, we found s-STIM1 and c-STIM1, except in cells overexpressing s-STIM1 (478) that lacked CAPN1 target residues. These results and the in silico analysis, lead us to conclude that STIM1 is cleaved at Q496 by CAPN1. Ca2+ imaging analysis and coimmunoprecipitation assay using MEG01 and HEK293 cells overexpressing SARAF together with s-STIM1 (478) reported a reduced slow Ca2+-dependent inactivation, so reproducing the Ca2+-homeostasis pattern observed in neonatal platelets. CONCLUSIONS: CAPN1 may cleave STIM1 in neonatal platelets, hence, impairing SARAF coupling after SOCE activation. s-STIM1 may avoid slow Ca2+-dependent inactivation and, subsequently, results in an enhanced TG-evoked SOCE as observed in neonatal platelets.


Asunto(s)
Plaquetas , Calpaína , Proteínas de la Membrana , Molécula de Interacción Estromal 1 , Femenino , Humanos , Recién Nacido , Plaquetas/metabolismo , Calcio/metabolismo , Señalización del Calcio , Calpaína/metabolismo , Células HEK293 , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteína ORAI1/genética , Proteína ORAI1/metabolismo , Molécula de Interacción Estromal 1/genética , Molécula de Interacción Estromal 1/metabolismo
3.
J Cell Physiol ; 238(4): 714-726, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36952615

RESUMEN

Store operated Ca2+ entry (SOCE) is a cornerstone for the maintenance of intracellular Ca2+ homeostasis and the regulation of a variety of cellular functions. SOCE is mediated by STIM and Orai proteins following the activation of inositol 1,4,5-trisphosphate receptors. Then, a reduction of the endoplasmic reticulum intraluminal Ca2+ concentration is sensed by STIM proteins, which undergo a conformational change and activate plasma membrane Ca2+ channels comprised by Orai proteins. STIM1/Orai-mediated Ca2+ signals are finely regulated and modulate the activity of different transcription factors, including certain isoforms of the nuclear factor of activated T-cells, the cAMP-response element binding protein, the nuclear factor κ-light chain-enhancer of activated B cells, c-fos, and c-myc. These transcription factors associate SOCE with a plethora of signaling events and cellular functions. Here we provide an overview of the current knowledge about the role of Orai channels in the regulation of transcription factors through Ca2+ -dependent signaling pathways.


Asunto(s)
Canales de Calcio Activados por la Liberación de Calcio , Señalización del Calcio , Factores de Transcripción , Calcio/metabolismo , Membrana Celular/metabolismo , Proteína ORAI1/metabolismo , Molécula de Interacción Estromal 1/metabolismo , Factores de Transcripción/metabolismo , Canales de Calcio Activados por la Liberación de Calcio/metabolismo
4.
Cell Mol Life Sci ; 79(1): 33, 2022 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-34988680

RESUMEN

The identification of two variants of the canonical pore-forming subunit of the Ca2+ release-activated Ca2+ (CRAC) channel Orai1, Orai1α and Orai1ß, in mammalian cells arises the question whether they exhibit different functional characteristics. Orai1α and Orai1ß differ in the N-terminal 63 amino acids, exclusive of Orai1α, and show different sensitivities to Ca2+-dependent inactivation, as well as distinct ability to form arachidonate-regulated channels. We have evaluated the role of both Orai1 variants in the activation of TRPC1 in HeLa cells. We found that Orai1α and Orai1ß are required for the maintenance of regenerative Ca2+ oscillations, while TRPC1 plays a role in agonist-induced Ca2+ influx but is not essential for Ca2+ oscillations. Using APEX2 proximity labeling, co-immunoprecipitation and the fluorescence of G-GECO1.2 fused to Orai1α our results indicate that agonist stimulation and Ca2+ store depletion enhance Orai1α-TRPC1 interaction. Orai1α is essential for TRPC1 plasma membrane location and activation. Thus, TRPC1 function in HeLa cells depends on Ca2+ influx through Orai1α exclusively.


Asunto(s)
Membrana Celular/metabolismo , Proteína ORAI1/metabolismo , Canales Catiónicos TRPC/metabolismo , Calcio/metabolismo , Cationes , Células HeLa , Humanos , Proteínas Mutantes/metabolismo , Unión Proteica , Molécula de Interacción Estromal 1/metabolismo
5.
Int J Mol Sci ; 24(19)2023 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-37833987

RESUMEN

Hypoxia is considered to be a stressful physiological condition, which may occur during labor and the later stages of pregnancy as a result of, among other reasons, an aged placenta. Therefore, when gestation or labor is prolonged, low oxygen supply to the tissues may last for minutes, and newborns may present breathing problems and may require resuscitation maneuvers. As a result, poor oxygen supply to tissues and to circulating cells may last for longer periods of time, leading to life-threatening conditions. In contrast to the well-known platelet activation that occurs after reperfusion of the tissues due to an ischemia/reperfusion episode, platelet alterations in response to reduced oxygen exposition following labor have been less frequently investigated. Newborns overcome temporal hypoxic conditions by changing their organ functions or by adaptation of the intracellular molecular pathways. In the present review, we aim to analyze the main platelet modifications that appear at the protein level during hypoxia in order to highlight new platelet markers linked to complications arising from temporal hypoxic conditions during labor. Thus, we demonstrate that hypoxia modifies the expression and activity of hypoxic-response proteins (HRPs), including hypoxia-induced factor (HIF-1), endoplasmic reticulum oxidase 1 (Ero1), and carbonic anhydrase (CIX). Finally, we provide updates on research related to the regulation of platelet function due to HRP activation, as well as the role of HRPs in intracellular Ca2+ homeostasis.


Asunto(s)
Anhidrasas Carbónicas , Trabajo de Parto , Recién Nacido , Embarazo , Femenino , Humanos , Anciano , Hipoxia/metabolismo , Oxígeno/metabolismo , Placenta/metabolismo , Anhidrasas Carbónicas/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo
6.
Int J Mol Sci ; 23(23)2022 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-36498894

RESUMEN

Orai1, the first identified member of the Orai protein family, is ubiquitously expressed in the animal kingdom. Orai1 was initially characterized as the channel responsible for the store-operated calcium entry (SOCE), a major mechanism that allows cytosolic calcium concentration increments upon receptor-mediated IP3 generation, which results in intracellular Ca2+ store depletion. Furthermore, current evidence supports that abnormal Orai1 expression or function underlies several disorders. Orai1 is, together with STIM1, the key element of SOCE, conducting the Ca2+ release-activated Ca2+ (CRAC) current and, in association with TRPC1, the store-operated Ca2+ (SOC) current. Additionally, Orai1 is involved in non-capacitative pathways, as the arachidonate-regulated or LTC4-regulated Ca2+ channel (ARC/LRC), store-independent Ca2+ influx activated by the secretory pathway Ca2+-ATPase (SPCA2) and the small conductance Ca2+-activated K+ channel 3 (SK3). Furthermore, Orai1 possesses two variants, Orai1α and Orai1ß, the latter lacking 63 amino acids in the N-terminus as compared to the full-length Orai1α form, which confers distinct features to each variant. Here, we review the current knowledge about the differences between Orai1α and Orai1ß, the implications of the Ca2+ signals triggered by each variant, and their downstream modulatory effect within the cell.


Asunto(s)
Canales de Calcio , Calcio , Animales , Proteína ORAI1/genética , Proteína ORAI1/metabolismo , Calcio/metabolismo , Canales de Calcio/genética , Canales de Calcio/metabolismo , Canales Catiónicos TRPC/metabolismo , Molécula de Interacción Estromal 1/genética , Molécula de Interacción Estromal 1/metabolismo , Transporte Iónico , Señalización del Calcio
7.
Cell Physiol Biochem ; 51(3): 1164-1178, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30481768

RESUMEN

BACKGROUND/AIMS: STIM1 and Orai1 are the key components of store-operated Ca2+ entry (SOCE). Among the proteins involved in the regulation of SOCE, SARAF prevents spontaneous activation of SOCE and modulates STIM1 function. METHODS: Cytosolic Ca2+ mobilization was estimated in fura-2-loaded cells using an epifluorescence inverted microscope. STIM1 interaction with Orai1, EFHB (EF-hand domain family member B, also known as CFAP21) and SARAF was detected by immunoprecipitation followed by Western blotting using specific antibodies. The involvement of EFHB in the translocation of NFAT to the nucleus was detected by confocal microscopy. RESULTS: Here, we report the identification of EFHB as a new SOCE regulator. EFHB interacts with STIM1 upon store depletion and dissociates through a Ca2+-dependent mechanism. RNAi-mediated silencing as well as overexpression studies revealed that EFHB plays a relevant role in the interaction of STIM1 and Orai1 upon store depletion, the activation of SOCE and NFAT translocation from the cytosol to the nucleus. Silencing EFHB expression abolished the dissociation of SARAF from STIM1, which indicates that EFHB might play an important role in the dynamic interaction between both proteins, which is relevant for the activation of Orai1 channels upon Ca2+ store depletion and their subsequent modulation via slow Ca2+-dependent inactivation. CONCLUSION: Our results indicate that EFHB is a new SOCE regulator that modulates STIM1-SARAF interaction.


Asunto(s)
Calcio/metabolismo , Proteínas Sensoras del Calcio Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Mapas de Interacción de Proteínas , Molécula de Interacción Estromal 1/metabolismo , Citosol/metabolismo , Motivos EF Hand , Células HEK293 , Células HeLa , Humanos
8.
J Physiol ; 595(10): 3111-3128, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28087881

RESUMEN

An endoplasmic reticulum (ER)-resident protein that regulates cytosolic and ER free-Ca2+ concentration by induction of store-operated calcium entry: that is the original definition of STIM2 and its function. While its activity strongly depends on the amount of calcium stored in the ER, its function goes further, to intracellular signalling and gene expression. Initially under-studied owing to the prominent function of STIM1, STIM2 came to be regarded as vital in mice, gradually emerging as an important player in the nervous system, and cooperating with STIM1 in the immune system. STIM2 has also been proposed as a relevant player in pathological conditions related to ageing, Alzheimer's and Huntington's diseases, autoimmune disorders and cancer. The discovery of additional functions, together with new splicing forms with opposite roles, has clarified existing controversies about STIM2 function in SOCE. With STIM2 being essential for life, but apparently not for development, newly available data demonstrate a complex and still intriguing behaviour that this review summarizes, updating current knowledge of STIM2 function.


Asunto(s)
Molécula de Interacción Estromal 2 , Animales , Sistema Cardiovascular/metabolismo , Humanos , Neoplasias/metabolismo , Sistema Nervioso/metabolismo , Molécula de Interacción Estromal 2/química , Molécula de Interacción Estromal 2/genética , Molécula de Interacción Estromal 2/inmunología , Molécula de Interacción Estromal 2/metabolismo
9.
Biochim Biophys Acta ; 1853(10 Pt A): 2684-96, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26239116

RESUMEN

Non-capacitative calcium entry (NCCE) contributes to cell activation in response to the occupation of G protein-coupled membrane receptors. Thrombin administration to platelets evokes the synthesis of diacylglycerol downstream of PAR receptor activation. Diacylglycerol evokes NCCE through activating TRPC3 and TRPC6 in human platelets. Although it is known that immunophilins interact with TRPCs, the role of immunophilins in the regulation of NCCE remains unknown. Platelet incubation with FK506, an immunophilin antagonist, reduced OAG-evoked NCCE in a concentration-dependent manner, an effect that was independent on the inactivation of calcineurin (CaN). FK506 was unable to reduce NCCE evoked by OAG in platelets from TRPC6-/- mice. In HEK-293 cells overexpressing TRPC6, currents through TRPC6 were altered in the presence of FK506. We have found interaction between FKBP38 and other FKBPs, like FKBP25, FKBP12, and FKBP52 that were not affected by FK506, as well as with calmodulin (CaM). FK506 modified the pattern of association between FKBP25 and TRPCs as well as impaired OAG-evoked TRPC3 and TRPC6 coupling in both human and mouse platelets. By performing biotinylation experiments we have elucidated that FKBP25 and FKBP38 might be found at different cellular location, the plasma membrane and the already described intracellular locations. Finally, FKBP25 and FKBP38 silencing significantly inhibits OAG-evoked NCCE in MEG-01 and HEK293 cells, while overexpression of FKBP38 does not modify NCCE in HEK293 cells. All together, these findings provide strong evidence for a role of immunophilins, including FKBP25 and FKBP38, in NCCE mediated by TRPC6.


Asunto(s)
Plaquetas/metabolismo , Inmunosupresores/farmacología , Canales Catiónicos TRPC/metabolismo , Animales , Plaquetas/citología , Calcio , Células HEK293 , Humanos , Ratones , Canales Catiónicos TRPC/genética , Canal Catiónico TRPC6
10.
Adv Exp Med Biol ; 898: 157-81, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27161229

RESUMEN

Agonist-induced changes in cytosolic Ca(2+) concentration ([Ca(2+)]c) are central events in platelet physiology. A major mechanism supporting agonist-induced Ca(2+) signals is store-operated Ca(2+) entry (SOCE), where the Ca(2+) sensor STIM1 and the channels of the Orai family, as well as TRPC members are the key elements. STIM1-dependent SOCE plays a major role in collagen-stimulated Ca(2+) signaling, phosphatidylserine exposure and thrombin generation. Furthermore, studies involving Orai1 gain-of-function mutants and platelets from Orai1-deficient mice have revealed the importance of this channel in thrombosis and hemostasis to those found in STIM1-deficient mice indicating that SOCE might play a prominent role in thrombus formation. Moreover, increase in TRPC6 expression might lead to thrombosis in humans. The role of STIM1, Orai1 and TRPCs, and thus SOCE, in thrombus formation, suggests that therapies directed against SOCE and targeting these molecules during cardiovascular and cerebrovascular events could significantly improve traditional anti-thrombotic treatments.


Asunto(s)
Plaquetas/fisiología , Canales de Calcio/fisiología , Canales Catiónicos TRPC/fisiología , Animales , Canales de Calcio/genética , Ratones , Ratones Noqueados , Molécula de Interacción Estromal 1
11.
Biochim Biophys Acta ; 1838(2): 658-64, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23333715

RESUMEN

Transient receptor potential (TRP) channels are six transmembrane-spanning proteins, with variable selectivity for cations, that play a relevant role in intracellular Ca(2+) homeostasis. There is a large body of evidence that shows association of TRP channels with the actin cytoskeleton or even the microtubules and demonstrating the functional importance of this interaction for TRP channel function. Conversely, cation currents through TRP channels have also been found to modulate cytoskeleton rearrangements. The interplay between TRP channels and the cytoskeleton has been demonstrated to be essential for full activation of a variety of cellular functions. Furthermore, TRP channels have been reported to take part of macromolecular complexes including different signal transduction proteins. Scaffolding proteins play a relevant role in the association of TRP proteins with other signaling molecules into specific microdomains. Especially relevant are the roles of the Homer family members for the regulation of TRPC channel gating in mammals and INAD in the modulation of Drosophila TRP channels. This article is part of a Special Issue entitled: Reciprocal influences between cell cytoskeleton and membrane channels, receptors and transporters. Guest Editor: Jean Claude Hervé.


Asunto(s)
Citoesqueleto/metabolismo , Transducción de Señal , Canales de Potencial de Receptor Transitorio/metabolismo , Animales , Humanos
12.
Biochim Biophys Acta ; 1843(4): 789-96, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24462772

RESUMEN

Cytosolic-free Ca(2+) plays a crucial role in blood platelet function and is essential for thrombosis and hemostasis. Therefore, cytosolic-free Ca(2+) concentration is tightly regulated in this cell. TRPC6 is expressed in platelets, and an important role for this Ca(2+) channel in Ca(2+) homeostasis has been reported in other cell types. The aim of this work is to study the function of TRPC6 in platelet Ca(2+) homeostasis. The absence of TRPC6 resulted in an 18.73% decreased basal [Ca(2+)]c in resting platelets as compared to control cells. Further analysis confirmed a similar Ca(2+) accumulation in wild-type and TRPC6-deficient mice; however, passive Ca(2+) leak rates from agonist-sensitive intracellular stores were significantly decreased in TRPC6-deficient platelets. Biotinylation studies indicated the presence of an intracellular TRPC6 population, and subcellular fractionation indicated their presence on endoplasmic reticulum membranes. Moreover, the presence of intracellular calcium release in platelets stimulated with 1-oleoyl-2-acetyl-sn-glycerol further suggested a functional TRPC6 population located on the intracellular membranes surrounding calcium stores. However, coimmunoprecipitation assay confirmed the absence of STIM1-TRPC6 interactions in resting conditions. This findings together with the absence of extracellular Mn(2+) entry in resting wild-type platelets indicate that the plasma membrane TRPC6 fraction does not play a significant role in the maintenance of basal [Ca(2+)]c in mouse platelets. Our results suggest an active participation of the intracellular TRPC6 fraction as a regulator of basal [Ca(2+)]c, controlling the passive Ca(2+) leak rate from agonist-sensitive intracellular Ca(2+) stores in resting platelets.


Asunto(s)
Plaquetas/metabolismo , Calcio/metabolismo , Homeostasis , Canales Catiónicos TRPC/metabolismo , Animales , Citosol/metabolismo , Diglicéridos/farmacología , Membranas Intracelulares/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Proteínas de Neoplasias/metabolismo , Molécula de Interacción Estromal 1 , Canales Catiónicos TRPC/genética , Canal Catiónico TRPC6
13.
Biochim Biophys Acta ; 1833(3): 652-62, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23228564

RESUMEN

Immunophilins are FK506-binding proteins that have been involved in the regulation of calcium homeostasis, either by modulating Ca(2+) channels located in the plasma membrane or in the rough endoplasmic reticulum (RE). We have investigated whether immunophilins would participate in the regulation of stored-operated Ca(2+) entry (SOCE) in human platelets and MEG 01. Both cell types were loaded with fura-2 for determining cytosolic calcium concentration changes ([Ca(2+)](c)), or stimulated and fixed to evaluate the protein interaction profile by performing immunoprecipitation and western blotting. We have found that incubation of platelets with FK506 increases Ca(2+) mobilization. Thapsigargin (TG)-evoked, Thr-evoked SOCE and TG-evoked Mn(2+) entry resulted in significant reduction by treatment of platelets with immunophilin antagonists. We confirmed by immunoprecipitation that immunophilins interact with transient receptor potential channel 1 (TRPC1) and Orai1 in human platelets. FK506 and rapamycin reduced the association between TRPC1 and Orai1 with FK506 binding protein (52) (FKBP52) in human platelets, and between TRPC1 and the type II IP(3)R, which association is known to be crucial for the maintenance of SOCE in human platelets. FKBP52 role in SOCE activation was confirmed by silencing FKBP52 using SiRNA FKBP52 in MEG 01 as demonstrated by single cell configuration imaging technique. TRPC1 silencing and depletion of cell of TRPC1 and FKBP52 simultaneously, impair activation of SOCE evoked by TG in MEG 01. Finally, in MEG 01 incubated with FK506 we observed a reduction in TRPC1/FKBP52 coupling, and similarly, FKBP52 silencing reduced the association between IP3R type II and TRPC1 during SOCE. All together, these results demonstrate that immunophilins participate in the regulation of SOCE in human platelets.


Asunto(s)
Plaquetas/metabolismo , Canales de Calcio/metabolismo , Calcio/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Células Progenitoras de Megacariocitos/metabolismo , Canales Catiónicos TRPC/metabolismo , Proteínas de Unión a Tacrolimus/metabolismo , Plaquetas/efectos de los fármacos , Western Blotting , Señalización del Calcio/efectos de los fármacos , Células Cultivadas , Inhibidores Enzimáticos/farmacología , Fura-2/metabolismo , Humanos , Inmunofilinas/farmacología , Inmunoprecipitación , Inmunosupresores/farmacología , Transporte Iónico/efectos de los fármacos , Células Progenitoras de Megacariocitos/citología , Células Progenitoras de Megacariocitos/efectos de los fármacos , Proteína ORAI1 , ARN Interferente Pequeño/genética , Canales Catiónicos TRPC/antagonistas & inhibidores , Canales Catiónicos TRPC/genética , Tacrolimus/farmacología , Proteínas de Unión a Tacrolimus/antagonistas & inhibidores , Proteínas de Unión a Tacrolimus/genética , Tapsigargina/farmacología
14.
Blood Cells Mol Dis ; 52(2-3): 108-15, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24074949

RESUMEN

The canonical transient receptor potential-6 (TRPC6) is a receptor-activated non-selective Ca(2+) channel regulated by a variety of modulators such as diacylglycerol, Ca(2+)/calmodulin or phosphorylation. The present study is aimed to investigate whether different situations, such as acidic pH, exposure to reactive oxygen species (ROS) or hypoxic-like conditions modulate TRPC6 channel function. Here we show normal aggregation and Ca(2+) mobilization stimulated by thrombin in TRPC6 KO platelets; however, OAG (1-oleoyl-2-acetyl-sn-glycerol)-evoked Ca(2+) entry was attenuated in the absence of TRPC6. Exposure of mouse platelets to acidic pH resulted in abolishment of thrombin-evoked aggregation and attenuated platelet aggregation induced by thapsigargin (TG) or OAG. Both OAG-induced Ca(2+) entry and platelet aggregation were greatly attenuated in cells expressing TRPC6 channels. Exposure of platelets to H2O2 or deferoxamine did not clearly alter thrombin, TG or OAG-induced platelet aggregation. Our results indicate that TRPC6 is sensitive to acidic pH but not to exposure to ROS or hypoxic-like conditions, which might be involved in the pathogenesis of the altered platelet responsiveness to DAG-generating agonists in disorders associated to acidic pH.


Asunto(s)
Plaquetas/fisiología , Espacio Extracelular/metabolismo , Canales Catiónicos TRPC/metabolismo , Animales , Plaquetas/efectos de los fármacos , Calcio/metabolismo , Deferoxamina/farmacología , Peróxido de Hidrógeno/farmacología , Concentración de Iones de Hidrógeno , Ratones , Ratones Noqueados , Agregación Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/genética , Canales Catiónicos TRPC/genética , Canal Catiónico TRPC6 , Trombina/farmacología
15.
J Cell Mol Med ; 17(5): 636-47, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23577651

RESUMEN

The use of the mammal target of rapamycin (mTOR) inhibitors has been consolidated as the therapy of election for preventing graft rejection in kidney transplant patients, despite their immunosuppressive activity is less strong than anti-calcineurin agents like tacrolimus and cyclosporine A. Furthermore, as mTOR is widely expressed, rapamycin (a macrolide antibiotic produced by Streptomyces hygroscopicus) is recommended in patients presenting neoplasia due to its antiproliferative actions. Hence, we have investigated whether rapamycin presents side effects in the physiology of other cell types different from leucocytes, such as platelets. Blood samples were drawn from healthy volunteers and kidney transplant patients long-term medicated with rapamycin: sirolimus and everolimus. Platelets were either loaded with fura-2 or directly stimulated, and immunoassayed or fixed with Laemmli's buffer to perform the subsequent analysis of platelet physiology. Our results indicate that rapamycin evokes a biphasic time-dependent alteration in calcium homeostasis and function in platelets from kidney transplant patients under rapamycin regime, as demonstrated by the reduction in granule secretion observed and subsequent impairment of platelet aggregation in these patients compared with healthy volunteers. Platelet count was also reduced in these patients, thus 41% of patients presented thrombocytopenia. All together our results show that long-term administration of rapamycin to kidney transplant patients evokes alteration in platelet function.


Asunto(s)
Plaquetas/patología , Calcio/metabolismo , Homeostasis/efectos de los fármacos , Trasplante de Riñón , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Adulto , Anciano , Plaquetas/efectos de los fármacos , Demografía , Activación Enzimática/efectos de los fármacos , Everolimus , Femenino , Humanos , Masculino , Persona de Mediana Edad , Fosforilación/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Sirolimus/administración & dosificación , Sirolimus/análogos & derivados , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/metabolismo , Factores de Tiempo
16.
Biochim Biophys Acta ; 1823(10): 1976-83, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23077736

RESUMEN

Two-pore channels (TCPS) are NAADP-sensitive receptor channels that conduct Ca(2+) efflux from the intracellular stores. Discharge of the internal Ca(2+) pools results in the activation of store-operated Ca(2+) entry (SOCE); however, the role of TPCs in the modulation of SOCE remains unexplored. Mammalian cells express three TPCs: TPC1, TPC2 and TPC3, a pseudogene in humans. Here we report that MEG01 and HEK293 cells endogenously express TPC1 and TPC2. Silencing TPC2 expression results in attenuation of the rate and extent of thapsigargin (TG)-evoked SOCE both in MEG01 and HEK293 cells, without having any effect on the ability of cells to accumulate Ca(2+) into the TG-sensitive stores. Similarly, silencing of native TPC2 expression reduced thrombin-induced Ca(2+) entry both in MEG01 and HEK293 cells. Biotinylation analysis revealed that TPC1 and TPC2 are expressed in internal membranes. Finally, co-immunoprecipitation experiments indicated that endogenously expressed TPC2, but not TPC1, assoicates STIM1 and Orai1, but not with TRPC1, in MEG01 cells with depleted intracellular Ca(2+) stores, but not in resting cells. These results provide strong evidence for modulation of SOCE by TPC2 involving de novo association between TPC2 and STIM1, as well as Orai1, in human cells.


Asunto(s)
Canales de Calcio/metabolismo , Calcio/metabolismo , Canales de Calcio/genética , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Silenciador del Gen/efectos de los fármacos , Células HEK293 , Humanos , Inmunoprecipitación , Proteínas de la Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Proteína ORAI1 , ARN Mensajero/genética , ARN Mensajero/metabolismo , Molécula de Interacción Estromal 1 , Tapsigargina/farmacología , Trombina/farmacología
17.
Biochim Biophys Acta ; 1823(8): 1242-51, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22640869

RESUMEN

Discharge of the intracellular Ca(2+) stores activates Ca(2+) entry through store-operated channels (SOCs). Since the recent identification of STIM1 and STIM2, as well as the Orai1 homologs, Orai2 and Orai3, the protein complexes involved in Ca(2+) signaling needs re-evaluation in native cells. Using real time PCR combined with Western blotting we have found the expression of the three Orai isoforms, STIM1, STIM2 and different TRPCs in human platelets. Depletion of the intracellular Ca(2+) stores with thapsigargin, independently of changes in cytosolic Ca(2+) concentration, enhanced the formation of a signaling complex involving STIM1, STIM2, Orai1, Orai2 and TRPC1. Furthermore, platelet treatment with the dyacylglicerol analog 1-oleoyl-2-acetyl-sn-glycerol (OAG) resulted in specific association of Orai3 with TRPC3. Treatment of platelets with arachidonic acid enhanced the association between Orai1 and Orai3 in human platelets and overexpression of Orai1 and Orai3 in HEK293 cells increased arachidonic acid-induced Ca(2+) entry. These results indicate that Ca(2+) store depletion results in the formation of exclusive signaling complexes involving STIM proteins, as well as Orai1, Orai2 and TRPC1, but not Orai3, which seems to be involved in non-capacitative Ca(2+) influx in human platelets.


Asunto(s)
Plaquetas/metabolismo , Canales de Calcio/metabolismo , Señalización del Calcio , Proteínas de la Membrana/metabolismo , Ácido Araquidónico/farmacología , Ácido Araquidónico/fisiología , Canales de Calcio/genética , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Diglicéridos/farmacología , Diglicéridos/fisiología , Expresión Génica , Células HEK293 , Humanos , Inmunoprecipitación , Proteínas de la Membrana/genética , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteína ORAI1 , Proteína ORAI2 , Unión Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Molécula de Interacción Estromal 1 , Molécula de Interacción Estromal 2 , Canales Catiónicos TRPC/genética , Canales Catiónicos TRPC/metabolismo , Canal Catiónico TRPC6
18.
IUBMB Life ; 65(6): 497-504, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23554128

RESUMEN

The Homer family of proteins consists of three adaptor proteins, Homer1, Homer2 and Homer3, each with various isoforms. Homer1 family presents an EVH1 domain, a coiled coil domain and two leucine zipper domains. Homer proteins regulate a number of Ca2+-handling proteins, including transient receptor potential channels and other Ca2+-permeable channels, ionotropic and metabotropic glutamate receptors, shank scaffolding proteins or endoplasmic reticulum Ca2+ release channels. This review article focuses on the association of Homer 1 proteins with Ca2+-handling proteins and their role on intracellular Ca2+-homeostasis.


Asunto(s)
Calcio/metabolismo , Proteínas Portadoras/fisiología , Animales , Canales de Calcio/metabolismo , ATPasas Transportadoras de Calcio/metabolismo , Acoplamiento Excitación-Contracción , Proteínas de Andamiaje Homer , Humanos , Receptores de Glutamato/metabolismo
19.
Biomolecules ; 13(9)2023 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-37759684

RESUMEN

The STIM family of proteins plays a crucial role in a plethora of cellular functions through the regulation of store-operated Ca2+ entry (SOCE) and, thus, intracellular calcium homeostasis. The two members of the mammalian STIM family, STIM1 and STIM2, are transmembrane proteins that act as Ca2+ sensors in the endoplasmic reticulum (ER) and, upon Ca2+ store discharge, interact with and activate the Orai/CRACs in the plasma membrane. Dysregulation of Ca2+ signaling leads to the pathogenesis of a variety of human diseases, including neurodegenerative disorders, cardiovascular diseases, cancer, and immune disorders. Therefore, understanding the mechanisms underlying Ca2+ signaling pathways is crucial for developing therapeutic strategies targeting these diseases. This review focuses on several rare conditions associated with STIM1 mutations that lead to either gain- or loss-of-function, characterized by myopathy, hematological and immunological disorders, among others, and due to abnormal activation of CRACs. In addition, we summarize the current evidence concerning STIM2 allele duplication and deletion associated with language, intellectual, and developmental delay, recurrent pulmonary infections, microcephaly, facial dimorphism, limb anomalies, hypogonadism, and congenital heart defects.


Asunto(s)
Líquidos Corporales , Enfermedades Cardiovasculares , Animales , Humanos , Alelos , Membrana Celular , Retículo Endoplásmico , Mamíferos
20.
J Biol Chem ; 286(14): 12257-70, 2011 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-21321120

RESUMEN

Mammalian cells accumulate Ca2+ into agonist-sensitive acidic organelles, vesicles that possess a vacuolar proton-ATPase. Acidic Ca2+ stores include secretory granules and lysosome-related organelles. Current evidence clearly indicates that acidic Ca2+ stores participate in cell signaling and function, including the activation of store-operated Ca2+ entry in human platelets upon depletion of the acidic stores, although the mechanism underlying the activation of store-operated Ca2+ entry controlled by the acidic stores remains unclear. STIM1 has been presented as the endoplasmic reticulum Ca2+ sensor, but its role sensing intraluminal Ca2+ concentration in the acidic stores has not been investigated. Here we report that STIM1 and STIM2 are expressed in the lysosome-related organelles and dense granules in human platelets isolated by immunomagnetic sorting. Depletion of the acidic Ca2+ stores using the specific vacuolar proton-ATPase inhibitor, bafilomycin A1, enhanced the association between STIM1 and STIM2 as well as between these proteins and the plasma membrane channel Orai1. Depletion of the acidic Ca2+ stores also induces time-dependent co-immunoprecipitation of STIM1 with the TRPC proteins hTRPC1 and hTRPC6, as well as between Orai1 and both TRPC proteins. In addition, bafilomycin A1 enhanced the association between STIM2 and SERCA3. These findings demonstrate the location of STIM1 and STIM2 in the acidic Ca2+ stores and their association with Ca2+ channels and ATPases upon acidic stores discharge.


Asunto(s)
Plaquetas/metabolismo , Moléculas de Adhesión Celular/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Western Blotting , Calcio/metabolismo , Canales de Calcio , Moléculas de Adhesión Celular/genética , Células Cultivadas , Electroporación , Técnica del Anticuerpo Fluorescente , Humanos , Inmunoprecipitación , Macrólidos/farmacología , Proteínas de la Membrana/genética , Proteínas de Neoplasias/genética , Proteína ORAI1 , Unión Proteica/efectos de los fármacos , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Molécula de Interacción Estromal 1 , Molécula de Interacción Estromal 2
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA