Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters











Publication year range
1.
Biosystems ; 237: 105138, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38340977

ABSTRACT

Pancreatic ß-cells are equipped with the molecular machinery allowing them to respond to high glucose levels in the form of electrical activity and Ca2+ oscillations. These oscillations drive insulin secretion. Two key ionic mechanisms involved in this response are the Store-Operated Current and the current through ATP-dependent K+ channels. Both currents have been shown to be regulated by the protein STIM1, but this dual regulation by STIM1 has not been studied before. In this paper, we use mathematical modelling to gain insight into the role of STIM1 in the ß-cell response. We extended a previous ß-cell model to include the dynamics of STIM1 and described the dependence of the ATP-dependent K+ current on STIM1. Our simulations suggest that the total concentration of STIM1 modifies the bursting frequency, the burst duration and the intracellular Ca2+ levels. These results are in good agreement with experimental reports, and the contribution of the studied currents to electrical activity and Ca2+ dynamics is discussed. The model predicts that in the absence of STIM1 the excitability of the plasma membrane increases and that the glucose threshold for electrical activity is shifted to lower concentrations. These computational predictions may be related to impaired insulin secretion under conditions of reduced STIM1 in the diabetic state.


Subject(s)
Insulin-Secreting Cells , Stromal Interaction Molecule 1 , Adenosine Triphosphate/metabolism , Calcium/metabolism , Calcium Signaling , Cell Membrane/metabolism , Glucose/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Stromal Interaction Molecule 1/metabolism , Humans
2.
Int J Mol Sci ; 24(6)2023 Mar 10.
Article in English | MEDLINE | ID: mdl-36982380

ABSTRACT

Triple-negative breast cancer has a poor prognosis and is non-responsive to first-line therapies; hence, new therapeutic strategies are needed. Enhanced store-operated Ca2+ entry (SOCE) has been widely described as a contributing factor to tumorigenic behavior in several tumor types, particularly in breast cancer cells. SOCE-associated regulatory factor (SARAF) acts as an inhibitor of the SOCE response and, therefore, can be a potential antitumor factor. Herein, we generated a C-terminal SARAF fragment to evaluate the effect of overexpression of this peptide on the malignancy of triple-negative breast cancer cell lines. Using both in vitro and in vivo approaches, we showed that overexpression of the C-terminal SARAF fragment reduced proliferation, cell migration, and the invasion of murine and human breast cancer cells by decreasing the SOCE response. Our data suggest that regulating the activity of the SOCE response via SARAF activity might constitute the basis for further alternative therapeutic strategies for triple-negative breast cancer.


Subject(s)
Membrane Proteins , Triple Negative Breast Neoplasms , Mice , Humans , Animals , Membrane Proteins/metabolism , Calcium/metabolism , Triple Negative Breast Neoplasms/metabolism , Ion Transport , Cytoplasm/metabolism , Calcium Signaling , Stromal Interaction Molecule 1/metabolism
3.
Cell Calcium ; 104: 102595, 2022 06.
Article in English | MEDLINE | ID: mdl-35561647

ABSTRACT

The use of a variety of techniques based on super-resolution (SR) microscopy unveiled a close and complex relationship between cytoskeleton reorganization and SOCE. By using SR microscopy many new proteins involved in SOCE regulation have been identified over the last few years. Many enigmas remain unsolved in this highly dynamic field, however, recent developments in SR microscopy promise new answers soon. In the present review, we describe the most relevant findings in SOCE components and SOCE modulation using different methods derived from SR microscopy.


Subject(s)
Calcium , Microscopy , Calcium/metabolism , Calcium Signaling/physiology , ORAI1 Protein/metabolism , Stromal Interaction Molecule 1/metabolism
4.
Curr Vasc Pharmacol ; 20(3): 272-283, 2022.
Article in English | MEDLINE | ID: mdl-35319374

ABSTRACT

Sustained and intermittent hypoxia produce vasoconstriction, arterial remodeling, and hypertension in the lung. Stromal interaction molecule (STIM)-activated transient receptor potential channels (TRPC) and calcium release-activated calcium channel protein (ORAI) channels (STOC) play key roles in the progression of pulmonary hypertension in pre-clinical models of animals subjected to sustained and intermittent hypoxia. The available evidence supports the theory that oxidative stress and hypoxic inducible factors upregulate and activate STIM-activated TRPC-ORAI Ca2+ channels, contributing to the pulmonary remodeling and hypertension induced by sustained hypoxia. However, less is known about the effects of oxidative stress and hypoxic inducible factors on the modulation of STIM-activated TRPC-ORAI channels following chronic intermittent hypoxia. In this review, we examined the emerging evidence supporting the theory that oxidative stress and hypoxic inducible factors induced by intermittent hypoxia upregulate and activate STIM-activated TRPC-ORAI Ca2+ channels. In addition, we used bioinformatics tools to search public databases for the genes involved in the upregulation of STIMactivated TRPC-ORAI Ca2+ channels and compare the differential gene expression and biological processes induced by intermittent and sustained hypoxia in lung cells.


Subject(s)
Calcium Release Activated Calcium Channels , Hypertension, Pulmonary , Hypertension , Stromal Interaction Molecules , Transient Receptor Potential Channels , Animals , Calcium/metabolism , Calcium Release Activated Calcium Channels/metabolism , Calcium Signaling , Hypertension/metabolism , Hypertension, Pulmonary/etiology , Hypoxia/complications , ORAI1 Protein/metabolism , Stromal Interaction Molecule 1/metabolism , Stromal Interaction Molecules/metabolism , Transient Receptor Potential Channels/metabolism
5.
Biol Res ; 52(1): 45, 2019 Aug 19.
Article in English | MEDLINE | ID: mdl-31426853

ABSTRACT

BACKGROUND: Resveratrol was reported to trigger the apoptosis of fibroblast-like synoviocytes in adjuvant arthritis rats but the subcellular mechanism remains unclear. Since ER stress, mitochondrial dysfunction and oxidative stress were involved in the effects of resveratrol with imbalance of calcium bio-transmission, store operated calcium entry (SOCE), a novel intracellular calcium regulatory pathway, may also participate in this process. RESULTS: In the present study, Resveratrol was found to suppress ORAI1 expression of a dose dependent manner while have no evident effects on STIM1 expressive level. Besides, resveratrol had no effects on ATP or TG induced calcium depletion but present partly dose-dependent suppression of SOCE. On the one hand, microinjection of ORAI1 overexpressed vector in sick toe partly counteracted the therapeutic effects of resveratrol on adjuvant arthritis and serum inflammatory cytokine including IL-1, IL-6, IL-8, IL-10 and TNF-α. On the other hand, ORAI1 SiRNA injection provided slight relief to adjuvant arthritis in rats. In addition, ORAI1 overexpression partly diminished the alleviation of hemogram abnormality induced by adjuvant arthritis after resveratrol treatment while ORAI1 knockdown presented mild resveratrol-like effect on hemogram in rats model. CONCLUSION: These results indicated that resveratrol reduced store-operated Ca2+ entry and enhanced the apoptosis of fibroblast-like synoviocytes in adjuvant arthritis rats model via targeting ORAI1-STIM1 complex, providing a theoretical basis for ORAI1 targeted therapy in future treatment with resveratrol on rheumatoid arthritis.


Subject(s)
Apoptosis/drug effects , Arthritis, Experimental/physiopathology , Calcium Channels/drug effects , Fibroblasts/drug effects , ORAI1 Protein/drug effects , Resveratrol/pharmacology , Stromal Interaction Molecule 1/drug effects , Synoviocytes/drug effects , Animals , Calcium Channels/physiology , Mitochondria/drug effects , Oxidative Stress/drug effects , Rats , Resveratrol/administration & dosage
6.
Biol. Res ; 52: 45, 2019. tab, graf
Article in English | LILACS | ID: biblio-1019509

ABSTRACT

BACKGROUND: Resveratrol was reported to trigger the apoptosis of fibroblast-like synoviocytes In adjuvant arthritis rats but the subcellular mechanism remains unclear. Since ER stress, mitochondrial dysfunction and oxidative stress were involved in the effects of resveratrol with imbalance of calcium bio-transmission, store operated calcium entry (SOCE), a novel intracellular calcium regulatory pathway, may also participate in this process. RESULTS: In the present study, Resveratrol was found to suppress ORAI1 expression of a dose dependent manner while have no evident effects on STIM1 expressive level. Besides, resveratrol had no effects on ATP or TG induced calcium depletion but present partly dose-dependent suppression of SOCE. On the one hand, microinjection of ORAI1 overexpressed vector in sick toe partly counteracted the therapeutic effects of resveratrol on adjuvant arthritis and serum inflammatory cytokine including IL-1, IL-6, IL-8, IL-10 and TNF-α. On the other hand, ORAI1 SiRNA injection provided slight relief to adjuvant arthritis in rats. In addition, ORAI1 overexpression partly diminished the alleviation of hemogram abnormality induced by adjuvant arthritis after resveratrol treatment while ORAI1 knockdown presented mild resveratrol-like effect on hemogram in rats model. CONCLUSION: These results indicated that resveratrol reduced store-operated Ca2+ entry and enhanced the apoptosis of fibroblast-like synoviocytes in adjuvant arthritis rats model via targeting ORAI1-STIM1 complex, providing a theoretical basis for ORAI1 targeted therapy in future treatment with resveratrol on rheumatoid arthritis.


Subject(s)
Animals , Rats , Arthritis, Experimental/physiopathology , Calcium Channels/drug effects , Apoptosis/drug effects , Fibroblasts/drug effects , Synoviocytes/drug effects , Stromal Interaction Molecule 1/drug effects , ORAI1 Protein/drug effects , Resveratrol/pharmacology , Calcium Channels/physiology , Oxidative Stress/drug effects , Resveratrol/administration & dosage , Mitochondria/drug effects
7.
Sci Rep ; 8(1): 13252, 2018 09 05.
Article in English | MEDLINE | ID: mdl-30185837

ABSTRACT

The involvement of inositol trisphosphate receptor (IP3R) in modulating store-operated calcium entry (SOCE) was established many years ago. Nevertheless, the molecular mechanism responsible for this observation has not been elucidated to this date. In the present study we show that IP3R associates to STIM1 upon depletion of the endoplasmic reticulum (ER) by activation of the inositol trisphosphate signaling cascade via G-protein coupled receptors. IP3R-STIM1 association results in enhanced STIM1 puncta formation and larger Orai-mediated whole-cell currents as well as increased calcium influx. Depleting the ER with a calcium ATPase inhibitor (thapsigargin, TG) does not induce IP3R-STIM1 association, indicating that this association requires an active IP3R. The IP3R-STIM1 association is only observed after IP3R activation, as evidenced by FRET experiments and co-immunoprecipitation assays. ER intraluminal calcium measurements using Mag-Fluo-4 showed enhanced calcium depletion when IP3R is overexpressed. A STIM1-GCaMP fusion protein indicates that STIM1 detects lower calcium concentrations near its EF-hand domain when IP3R is overexpressed when compared with the fluorescence reported by a GCaMP homogenously distributed in the ER lumen (ER-GCaMP). All these data together strongly suggest that activation of inositol trisphosphate signaling cascade induces the formation of the IP3R-STIM1 complex. The activated IP3R provides a reduced intraluminal calcium microenvironment near STIM1, resulting in enhanced activation of Orai currents and SOCE.


Subject(s)
Calcium/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Neoplasm Proteins/metabolism , Stromal Interaction Molecule 1/metabolism , EF Hand Motifs , Endoplasmic Reticulum/metabolism , HEK293 Cells , Humans , Inositol Phosphates/metabolism , Neoplasm Proteins/chemistry , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Stromal Interaction Molecule 1/chemistry
8.
Toxicol In Vitro ; 48: 121-127, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29337250

ABSTRACT

Stromal-interaction molecule 1 (STIM1)-mediated store-operated Ca2+ entry (SOCE) plays a key role in mediating cardiomyocyte hypertrophy, both in vitro and in vivo. Moreover, there is growing support for the contribution of SOCE to the Ca2+ overload associated with ischemia/reperfusion injury. Therefore, STIM1 inhibition is proposed as a novel target for controlling both hypertrophy and ischemia/reperfusion-induced Ca2+ overload. Our aim was to evaluate the effect of ML9, a STIM1 inhibitor, on cardiomyocyte viability. ML9 was found to induce cell death in cultured neonatal rat cardiomyocytes. Caspase-3 activation, apoptotic index and release of the necrosis marker lactate dehydrogenase to the extracellular medium were evaluated. ML9-induced cardiomyocyte death was not associated with increased intracellular ROS or decreased ATP levels. Moreover, treatment with ML9 significantly increased levels of the autophagy marker LC3-II, without altering Beclin1 or p62 protein levels. However, treatment with ML9 followed by bafilomycin-A1 did not produce further increases in LC3-II content. Furthermore, treatment with ML9 resulted in decreased LysoTracker® Green staining. Collectively, these data suggest that ML9-induced cardiomyocyte death is triggered by a ML9-dependent disruption of autophagic flux due to lysosomal dysfunction.


Subject(s)
Autophagy/drug effects , Azepines/toxicity , Lysosomes/drug effects , Lysosomes/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Stromal Interaction Molecule 1/antagonists & inhibitors , Animals , Animals, Newborn , Apoptosis/drug effects , Cell Death/drug effects , Cells, Cultured , Mitochondria/drug effects , Mitochondria/metabolism , Necrosis/chemically induced , Necrosis/pathology , Rats , Reactive Oxygen Species/metabolism
9.
Virus Res ; 245: 17-28, 2018 02 02.
Article in English | MEDLINE | ID: mdl-29269104

ABSTRACT

The role of Ca2+ during dengue virus (DENV) replication is unknown; thus, changes in Ca2+ homeostasis in DENV infected human hepatic HepG2 and Huh-7 cells were analyzed. Infected HepG2 cells, but not Huh-7 cells, showed a significant increase in plasma membrane permeability to Ca2+, while both cell lines showed marked reduced levels of Ca2+ stored in the endoplasmic reticulum. While the expression levels of STIM1 and ORAI1 showed no changes, STIM1 and ORAI1 were shown to co-localized in infected cells, indicating activation of the store-operated Ca2+ entry (SOCE) pathway. Finally, manipulation in the infected cells of the intra and extracellular Ca2+ levels by chelators (BAPTA-AM and EGTA), SOC inhibitor (SKF96365), IP3 Receptor antagonist (2APB) or increase of extracellular [Ca2+], significantly reduced DENV yield, but not vesicular stomatitis virus yield, used as a control. These results show that DENV infection alters cell Ca2+ homeostasis and that such changes favor viral replication.


Subject(s)
Calcium Chelating Agents/pharmacology , Calcium/metabolism , Dengue Virus/drug effects , Homeostasis/drug effects , Host-Pathogen Interactions , Virus Replication/drug effects , Animals , Boron Compounds/pharmacology , Calcium Channel Blockers/pharmacology , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Cell Membrane/virology , Cell Membrane Permeability , Chlorocebus aethiops , Dengue Virus/physiology , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/ultrastructure , Endoplasmic Reticulum/virology , Gene Expression , Hep G2 Cells , Humans , Imidazoles/pharmacology , Inositol 1,4,5-Trisphosphate Receptors/antagonists & inhibitors , Inositol 1,4,5-Trisphosphate Receptors/genetics , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Ion Transport , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , ORAI1 Protein/antagonists & inhibitors , ORAI1 Protein/genetics , ORAI1 Protein/metabolism , Stromal Interaction Molecule 1/antagonists & inhibitors , Stromal Interaction Molecule 1/genetics , Stromal Interaction Molecule 1/metabolism , Vero Cells , Vesicular stomatitis Indiana virus/drug effects , Vesicular stomatitis Indiana virus/physiology , Virus Replication/genetics
10.
Am J Hypertens ; 30(4): 389-399, 2017 Apr 01.
Article in English | MEDLINE | ID: mdl-28164209

ABSTRACT

BACKGROUND: We investigated the influence of salt overconsumption on the functionality of the RhoA/Rho-associated kinase (ROCK) pathway and calcium regulation in arteries. METHODS: The aorta and small mesenteric arteries from rats fed a chow containing 2%, 4%, or 8% NaCl were evaluated in organ baths for the activity of the RhoA/ROCK pathway and intracellular calcium mobilization. Components of these pathways and intracellular calcium levels were also assessed in samples from 4% NaCl group. RESULTS: In arteries from animals fed regular chow, the ROCK inhibitor Y-27632 reduced the responses to phenylephrine, even when the smallest concentrations (1 and 3 µM) were tested. However, only higher concentrations of Y-27632 (10 and 50 µM) reduced phenylephrine-induced contraction in vessels from high-salt groups. Immunoblotting revealed augmented phosphorylation of the myosin phosphatase targeting subunit 1 and increased amounts of RhoA in the membrane fraction of aorta homogenates from the 4% NaCl group. Under calcium-free solution, vessels from NaCl groups presented reduced contractile responses to phenylephrine and caffeine, compared with the regular chow group. Moreover, decreased intracellular calcium at rest and after stimulation with ATP were found in aortic smooth muscle cells from 4% NaCl-fed rats, which also showed diminished levels of SERCA2 and SERCA3, but not of IP3 and ryanodine receptors, or STIM1 and Orai1 proteins. CONCLUSIONS: Arteries from rats subjected to high-salt intake are unable to properly regulate intracellular calcium levels and present augmented activity of the calcium sensitization pathway RhoA/ROCK. These changes may precede the development of vascular diseases induced by high-salt intake.


Subject(s)
Aorta/drug effects , Calcium/metabolism , Mesenteric Arteries/drug effects , Myocytes, Smooth Muscle/drug effects , Sodium Chloride, Dietary/pharmacology , Vasoconstriction/drug effects , rho GTP-Binding Proteins/drug effects , rho-Associated Kinases/drug effects , Amides/pharmacology , Animals , Aorta/cytology , Aorta/metabolism , Enzyme Inhibitors/pharmacology , Inositol 1,4,5-Trisphosphate Receptors/drug effects , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Male , Mesenteric Arteries/cytology , Mesenteric Arteries/metabolism , Myocytes, Smooth Muscle/metabolism , ORAI1 Protein/drug effects , ORAI1 Protein/metabolism , Phenylephrine/pharmacology , Phosphorylation/drug effects , Protein Phosphatase 1/drug effects , Protein Phosphatase 1/metabolism , Pyridines/pharmacology , Rats , Rats, Wistar , Ryanodine Receptor Calcium Release Channel/drug effects , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/drug effects , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Signal Transduction/drug effects , Stromal Interaction Molecule 1/drug effects , Stromal Interaction Molecule 1/metabolism , Vasoconstrictor Agents/pharmacology , rho GTP-Binding Proteins/metabolism , rho-Associated Kinases/metabolism
11.
Curr Vasc Pharmacol ; 15(3): 265-281, 2017.
Article in English | MEDLINE | ID: mdl-28155613

ABSTRACT

BACKGROUND: Chronic ethanol (EtOH) consumption has been associated with deleterious effects on the cardiovascular system by abnormal calcium (Ca2+) handling. Store-operated Ca2+ entry (SOCE) is related to cardiovascular remodeling which leads to the hypertension development, and the coupling between STIM-1 (ER Ca2+ sensor) and Orai-1 (channel pore) is a key mechanism to control SOCE through of store-operated Ca2+ channels (SOCCs). However, the role of STIM-1/Orai-1-mediated SOCE and its cross-talk with EtOH-triggered vascular remodeling and hypertension remain poorly understood. We address this subject in the present study by evaluating how chronic EtOH consumption induces alterations in Ca2+ handling via SOCE. METHODS: Male Wistar Kyoto (WKY) and Spontaneously Hypertensive (SHR) rats were subjected to the intake of increasing EtOH concentrations (5-20%, for 30 days). Systolic blood pressure (SBP) and EtOH concentration were measured; cardiovascular remodeling was assessed by histomorphometry; and function/ expression of STIM-1/Orai-1-mediated Ca2+ influx were evaluated by isometric contraction and western blot experiments. RESULTS: Compared to the WKY-Control, our results show that: (1) chronic EtOH consumption caused a significant elevation of SBP in both strains; (2) cardiac hypertrophy and hypertrophic aortic wall remodeling much more pronounced in WKY-EtOH; (3) decreased capacity of ER to store and release Ca2+; (4) increased STIM-1/Orai-1-mediated SOCCs activation, which was selectively inhibited by YM-58483; and (5) increased expression of STIM-1 in WKY-EtOH and SHR-Control rats. CONCLUSION: These findings suggest that hypertrophic aortic remodeling and abnormal contraction triggered mainly by Ca2+ overload via STIM-1/Orai-1-mediated SOCE through SOCCs are involved hypertension developed by EtOH consumption.


Subject(s)
Alcohol Drinking/adverse effects , Calcium Signaling , Calcium/metabolism , Ethanol , Hypertension/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , ORAI1 Protein/metabolism , Stromal Interaction Molecule 1/metabolism , Animals , Aorta, Thoracic/metabolism , Aorta, Thoracic/physiopathology , Blood Pressure , Disease Models, Animal , Endoplasmic Reticulum/metabolism , Hypertension/etiology , Hypertension/physiopathology , Male , Muscle, Smooth, Vascular/physiopathology , Myocytes, Cardiac/metabolism , Rats, Inbred SHR , Rats, Inbred WKY , Time Factors , Up-Regulation , Vascular Remodeling , Vasoconstriction
12.
Inflammation ; 39(3): 1188-97, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27099074

ABSTRACT

We hypothesized that SIRS/endotoxemia-associated hyporesponsiveness to vasoconstrictors is mediated by smaller increases in intracellular Ca(2+) levels due to reduced signaling via the STIM/Orai. Male Wistar rats were injected either with saline or bacterial LPS (i.p.; 10 mg/kg), and experiments were performed 24 h later. LPS-injected rats exhibited decreased systolic blood pressure, increased heart rate, neutrophils' migration into the peritoneal cavity, and elevated alanine aminotransferase levels. Additionally, second-order mesenteric arteries from endotoxemic rats displayed hyporeactivity to contractile agents such as phenylephrine and potassium chloride; decreased contractile responses to Ca(2+); reduced contraction during Ca(2+) loading; and smaller intracellular Ca(2+) stores. Decreased Orai1, but not STIM1, expression was found in resistance mesenteric arteries from LPS-treated rats. Additionally, cultured vascular smooth muscle cell (VSMC) treated with LPS resulted in increased TLR-4 expression, but Myd-88 and STIM-1 expression were not changed. Our data suggest that in endotoxemia, Ca(2+) homeostasis is disrupted in VSMC, with decreased Ca(2+) influx, smaller concentrations of Ca(2+) in the sarcoplasmic reticulum, and decreased activation of Orai1. Abnormal Ca(2+) handling contributes to LPS-associated vascular hyporeactivity.


Subject(s)
Calcium/metabolism , Endotoxemia/physiopathology , Homeostasis , Mesenteric Arteries/physiopathology , ORAI1 Protein/metabolism , Animals , Cells, Cultured , Endotoxemia/metabolism , Lipopolysaccharides/pharmacology , Male , Muscle, Smooth, Vascular/pathology , ORAI1 Protein/analysis , Rats , Rats, Wistar , Stromal Interaction Molecule 1/analysis , Stromal Interaction Molecule 1/metabolism , Vasoconstrictor Agents/pharmacology
13.
Biocell ; 36(2): 73-81, 2012 Aug.
Article in English | MEDLINE | ID: mdl-23185782

ABSTRACT

After depletion of intracellular Ca2+ stores the capacitative response triggers an extracellular Ca2+ influx through store-operated channels (SOCs) which refills these stores. Our objective was to explore if human umbilical artery smooth muscle presented this response and if it was involved in the mechanism of serotonin- and histamine-induced contractions. Intracellular Ca2+ depletion by a Ca(2+)-free extracellular solution followed by Ca2+ readdition produced a contraction in artery rings which was inhibited by the blocker of Orai and TRPC channels 2-aminoethoxydiphenyl borate (2-APB), suggesting a capacitative response. In presence of 2-APB the magnitude of a second paired contraction by serotonin or histamine was significantly less than a first one, likely because 2-APB inhibited store refilling by capacitative Ca2+ entry. 2-APB inhibition of sarcoplasmic reticulum Ca2+ release was excluded because this blocker did not affect serotonin force development in a Ca(2+)-free solution. The PCR technique showed the presence of mRNAs for STIM proteins (1 and 2), for Orai proteins (1, 2 and 3) and for TRPC channels (subtypes 1, 3, 4 and 6) in the smooth muscle of the human umbilical artery. Hence, this artery presents a capacitative contractile response triggered by stimulation with physiological vasoconstrictors and expresses mRNAs for proteins and channels previously identified as SOCs.


Subject(s)
Boron Compounds/pharmacology , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Muscle, Smooth/metabolism , RNA, Messenger/genetics , Umbilical Arteries/drug effects , Vascular Capacitance/drug effects , Blotting, Western , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Calcium Channels/chemistry , Calcium Channels/genetics , Calcium Channels/metabolism , Cells, Cultured , Histamine/pharmacology , Histamine Agonists/pharmacology , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Muscle, Smooth/cytology , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , ORAI1 Protein , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum/metabolism , Serotonin/pharmacology , Serotonin Receptor Agonists/pharmacology , Stromal Interaction Molecule 1 , TRPC Cation Channels/genetics , TRPC Cation Channels/metabolism , Umbilical Arteries/cytology , Umbilical Arteries/metabolism
14.
Cell Calcium ; 52(6): 457-67, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22944608

ABSTRACT

In astrocytes, thrombin leads to cytoplasmic Ca(2+) elevations modulating a variety of cytoprotective and cytotoxic responses. Astrocytes respond to thrombin stimulation with a biphasic Ca(2+) increase generated by an interplay between ER-Ca(2+) release and store-operated Ca(2+) entry (SOCE). In many cell types, STIM1 and Orai1 have been demonstrated to be central components of SOCE. STIM1 senses the ER-Ca(2+) depletion and binds Orai1 to activate Ca(2+) influx. Here we used immunocytochemistry, overexpression and siRNA assays to investigate the role of STIM1 and Orai1 in the thrombin-induced Ca(2+) response in primary cultures of rat cortical astrocytes. We found that STIM1 and Orai1 are endogenously expressed in cortical astrocytes and distribute accordingly with other mammalian cells. Importantly, native and overexpressed STIM1 reorganized in puncta under thrombin stimulation and this reorganization was reversible. In addition, the overexpression of STIM1 and Orai1 increased by twofold the Ca(2+) influx evoked by thrombin, while knockdown of endogenous STIM1 and Orai1 significantly decreased this Ca(2+) influx. These results indicate that STIM1 and Orai1 underlie an important fraction of the Ca(2+) response that astrocytes exhibit in the presence of thrombin. Thrombin stimulation in astrocytes leads to ER-Ca(2+) release which causes STIM1 reorganization allowing the activation of Orai1 and the subsequent Ca(2+) influx.


Subject(s)
Astrocytes/drug effects , Calcium Channels/metabolism , Calcium/metabolism , Membrane Glycoproteins/metabolism , Thrombin/pharmacology , Animals , Animals, Newborn , Astrocytes/cytology , Astrocytes/metabolism , Calcium Channels/chemistry , Calcium Channels/genetics , Cells, Cultured , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/genetics , ORAI1 Protein , RNA Interference , RNA, Small Interfering/metabolism , Rats , Rats, Wistar , Stromal Interaction Molecule 1 , Thapsigargin/pharmacology
15.
Biochim Biophys Acta ; 1820(12): 1997-2006, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23000491

ABSTRACT

BACKGROUND: Sleep is a physiological event that directly influences health by affecting the immune system, in which calcium (Ca(2+)) plays a critical signaling role. We performed live cell measurements of cytosolic Ca(2+) mobilization to understand the changes in Ca(2+) signaling that occur in splenic immune cells after various periods of sleep deprivation (SD). METHODS: Adult male mice were subjected to sleep deprivation by platform technique for different periods (from 12 to 72h) and Ca(2+) intracellular fluctuations were evaluated in splenocytes by confocal microscopy. We also performed spleen cell evaluation by flow cytometry and analyzed intracellular Ca(2+) mobilization in endoplasmic reticulum and mitochondria. Additionally, Ca(2+) channel gene expression was evaluated RESULTS: Splenocytes showed a progressive loss of intracellular Ca(2+) maintenance from endoplasmic reticulum (ER) stores. Transient Ca(2+) buffering by the mitochondria was further compromised. These findings were confirmed by changes in mitochondrial integrity and in the performance of the store operated calcium entry (SOCE) and stromal interaction molecule 1 (STIM1) Ca(2+) channels. CONCLUSIONS AND GENERAL SIGNIFICANCE: These novel data suggest that SD impairs Ca(2+) signaling, most likely as a result of ER stress, leading to an insufficient Ca(2+) supply for signaling events. Our results support the previously described immunosuppressive effects of sleep loss and provide additional information on the cellular and molecular mechanisms involved in sleep function.


Subject(s)
Calcium Signaling/physiology , Calcium/metabolism , Mitochondria/immunology , Mitochondria/metabolism , Sleep Deprivation/immunology , Spleen/cytology , Animals , Endoplasmic Reticulum/metabolism , Lysosomes/metabolism , Male , Membrane Potential, Mitochondrial , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Sleep Deprivation/metabolism , Sleep Deprivation/pathology , Spleen/immunology , Spleen/metabolism , Stromal Interaction Molecule 1
16.
Braz J Med Biol Res ; 44(11): 1080-7, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22002090

ABSTRACT

Highly efficient mechanisms regulate intracellular calcium (Ca2+) levels. The recent discovery of new components linking intracellular Ca2+ stores to plasma membrane Ca2+ entry channels has brought new insight into the understanding of Ca2+ homeostasis. Stromal interaction molecule 1 (STIM1) was identified as a Ca2+ sensor essential for Ca2+ store depletion-triggered Ca2+ influx. Orai1 was recognized as being an essential component for the Ca2+ release-activated Ca2+ (CRAC) channel. Together, these proteins participate in store-operated Ca2+ channel function. Defective regulation of intracellular Ca2+ is a hallmark of several diseases. In this review, we focus on Ca2+ regulation by the STIM1/Orai1 pathway and review evidence that implicates STIM1/Orai1 in several pathological conditions including cardiovascular and pulmonary diseases, among others.


Subject(s)
Calcium Channels/metabolism , Calcium Signaling/physiology , Calcium/metabolism , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Cardiovascular Diseases/metabolism , Humans , Lung Diseases/metabolism , ORAI1 Protein , Stromal Interaction Molecule 1
17.
Cell Calcium ; 45(5): 439-46, 2009 May.
Article in English | MEDLINE | ID: mdl-19327826

ABSTRACT

Depletion of intracellular calcium stores leads to the activation of calcium influx via the so-called store-operated channels (SOCs). Recent evidence positions Orai proteins as the putative channels responsible for this process. The stromal interacting molecule (STIM1) has been recently identified as the calcium sensor located at the endoplasmic reticulum (ER), and responsible for communicating the deplete state of calcium stores to Orai at the plasma membrane (PM). However, recent experimental findings suggest that Orai and STIM1 are only part of a larger molecular complex required to modulate store-operated calcium entry (SOCE). In the present study we describe the assembly of the several of the components from the SOC complex in real-time, utilizing a novel imaging method. Using FRET imaging we show that under resting conditions (with calcium stores replenished) STIM1 travels continuously through the ER associated to the microtubule tracking protein, EB1. Upon depletion of the ER STIM1 dissociates from EB1 and aggregates into macromolecular complexes at the ER which includes the microsomal calcium ATPase. This association follows the assembly of Orai into macromolecular aggregates at the PM. We show that STIM1-Orai association follows a similar time course as that of Orai aggregation at the PM. During this last step of the process, calcium-selective, whole-cell inward currents developed, simultaneously. We show that this process is fully reversible. Replenishing intracellular calcium stores induces STIM1-Orai complex dissociation and shuts down inward currents. Under these conditions STIM1 re-associates to EB1, and reinitiates its travel through the ER.


Subject(s)
Calcium Channels/metabolism , Calcium/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Calcium Channels/genetics , Cell Line , Endoplasmic Reticulum/metabolism , Enzyme Inhibitors/metabolism , Fluorescence Resonance Energy Transfer , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , ORAI1 Protein , Patch-Clamp Techniques , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Stromal Interaction Molecule 1 , Thapsigargin/metabolism
18.
Cell Calcium ; 44(5): 479-91, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18420269

ABSTRACT

While the role of members from the TRPC family of channels as receptor-operated channels (ROC) is well established and supported by numerous studies, the role of this family of channels as store-operated channels (SOC) has been the focus of a heated controversy over the last few years. In the present study, we have explored the modulation of STIM1 on human TRPC1 channel. We show that the association of STIM1 to TRPC1 favors the insertion of TRPC1 into lipid rafts, where TRPC1 functions as a SOC. In the absence of STIM1, TRPC1 associates to other members from the TRPC family of channels to form ROCs. A novel TIRFM-FRET method illustrates the relevance of the dynamic association between STIM1 and TRPC1 for the activation of SOC and the lipid raft localization of the STIM1-TRPC1 complex. This study provides new evidence about the dual activity of TRPC1 (forming ROC or SOC) and the partners needed to determine TRPC1 functional fate. It highlights also the role of plasma membrane microdomains and ER-PM junctions in modulating TRPC1 channel function and its association to STIM1.


Subject(s)
Membrane Microdomains/metabolism , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , TRPC Cation Channels/metabolism , Cell Line , Enzyme Inhibitors/metabolism , Humans , Membrane Proteins/genetics , Neoplasm Proteins/genetics , Patch-Clamp Techniques , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Stromal Interaction Molecule 1 , TRPC Cation Channels/genetics , Thapsigargin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL