Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Adv Exp Med Biol ; 1131: 445-469, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31646520

RESUMEN

Store-operated Ca2+ entry (SOCE) is a ubiquitous mechanism for Ca2+ influx in mammalian cells with important physiological implications. Since the discovery of SOCE more than three decades ago, the mechanism that communicates the information about the amount of Ca2+ accumulated in the intracellular Ca2+ stores to the plasma membrane channels and the nature of these channels have been matters of intense investigation and debate. The stromal interaction molecule-1 (STIM1) has been identified as the Ca2+ sensor of the intracellular Ca2+ compartments that activates the store-operated channels. STIM1 regulates two types of store-dependent channels: the Ca2+ release-activated Ca2+ (CRAC) channels, formed by Orai1 subunits, that conduct the highly Ca2+ selective current I CRAC and the cation permeable store-operated Ca2+ (SOC) channels, which consist of Orai1 and TRPC1 proteins and conduct the non-selective current I SOC. While the crystal structure of Drosophila CRAC channel has already been solved, the architecture of the SOC channels still remains unclear. The dynamic interaction of STIM1 with the store-operated channels is modulated by a number of proteins that either support the formation of the functional STIM1-channel complex or protect the cell against Ca2+ overload.


Asunto(s)
Canales de Calcio , Calcio , Transporte Iónico , Animales , Calcio/metabolismo , Canales de Calcio/genética , Canales de Calcio/metabolismo , Señalización del Calcio , Transporte Iónico/genética , Molécula de Interacción Estromal 1/metabolismo
2.
Biochim Biophys Acta Mol Cell Res ; 1865(3): 463-469, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29223474

RESUMEN

Store-operated Ca2+ entry (SOCE) is a functionally relevant mechanism for Ca2+ influx present in electrically excitable and non-excitable cells. Regulation of Ca2+ entry through store-operated channels is essential to maintain an appropriate intracellular Ca2+ homeostasis and prevent cell damage. Calcium-release activated channels exhibit Ca2+-dependent inactivation mediated by two temporally separated mechanisms: fast Ca2+-dependent inactivation takes effect in the order of milliseconds and involves the interaction of Ca2+ with residues in the channel pore while slow Ca2+-dependent inactivation (SCDI) develops over tens of seconds, requires a global rise in [Ca2+]cyt and is a mechanism regulated by mitochondria. Recent studies have provided evidence that the protein SARAF (SOCE-associated regulatory factor) is involved in the mechanism underlying SCDI of Orai1. SARAF is an endoplasmic reticulum (ER) membrane protein that associates with STIM1 and translocate to plasma membrane-ER junctions in a STIM1-dependent manner upon store depletion to modulate SOCE. SCDI mediated by SARAF depends on the location of the STIM1-Orai1 complex within a PI(4,5)P2-rich microdomain. SARAF also interacts with Orai1 and TRPC1 in cells endogenously expressing STIM1 and cells with a low STIM1 expression and modulates channel function. This review focuses on the modulation by SARAF of SOCE and other forms of Ca2+ influx mediated by Orai1 and TRPC1 in order to provide spatio-temporally regulated Ca2+ signals.


Asunto(s)
Señalización del Calcio/genética , Calcio/metabolismo , Retículo Endoplásmico/genética , Proteínas de la Membrana/genética , Canales de Calcio/genética , Membrana Celular/genética , Humanos , Proteínas Sensoras del Calcio Intracelular , Mitocondrias/genética , Proteína ORAI1/genética , Molécula de Interacción Estromal 1/genética , Canales Catiónicos TRPC/genética
3.
Arterioscler Thromb Vasc Biol ; 38(2): 386-397, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29284605

RESUMEN

OBJECTIVE: Here, we provide evidence for the role of FLNA (filamin A) in the modulation of store-operated calcium entry (SOCE). APPROACH AND RESULTS: SOCE is a major mechanism for calcium influx controlled by the intracellular Ca2+ stores. On store depletion, the endoplasmic reticulum calcium sensor STIM1 (stromal interaction molecule 1) redistributes into puncta at endoplasmic reticulum/plasma membrane junctions, a process supported by the cytoskeleton, where it interacts with the calcium channels; however, the mechanism for fine-tuning SOCE is not completely understood. Our results demonstrate that STIM1 interacts with FLNA on calcium store depletion in human platelets. The interaction is dependent on the phosphorylation of FLNA at Ser2152 by the cAMP-dependent protein kinase. Impairment of FLNA phosphorylation and knockdown of FLNA expression using siRNA increased SOCE in platelets. Similarly, SOCE was significantly greater in FLNA-deficient melanoma M2 cells than in the FLNA-expressing M2 subclone A7. Expression of FLNA in M2 cells attenuated SOCE, an effect prevented when the cells were transfected with the nonphosphorylatable FLNA S2152A mutant. Transfection of M2 cells with the STIM1(K684,685E) mutant reduced the STIM1-FLNA interaction. In platelets, attenuation of FLNA expression using siRNA resulted in enhanced association of STIM1 with the cytoskeleton, greater STIM1-Orai1 interaction, and SOCE. Introduction of an anti-FLNA (2597-2647) antibody attenuated the STIM1-FLNA interaction and enhanced thrombin-induced platelet aggregation. CONCLUSIONS: Our results indicate that FLNA modulates SOCE and then the correct platelet function, by fine-tuning the distribution of STIM1 in the cytoskeleton and the interaction with Orai1 channels.


Asunto(s)
Plaquetas/metabolismo , Señalización del Calcio , Calcio/metabolismo , Filaminas/metabolismo , Proteínas de Neoplasias/metabolismo , Proteína ORAI1/metabolismo , Molécula de Interacción Estromal 1/metabolismo , Línea Celular Tumoral , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Citoesqueleto/metabolismo , Filaminas/genética , Humanos , Activación del Canal Iónico , Melanoma/genética , Melanoma/metabolismo , Proteínas de Neoplasias/genética , Proteína ORAI1/genética , Fosforilación , Agregación Plaquetaria , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Serina , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/metabolismo , Molécula de Interacción Estromal 1/genética
4.
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
5.
J Biol Chem ; 291(13): 6982-8, 2016 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-26817842

RESUMEN

The store-operated Ca(2+)entry-associated regulatory factor (SARAF) has recently been identified as a STIM1 regulatory protein that facilitates slow Ca(2+)-dependent inactivation of store-operated Ca(2+)entry (SOCE). Both the store-operated channels and the store-independent arachidonate-regulated Ca(2+)(ARC) channels are regulated by STIM1. In the present study, we show that, in addition to its location in the endoplasmic reticulum, SARAF is constitutively expressed in the plasma membrane, where it can interact with plasma membrane (PM)-resident ARC forming subunits in the neuroblastoma cell line SH-SY5Y. Using siRNA-based and overexpression approaches we report that SARAF negatively regulates store-independent Ca(2+)entry via the ARC channels. Arachidonic acid (AA) increases the association of PM-resident SARAF with Orai1. Finally, our results indicate that SARAF modulates the ability of AA to promote cell survival in neuroblastoma cells. In addition to revealing new insight into the biology of ARC channels in neuroblastoma cells, these findings provide evidence for an unprecedented location of SARAF in the plasma membrane.


Asunto(s)
Ácido Araquidónico/farmacología , Membrana Celular/efectos de los fármacos , Retículo Endoplásmico/efectos de los fármacos , Regulación de la Expresión Génica , Proteínas de la Membrana/metabolismo , Neuronas/efectos de los fármacos , Ácido Araquidónico/metabolismo , Calcio/metabolismo , Canales de Calcio/genética , Canales de Calcio/metabolismo , Línea Celular Tumoral , Membrana Celular/metabolismo , Supervivencia Celular/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Humanos , Proteínas Sensoras del Calcio Intracelular , Transporte Iónico , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neuronas/citología , Neuronas/metabolismo , Proteína ORAI1 , Unión Proteica , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Molécula de Interacción Estromal 1
6.
Biochim Biophys Acta ; 1863(8): 2037-43, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27130253

RESUMEN

Three decades ago, store-operated Ca(2+) entry (SOCE) was identified as a unique mechanism for Ca(2+) entry through plasma membrane (PM) Ca(2+)-permeable channels modulated by the intracellular Ca(2+) stores, mainly the endoplasmic reticulum (ER). Extensive analysis of the communication between the ER and the PM leads to the identification of the protein STIM1 as the ER-Ca(2+) sensor that gates the Ca(2+) channels in the PM. Further analysis on the biophysical, electrophysiological and biochemical properties of STIM1-dependent Ca(2+) channels has revealed the presence of a highly Ca(2+)-selective channel termed Ca(2+) release-activated Ca(2+) channel (CRAC), consisting of Orai1 subunits, and non-selective cation channels named store-operated channels (SOC), including both Orai1 and TRPC channel subunits. Since the identification of the key elements of CRAC and SOC channels a number of intracellular modulators have been reported to play essential roles in the stabilization of STIM-Orai interactions, collaboration with STIM1 conformational changes or mediating slow Ca(2+)-dependent inactivation. Here, we review our current understanding of some of the key modulators of STIM1-Orai1 interaction, including the proteins CRACR2A, STIMATE, SARAF, septins, golli and ORMDL3.


Asunto(s)
Canales de Calcio/metabolismo , Señalización del Calcio/fisiología , Calcio/metabolismo , Animales , Canales de Calcio Activados por la Liberación de Calcio/metabolismo , Retículo Endoplásmico/metabolismo , Humanos , Proteínas Sensoras del Calcio Intracelular , Proteínas de la Membrana/fisiología , Modelos Biológicos , Proteína ORAI1/fisiología , Conformación Proteica , Subunidades de Proteína , Molécula de Interacción Estromal 1/fisiología , Canales Catiónicos TRPC/metabolismo
7.
Biochem J ; 473(20): 3581-3595, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-27506849

RESUMEN

Canonical transient receptor potential-1 (TRPC1) is an almost ubiquitously expressed channel that plays a relevant role in cell function. As other TRPC members, TRPC1 forms receptor-operated cation channels that exhibit both STIM1-dependent and store-independent behaviour. The STIM1 inhibitor SARAF (for store-operated Ca2+ entry (SOCE)-associated regulatory factor) modulates SOCE by interaction with the STIM1 region responsible for Orai1 activation (SOAR). Furthermore, SARAF modulates Ca2+ entry through the arachidonate-regulated Ca2+ (ARC) channels, consisting of Orai1 and Orai3 heteropentamers and plasma membrane-resident STIM1. While a role for STIM1-Orai1-mediated signals has been demonstrated, the possible role of SARAF in TRPC1 function remains unknown. Here, we provide evidence for the interaction of SARAF with TRPC1, independently of STIM1 both in STIM1-deficient NG115-401L cells and SH-SY5Y cells endogenously expressing STIM1. Silencing of SARAF expression in STIM1-deficient cells demonstrated that SARAF plays a negative regulatory role in TRPC1-mediated Ca2+ entry. The interaction of SARAF with TRPC1 in STIM1-deficient cells, as well as with the TRPC1 pool not associated with STIM1 in STIM1-expressing cells was enhanced by stimulation with the physiological agonist ATP. In contrast with TRPC1, we found that the interaction between SARAF and TRPC6 was constitutive rather than inducible by agonist stimulation. Furthermore, we found that SARAF expression silencing was without effect on Ca2+ entry evoked by agonists in TRPC6 overexpressing cells, as well as in Ca2+ influx evoked by the TRPC6 activator Hyp9. These findings provide evidence for a new regulator of TRPC1 channel function and highlight the relevance of SARAF in intracellular Ca2+ homeostasis.


Asunto(s)
Calcio/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Molécula de Interacción Estromal 1/metabolismo , Canales Catiónicos TRPC/metabolismo , Transporte Biológico/genética , Transporte Biológico/fisiología , Western Blotting , Línea Celular Tumoral , Humanos , Inmunoprecipitación , Proteínas Sensoras del Calcio Intracelular , Proteínas de la Membrana/genética , Proteínas de Neoplasias/genética , Unión Proteica/genética , Unión Proteica/fisiología , Molécula de Interacción Estromal 1/genética , Canales Catiónicos TRPC/genética , Canal Catiónico TRPC6
8.
Adv Exp Med Biol ; 898: 3-24, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27161222

RESUMEN

Calcium influx is an essential mechanism for the activation of cellular functions both in excitable and non-excitable cells. In non-excitable cells, activation of phospholipase C by occupation of G protein-coupled receptors leads to the generation of inositol 1,4,5-trisphosphate (IP3) and diacylglycerol (DAG), which, in turn, initiate two Ca(2+) entry pathways: Ca(2+) release from intracellular Ca(2+) stores, signaled by IP3, leads to the activation of store-operated Ca(2+) entry (SOCE); on the other hand, DAG activates a distinct second messenger-operated pathway. SOCE is regulated by the filling state of the intracellular calcium stores. The search for the molecular components of SOCE has identified the stromal interaction molecule 1 (STIM1) as the Ca(2+) sensor in the endoplasmic reticulum and Orai1 as a store-operated channel (SOC) subunit. Furthermore, a number of reports have revealed that several members of the TRPC family of channels also take part of the SOC macromolecular complex. This introductory chapter summarizes the early pieces of evidence that led to the concept of SOCE and the components of the store-operated signaling pathway.


Asunto(s)
Calcio/metabolismo , Animales , Humanos , Transporte Iónico , Proteínas de la Membrana/metabolismo , Canales Catiónicos TRPC/metabolismo
9.
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
10.
Biochem J ; 463(3): 373-81, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-25088676

RESUMEN

STIM1 (stromal interaction molecule 1) regulates store-operated channels in the plasma membrane, but the regulation of TRPC (transient receptor potential canonical) heteromultimerization and location by STIM1 is poorly understood. STIM1 is a single transmembrane protein that communicates the filling state of the endoplasmic reticulum to store-operated channels. STIM1 has been reported to regulate the activity of all of the TRPC family members, except TRPC7. TRPC6 has been predominantly associated to second messenger-activated Ca2+ entry pathways. In the present paper we report that STIM1 regulates the expression of TRPC6 in the plasma membrane and evokes translocation of this channel to the endoplasmic reticulum. Attenuation of TRPC6 expression in the plasma membrane resulted in a reduction in the association of this channel with TRPC1 and TRPC3. We have found that expression of TRPC6 in the endoplasmic reticulum results in an increase in the passive Ca2+ efflux and basal cytosolic Ca2+ concentration, but not in the ability of cells to accumulate Ca2+ into the endoplasmic reticulum. We propose a novel mechanism for the regulation of TRPC6 channel location and function by STIM1, probably as a mechanism to modulate second messenger-operated Ca2+ entry while potentiating store-operated Ca2+ influx.


Asunto(s)
Membrana Celular/metabolismo , Retículo Endoplásmico/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Canales Catiónicos TRPC/metabolismo , Animales , Línea Celular Tumoral , Humanos , Ratones , Multimerización de Proteína , Transporte de Proteínas , Ratas , Molécula de Interacción Estromal 1
11.
Biochim Biophys Acta ; 1833(12): 3025-3034, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23994313

RESUMEN

TRPA1 is a non-selective Ca(2+) permeable channel located in the plasma membrane that functions as a cellular sensor detecting mechanical, chemical and thermal stimuli, being a component of neuronal, epithelial, blood and smooth muscle tissues. TRPA1 has been shown to influence a broad range of physiological processes that involve Ca(2+)-dependent signaling pathways. Here we report that TRPA1 is expressed in MEG01 but not in platelets at the protein level. MEG01 cells maturation induced by PMA results in attenuation of TRPA1 protein expression and enhances thapsigargin-evoked Ca(2+) entry without altering the release of Ca(2+) from intracellular stores. Inhibition of TRPA1 by HC-030031 results in enhancement of both thrombin- and thapsigargin-stimulated Ca(2+) entry. Co-immunoprecipitation experiments revealed that TRPA1 associates with STIM1, as well as Orai1, TRPC1 and TRPC6. Downregulation of TRPA1 expression by MEG01 maturation, as well as pharmacological inhibition of TRPA1 by HC-030031, results in enhancement of the association between STIM1 and Orai1. Altogether, these findings provide evidence for a new and interesting function of TRPA1 in cellular function associated to the regulation of agonist-induced Ca(2+) entry by the modulation of STIM1/Orai1 interaction.


Asunto(s)
Canales de Calcio/metabolismo , Calcio/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Canales de Potencial de Receptor Transitorio/metabolismo , Acetanilidas/farmacología , Plaquetas/citología , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular , Humanos , Isotiocianatos/farmacología , Megacariocitos/citología , Megacariocitos/efectos de los fármacos , Megacariocitos/metabolismo , Proteína ORAI1 , Unión Proteica/efectos de los fármacos , Purinas/farmacología , Molécula de Interacción Estromal 1 , Canal Catiónico TRPA1 , Acetato de Tetradecanoilforbol/farmacología , Tapsigargina/farmacología , Trombina/farmacología , Proteínas de Unión al GTP rap/metabolismo
12.
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
13.
Arterioscler Thromb Vasc Biol ; 33(12): 2750-8, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24115034

RESUMEN

OBJECTIVE: Apoptotic-like phase is an essential step for the platelet formation from megakaryocytes. How controlled is this signaling pathway remained poorly understood. The aim of this study was to determine whether endoplasmic reticulum (ER) stress-induced apoptosis occurs during thrombopoiesis. APPROACH AND RESULTS: Investigation of ER stress and maturation markers in different models of human thrombopoiesis (CHRF, DAMI, MEG-01 cell lines, and hematopoietic stem cells: CD34(+)) as well as in immature pathological platelets clearly indicated that ER stress occurs transiently during thrombopoiesis. Direct ER stress induction by tunicamycin, an inhibitor of N-glycosylation, or by sarco/endoplasmic reticulum Ca(2+) ATPase type 3b overexpression, which interferes with reticular calcium, leads to some degree of maturation in megakaryocytic cell lines. On the contrary, exposure to salubrinal, a phosphatase inhibitor that prevents eukaryotic translation initiation factor 2α-P dephosphorylation and inhibits ER stress-induced apoptosis, decreased both expression of maturation markers in MEG-01 and CD34(+) cells as well as numbers of mature megakaryocytes and proplatelet formation in cultured CD34(+) cells. CONCLUSIONS: Taken as a whole, our research suggests that transient ER stress activation triggers the apoptotic-like phase of the thrombopoiesis process.


Asunto(s)
Estrés del Retículo Endoplásmico , Retículo Endoplásmico/metabolismo , Células Madre Hematopoyéticas/metabolismo , Megacariocitos/metabolismo , Trombopoyesis , Antígenos CD34/metabolismo , Apoptosis , Biomarcadores/metabolismo , Línea Celular , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/patología , Estrés del Retículo Endoplásmico/efectos de los fármacos , Factor 2 Eucariótico de Iniciación/metabolismo , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/patología , Humanos , Megacariocitos/efectos de los fármacos , Megacariocitos/patología , Fosforilación , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/genética , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Trombopoyesis/efectos de los fármacos , Factores de Tiempo , Transfección
14.
Arch Biochem Biophys ; 538(2): 130-7, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-23988350

RESUMEN

G protein-coupled receptors can be directly modulated by changes in transmembrane voltage in a variety of cell types. Here we show that, while changes in the membrane voltage itself do not induce detectable modifications in the cytosolic Ca(2+) concentration, platelet stimulation with thrombin or the PAR-1 and PAR-4 agonist peptides SFLLRN and AYPGKF, respectively, results in Ca(2+) release from intracellular stores that is sensitive to the membrane depolarisation. Direct activation of G proteins or phospholipase C by AlF4(-) and m-3M3FBS, respectively, leads to Ca(2+) release that is insensitive to changes in the membrane potential. Thapsigargin-, as well as OAG-induced Ca(2+) entry are affected by the membrane voltage, probably as a result of the modification in the driving force for Ca(2+) influx; however, hyperpolarisation does not enhance thrombin- or OAG-evoked Ca(2+) entry probably revealing the presence of a voltage-sensitive regulatory mechanism. Transmembrane voltage also modulates the activity of the plasma membrane Ca(2+)-ATPase (PMCA) most likely due to a decrease in the phosphotyrosine content of the pump. Thrombin-stimulated platelet aggregation is modulated by membrane depolarisation by a mechanism that is, at least partially, independent of Ca(2+). These observations indicate that PAR-1 and PAR-4 receptors are modulated by the membrane voltage in human platelets.


Asunto(s)
Plaquetas/citología , Calcio/metabolismo , Potenciales de la Membrana , Agregación Plaquetaria , Trombina/metabolismo , Plaquetas/metabolismo , Señalización del Calcio , Humanos , ATPasas Transportadoras de Calcio de la Membrana Plasmática/metabolismo , Receptor PAR-1/metabolismo , Receptores de Trombina/metabolismo
15.
Biochem J ; 445(1): 29-38, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22506990

RESUMEN

Homer is a family of cytoplasmic adaptor proteins that play different roles in cell function, including the regulation of G-protein-coupled receptors. These proteins contain an Ena (Enabled)/VASP (vasodilator-stimulated phosphoprotein) homology 1 domain that binds to the PPXXF sequence motif, which is present in different Ca²âº-handling proteins such as IP3 (inositol 1,4,5-trisphosphate) receptors and TRPC (transient receptor potential canonical) channels. In the present study we show evidence for a role of Homer proteins in the STIM1 (stromal interaction molecule 1)-Orai1 association, as well as in the TRPC1-IP3RII (type II IP3 receptor) interaction, which might be of relevance in platelet function. Treatment of human platelets with thapsigargin or thrombin results in a Ca²âº-independent association of Homer1 with TRPC1 and IP3RII. In addition, thapsigargin and thrombin enhanced the association of Homer1 with STIM1 and Orai1 in a Ca²âº-dependent manner. Interference with Homer function by introduction of the synthetic PPKKFR peptide into cells, which emulates the proline-rich sequences of the PPXXF motif, reduced STIM1-Orai1 and TRPC1- IP3RII associations, as compared with the introduction of the inactive PPKKRR peptide. The PPKKFR peptide attenuates thrombin-evoked Ca²âº entry and the maintenance of thapsigargin-induced store-operated Ca²âº entry. Finally, the PPKKFR peptide attenuated thrombin-induced platelet aggregation. The findings of the present study support an important role for Homer proteins in thrombin-stimulated platelet function, which is likely to be mediated by the support of agonist-induced Ca²âº entry.


Asunto(s)
Plaquetas/metabolismo , Canales de Calcio/metabolismo , Señalización del Calcio/fisiología , Proteínas Portadoras/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Canales Catiónicos TRPC/metabolismo , Plaquetas/efectos de los fármacos , Western Blotting , Señalización del Calcio/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Proteínas de Andamiaje Homer , Humanos , Inmunoprecipitación , Inositol 1,4,5-Trifosfato/metabolismo , Proteína ORAI1 , Fragmentos de Péptidos/metabolismo , Agregación Plaquetaria/efectos de los fármacos , Unión Proteica , Molécula de Interacción Estromal 1 , Tapsigargina/farmacología
16.
Biochim Biophys Acta ; 1813(8): 1483-94, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21601596

RESUMEN

BACKGROUND: A novel family of intracellular Ca(2+)-release channels termed two-pore channels (TPCs) has been presented as the receptors of NAADP (nicotinic acid adenine dinucleotide phosphate), the most potent Ca(2+) mobilizing intracellular messenger. TPCs have been shown to be exclusively localized to the endolysosomal system mediating NAADP-evoked Ca(2+) release from the acidic compartments. OBJECTIVES: The present study is aimed to investigate NAADP-mediated Ca(2+) release from intracellular stores in the megakaryoblastic cell line MEG01. METHODS: Changes in cytosolic and intraluminal free Ca(2+) concentrations were registered by fluorimetry using fura-2 and fura-ff, respectively; TPC expression was detected by PCR. RESULTS: Treatment of MEG01 cells with the H(+)/K(+) ionophore nigericin or the V-type H(+)-ATPase selective inhibitor bafilomycin A1 revealed the presence of acidic Ca(2+) stores in these cells, sensitive to the SERCA inhibitor 2,5-di-(tert-butyl)-1,4-hydroquinone (TBHQ). NAADP releases Ca(2+) from acidic lysosomal-like Ca(2+) stores in MEG01 cells probably mediated by the activation of TPC1 and TPC2 as demonstrated by TPC1 and TPC2 expression silencing and overexpression. Ca(2+) efflux from the acidic lysosomal-like Ca(2+) stores or the endoplasmic reticulum (ER) results in ryanodine-sensitive activation of Ca(2+)-induced Ca(2+) release (CICR) from the complementary Ca(2+) compartment. CONCLUSION: Our results show for the first time NAADP-evoked Ca(2+) release from acidic compartments through the activation of TPC1 and TPC2, and CICR, in a megakaryoblastic cell line.


Asunto(s)
Señalización del Calcio/fisiología , Células Progenitoras de Megacariocitos/metabolismo , NADP/análogos & derivados , Secuencia de Bases , Canales de Calcio/genética , Canales de Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Compartimento Celular , Línea Celular , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Inhibidores Enzimáticos/farmacología , Humanos , Concentración de Iones de Hidrógeno , Hidroquinonas/farmacología , Ionóforos/farmacología , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Macrólidos/farmacología , Células Progenitoras de Megacariocitos/efectos de los fármacos , NADP/metabolismo , Nigericina/farmacología , ARN Interferente Pequeño/genética , ATPasas de Translocación de Protón Vacuolares/antagonistas & inhibidores
17.
Curr Med Chem ; 26(22): 4119-4144, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-29210636

RESUMEN

A large number of studies have been focused on investigating serum biomarkers associated with risk or diagnosis of type-2 diabetes mellitus. In the last decade, promising studies have shown that circulating levels of adipokines could be used as a relevant biomarker for diabetes mellitus progression as well as therapeutic future targets. Here, we discuss the possible use of recently described adipokines, including apelin, omentin-1, resistin, FGF-21, neuregulin-4 and visfatin, as early biomarkers for diabetes. In addition, we also include recent findings of other well known adipokines such as leptin and adiponectin. In conclusion, further studies are needed to clarify the pathophysiological significance and clinical value of these biological factors as potential biomarkers in type-2 diabetes and related dysfunctions.


Asunto(s)
Adipoquinas/sangre , Diabetes Mellitus Tipo 2/sangre , Biomarcadores/sangre , Diabetes Mellitus Tipo 2/metabolismo , Humanos
18.
Channels (Austin) ; 11(1): 84-88, 2017 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-27414851

RESUMEN

The store-operated Ca2+ entry-associated regulatory factor (SARAF), a protein expressed both in the endoplasmic reticulum and the plasma membrane, has been presented as a STIM1-interacting protein with the ability to modulate intracellular Ca2+ homeostasis. SARAF negatively modulates store-operated Ca2+ entry (SOCE) by preventing STIM1 spontaneous activation and regulating STIM1-Orai1 complex formation. In addition, SARAF is a negative regulator of Ca2+ entry through the arachidonate-regulated Ca2+ (ARC) channels. Here we explored the possible role of the surface expression of SARAF on the location of STIM1 in the plasma membrane. In NG115-401L cells, lacking a detectable expression of native STIM1, transfection with pHluorin-STIM1, which is able to translocate to the cell surface, enhances the plasma membrane location of SARAF as compared to cells transfected with YFP-STIM1, lacking the ability to translocate to the cell surface. These findings suggest that the surface location of SARAF is dependent on the expression of STIM1 in the plasma membrane.


Asunto(s)
Membrana Celular/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Molécula de Interacción Estromal 1/metabolismo , Animales , Línea Celular Tumoral , Proteínas Sensoras del Calcio Intracelular , Ratones , Ratas
19.
Front Physiol ; 8: 392, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28649203

RESUMEN

According to the International Association for the Study of Pain (IASP) pain is characterized as an "unpleasant sensory and emotional experience associated with actual or potential tissue damage". The TRP super-family, compressing up to 28 isoforms in mammals, mediates a myriad of physiological and pathophysiological processes, pain among them. TRP channel might be constituted by similar or different TRP subunits, which will result in the formation of homomeric or heteromeric channels with distinct properties and functions. In this review we will discuss about the function of TRPs in pain, focusing on TRP channles that participate in the transduction of noxious sensation, especially TRPV1 and TRPA1, their expression in nociceptors and their sensitivity to a large number of physical and chemical stimuli.

20.
Sci Rep ; 6: 24452, 2016 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-27068144

RESUMEN

Ca(2+) influx by store-operated Ca(2+) channels is a major mechanism for intracellular Ca(2+) homeostasis and cellular function. Here we present evidence for the dynamic interaction between the SOCE-associated regulatory factor (SARAF), STIM1 and Orai1. SARAF overexpression attenuated SOCE and the STIM1-Orai1 interaction in cells endogenously expressing STIM1 and Orai1 while RNAi-mediated SARAF silencing induced opposite effects. SARAF impaired the association between Orai1 and the Orai1-activating small fragment of STIM1 co-expressed in the STIM1-deficient NG115-401L cells. Cell treatment with thapsigargin or physiological agonists results in direct association of SARAF with Orai1. STIM1-independent interaction of SARAF with Orai1 leads to activation of this channel. In cells endogenously expressing STIM1 and Orai1, Ca(2+) store depletion leads to dissociation of SARAF with STIM1 approximately 30s after treatment with thapsigargin, which paralleled the increase in SARAF-Orai1 interaction, followed by reinteraction with STIM1 and dissociation from Orai1. Co-expression of SARAF and either Orai1 or various N-terminal deletion Orai1 mutants did not alter SARAF-Orai1 interaction; however, expression of C-terminal deletion Orai1 mutants or blockade of the C-terminus of Orai1 impair the interaction with SARAF. These observations suggest that SARAF exerts an initial positive role in the activation of SOCE followed by the facilitation of SCDI of Orai1.


Asunto(s)
Calcio/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Proteína ORAI1/metabolismo , Mapas de Interacción de Proteínas , Molécula de Interacción Estromal 1/metabolismo , Homeostasis , Humanos , Proteínas Sensoras del Calcio Intracelular , Mapeo de Interacción de Proteínas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA