IMR Press / RCM / Volume 20 / Issue 3 / DOI: 10.31083/j.rcm.2019.03.522
Open Access Review
Store-operated calcium channels: Potential target for the therapy of hypertension
Show Less
1 Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Max Rady College of Medicine, University of Manitoba, Winnipeg, R2H 2A6, Canada
2 Kinesiology and Nutritional Science, California State University, Los Angeles, CA, 90032, USA
*Correspondence: nsdhalla@sbrc.ca (Naranjan S. Dhalla)
Rev. Cardiovasc. Med. 2019, 20(3), 139–151; https://doi.org/10.31083/j.rcm.2019.03.522
Submitted: 5 July 2019 | Accepted: 22 August 2019 | Published: 30 September 2019
Copyright: © 2019 Bhullar et al. Published by IMR press.
This is an open access article under the CC BY-NC 4.0 license https://creativecommons.org/licenses/by/4.0/.
Abstract

Effective therapy of hypertension represents a key strategy for reducing the burden of cardiovascular disease and its associated mortality. The significance of voltage dependent L-type Ca 2+ channels to Ca 2+ influx, and of their regulatory mechanisms in the development of heart disease, is well established. A wide variety of L-type Ca2+ channel inhibitors and Ca2+ antagonists have been found to be beneficial not only in the treatment of hypertension, but also in myocardial infarction and heart failure. Over the past two decades, another class of Ca 2+ channel - the voltage independent store-operated Ca 2+ channel - has been implicated in the regulation and fine tuning of Ca2+ entry in both cardiac and smooth muscle cells. Store-operated Ca2+ channels are activated by the depletion of Ca2+ stores within the endoplasmic/sarcoplasmic reticulum, or by low levels of cytosolic Ca 2+, thereby facilitating agonist-induced Ca 2+ influx. Store-operated Ca 2+ entry through this pivotal pathway involves both stromal interaction molecule (STIM) and Orai channels. Different degrees of changes in these proteins are considered to promote Ca2+ entry and hence contribute to the pathogenesis of cardiovascular dysfunction. Several blockers of store-operated Ca 2+ channels acting at the level of both STIM and Orai channels have been shown to depress Ca 2+ influx and lower blood pressure. However, their specificity, safety, and clinical significance remain to be established. Thus, there is an ongoing challenge in the development of selective inhibitors of store-operated Ca2+ channels that act in vascular smooth muscles for the improved treatment of hypertension.

Keywords
Store-operated Ca2+-channels
endoplasmic/sarcoplasmic Ca2+ stores
stromal interaction molecule
orai channels
hypertension therapy
1.Introduction

Although significant improvements in the management and treatment of coronary heart disease and stroke have reduced overall cardiovascular mortality in recent decades, it remains the number one killer worldwide (Forouzanfar et al., 2017; Lawes et al., 2008; Wellman et al., 2001). As hypertension is the most salient risk factor for cardiovascular disease, contributing to approximately 54% of all strokes and 47% of ischemic heart disease occurrences worldwide (Chaturvedi, 2003; Forouzanfar et al., 2017; Stanaway et al., 2018), this review is focused upon its pathophysiology and treatment. In particular, in view of the critical role of Ca2+ in determining the status of cardiovascular function (Barlow et al., 2006; Bers, 2008; Berridge et al., 2000; Carafoli, 2003; Cortes et al., 1997; Dhalla et al., 1977, 1982), we also describe the roles of various types of Ca2+ channels in health and disease, as well as therapeutic interventions that inhibit the vascular contractile response via blockage of Ca2+ entry into vascular smooth muscle. In doing so, we highlight recent discoveries in types of Ca2+ channels and the development of their inhibitors for the therapy of hypertension.

2.Cardiovascular Abnormalities and Ca2+-channel Antagonists

According to Global Health Observatory data, 1.13 billion people globally are affected with elevated blood pressure (BP), which increases morbidity of conditions such as left ventricular hypertrophy, coronary heart disease, heart failure, atrial fibrillation, and peripheral artery disease (Lewington et al., 2002; Manolis, et al., 2015; Mrowka, 2019; Wei et al., 2017). Hypertension itself, however, may not be associated with symptoms (Dorans, et al, 2018; Khoury and Ratchford, 2018; Whelton et al, 2018). Many pathophysiological factors are known to be involved in the pathogenesis of hypertension, including structural and functional abnormalities as well as molecular and cellular mechanisms underlying cardiovascular alterations (e.g. cardiac output, peripheral resistance, the renin-angiotensin-aldosterone system, the sympathetic nervous system, endothelial dysfunction, and loss of nitric oxide (NO) bioavailability)(Bartekova et al., 2015; Beevers et al., 2001; Bhatt et al., 2014; Cain and Khalil, 2002; Chiong et al., 2008; Eid et al., 2018; Oparil et al., 2003). Furthermore, impaired vasodilation, impaired Ca2+ signaling, oxidative stress, and the production of pro-inflammatory cytokines and pro-fibrotic growth factors are thought to play a role (Beevers et al., 2001; Carretero and Oparil., 2000; Fritze et al., 2012; Gates et al., 2009; Green et al., 2010; Van den et al., 2012). Several clinical trials have demonstrated that antihypertensive therapy reduces cardiovascular disease events and all-cause mortality (Dorans et al., 2018; Tocci et al., 2015). Treatments for hypertension include diuretics, beta- blockers, angiotensin-converting enzyme inhibitors, angiotensin II receptor blockers, vasodilators, and calcium channel antagonists (Bhatt et al., 2014; Chobanian et al., 2003; Fleckenstein, 1977; Godfraind, 2017; Gong et al., 1996; Kuramoto, 1999; Liu et al., 1998; Ozawa et al., 2006; Staessen et al., 1997; Tocci et al., 2015).

Since Ca2+ is known to play a critical role in transforming extracellular stimuli into intracellular signalling, its entry is controlled by the presence of different types of Ca2+ channels within the cell plasma membrane (Bers, 2008; Bean and McDonough,2010; Carafoli, 2003). The concept of Ca2+ entry blockade by drugs in hypertension was developed in the 1960s in pharmacological screening studies of coronary dilators; these agents were later called Ca2+ channel blockers or Ca2+ antagonists (Dhalla et al., 1982; Fleckenstein, 1977; Godfraind, 2017). The classification of numerous Ca2+ -channel antagonists used therapeutically is given in Table 1 (Bangalore et al., 1994; Dilmac et al., 2003; Hockerman et al., 1997; Hofmann et al., 1999; Kurokawa et al., 1997; Remuzzi et al., 2002; Scultety and Tamaskovits, 1991; Wang et al., 1994). Ca2+ antagonists are most frequently used in the treatment of cardiovascular disease and have been demonstrated to work by blocking Ca2+ entry through voltage dependent L-type calcium channels (Abernethy and Schwartz, 1999). The use of N-type and T-type calcium channel blockers has also been associated with reductions in cardiovascular events and renal injury, as well as the alleviation of hypertension (Ozawa et al., 2006). According to existing evidence, Ca2+ antagonists exert vasodilatory action on vascular smooth muscle cells by inhibiting Ca2+ entry through L-type calcium channels, whereas the blockade of N-type or T-type calcium channels in cardiac pacemaker cells may suppress heart rate and thus reduce cardiac events and improve survival (Harada et al., 2003; Takahara et al.,2004). Several L-type Ca2+ channel antagonists including verapamil, nifedipine, and diltiazem are now known for their beneficial effects in reducing BP and in the treatment of hypertension (Table 1). However a major side effect of Ca2+ channel antagonists, namely the depression of cardiac function, limits their clinical use in hypertension. As such,efforts are being made to discover other types of Ca2+entry blockers acting upon voltage independent Ca2+-channels, which may confer fewer side effects (Collins et al., 2013; Colovina et al., 1999; Guibert et al., 2008; Leung et al., 2008; Xu et al., 2015).

10.31083/j.rcm.2019.03.522.t0001 Table 1 Classification and Effects of Different Types of Ca2+-antagonists
Classification Antagonists Effects References
A. Dihydropyridines Amlodipine, Aranidipine, Azelnidipine, Barnidipine, Benidipine, Cilnidipine, Clevidipine, Efonidipine, Felodipine, Isradipine, Lacidipine, Lercanidipine, Manidipine, Nicardipine, Nifedipine, Nilvadipine, Nimodipine, Nisoldipine,Nitrendipine, Pranidipine Reduce systemic vascular resistance and arterial pressure. Bangalore et al., 1994; Kurokawa et al., 1997; Remuzzi et al., 2002
B. Non-Dihydropyridines
a. Benzothiazepines Clenazem, Diltiazem Reduce arterial pressure Hofmann et al., 1999
b. Phenylalkylamines Gallopamil, Verapamil, Fendiline Reduce myocardial oxygen demand, reverse coronary vasospasm Dilmac et al.,2003; Hockerman et al., 1997
C. Non-selective Bepridil, Flunarizine, Fluspirilene, Fendiline, Mibefradil Treat epilepsy and neuropathic pain Scultety and Tamaskovits, 1991
D. Non-medical Ethanol Induce muscle relaxation Wang et al., 1994
3.Store-operated Calcium Channels in Health and Disease

To date, knowledge is incomplete regarding the role of store-operated calcium channels (SOCC) in the pathogenesis of cardiovascular disease. A recently conducted cursory PubMed search found 143 research articles (of which 35 were reviews) for their role in cardiovascular diseases, including 123 research articles and 21 reviews for their role in hypertension. Although some investigators (Avila-Medina et al., 2018; Bolotina, 2008; Godfraind, 2017; Ozawa et al., 2006; Parekh and Putney, 2005; Tanwar et al., 2017) have attempted to analyze the existing data on SOCC, the mechanisms of the store-operated Ca2+ entry (SOCE) and the components of this pathway remain to be fully elucidated. It should be noted that SOCC are activated by depletion of Ca2+ stores within the endoplasmic/sarcoplasmic reticulum (ER/S R) or by low levels of cytosolic Ca2+([Ca2+]i) (Lambert et al., 2018; McFadzean and Gibson, 2002). These channels co-exist with voltage dependent Ca2+ channels within the plasma membrane of excitable tissues including cardiomyocytes, neurons, vascular myocytes and skeletal muscle cells (Arakawa et al., 2000; Collins et al., 2013; Pang et al., 2002; Trepakova et al., 2000) and are involved in Ca2+ entry from the extracellular space (Putney, 2018). The main feature differentiating voltage independent Ca2+ channels from all other types of Ca2+ channels is their activation, which occurs by the depletion of Ca2+ stores from the lumen of ER/SR (Lewis, 2011). SOCE was first conceptualized in the early 1980s (during which period it was known as ‘capacitative Ca2+ entry’), describing direct Ca2+ entry from the extracellular space to refill ER/SR Ca2+ stores (Putney, 1986, 1990, 2009). It was shown that Ca2+ is released form the intracellular inositol-1, 4, 5-trisphosphate (IP3)- or ryanodine (RyR)-sensitive SR stores in response to physiological stimuli (Lewis, 2011). This Ca2+ efflux and consequent depletion of intracellular Ca2+ stores thus demands the influx of extracellular Ca2+ into the cytosol in order for stores to be replenished (Bose, 2017; Parekh and Putney, 2005; Prakriya and Lewis, 2015; Putney, 2011). The accumulated evidence supporting the notion of sensing of ER/SR Ca2+ stores for the control of Ca2+ influx represents the nascence of the SOCC model (Putney, 1986, 1990).

3.1 Physiological and Pathophysiological role of SOCC

SOCC are notable particularly with regard to their activation via retrograde signalling mechanisms, as well as their imperative role under both physiological and pathophysiological conditions(Leung and Kwan, 1999). The replenishment of intracellular Ca2+ stores following their depletion during intracellular Ca2+ signaling has long been thought as the main function of SOCC; however, they have recently been found to carry out other tasks, which may offer direct Ca2+ signals to locations near Ca2+ entry sites to recruit particular pathways. A growing number of studies have supported the role of SOCC in biological processes including endothelial cell proliferation (Abdullaev et al., 2008), breast cancer cell migration and metastasis (Yang et al., 2009), skeletal muscle contraction (Stiber et al., 2008), and smooth muscle migration and proliferation (Berra-Romani et al., 2008; Potier et al., 2009). SOCC have also been implicated in a number of human disorders including cardiovascular diseases, immunodeficiency, acute pancreatitis, Alzheimer’s disease, Duchenne muscular dystrophy, and vascular disorders, positioning them amongst the important therapeutic targets in these diseases (Karlstad et al., 2012; Leung et al., 2008; Putney, 2011; Ruhle and Trebak, 2013; Spinelli and Trebak, 2016; Tian et al., 2016; Xu et al., 2015; Zhang and Trebak, 2011). In both cardiac and vascular myocytes, SOCCs exhibit sensitivity to an array of deleterious factors including redox stress, hyperglycemia, hypoxia, and acidosis which modify and/or disrupt SOCE pathways as an early event in diseases such as diabetic angiopathy, atherosclerosis and hypertension(Freichel et al., 2001; Groschner et al., 2017; Nakayama et al., 2006; Spinelli and Trebak, 2016). Investigations into the molecular regulation of SOCC has provided support for their role in cardiomyocyte function and for their pathophysiological role in cardiac hypertrophy, as well as in ischemia reperfusion-induced Ca2+ overload(Collins et al., 2013; Hulot et al., 2011; Luoet al., 2012).The involvement of SOCC in cardiovascular biology has also been explored and the potential for the development of therapy associated with the sechannels in apoptosis, hypertrophy, and arrhythmias has been suggested (Inoue et al., 2006; Watanabe et al., 2008). It has also been documented that disturbance in cardiovascular SOCC is of pathological significance, and thus therapeutic targeting of these channels has emerged as a favourable strategy for the treatment of cardiovascular diseases (Spinelli and Trebak, 2016).

3.2 Store-operated Ca2+ entry pathways

Two major functional molecular components of SOCC have been identified, namely: (i) Stromal interaction molecule (STIM), which serves as the ER/SR Ca2+ sensor; and (ii) Orai protein, which acts as a pore-forming channelin the plasma membrane(Cahalan, 2009; Fahrner et al., 2013; Liou et al., 2005; Putney, 2011; Shaw and Feske, 2012). There are three human Orai proteins-Orai 1, Orai 2 and Orai 3-as well as two human STIM proteins-STIM1 and STIM 2 (Hogan and Rao, 2015). The ER/SR Ca2+ sensors, STIM 1 and 2, differentially regulate and control the gating of plasma membrane Ca2+ release-activated Ca2+ channels (CRAC) in many cells (Hogan and Rao, 2015). STIM1 and STIM2 are identical in their overall structure, with a N-terminal domain in the ER/SR lumen, a single transmembrane segment anchoring the protein in ER, and a C-terminal cytoplasmic domain (Soboloff et al., 2012). The functional regions of this protein in the ER/SR lumen are: (i) the Ca2+-sensing sterile α motif domain; (ii) the cytoplasmic region that both stabilizes inactive STIM and, upon Ca2+ store depletion, transmits the activating conformational change; (iii) the STIM-Orai activated region and CRAC activation region domain that recruits and gates Orai channels; (iv) the polybasic tail that interacts with plasma membrane phosphoinositides; and (v) a full length dimer in unstimulated cell(Hogan and Rao, 2015; Muik et al., 2011; Yang et al., 2012; Yuan et al., 2009; Zhou et al., 2010). Sensing Ca2+ in the ER/SR lumen and networking store depletion to other proteins (including Orai channels) are the key functions of STIM. Upon depletion of IP3- or RyR- sensitive Ca2+ stores, the localized sensor in STIM directly couples with plasma membrane Orai channels mediating Ca2+ influx (Bose, 2017; Lur et al., 2009). This is considered to be the basis STIM-Orai signaling. The overall underlying mechanism for this SOCC activation and inactivation cycle is shown in Fig. 1. It should be noted that it is upon ER/SR Ca2+ depletion that STIM proteins oligomerize into multiple punctae and relocate to the proximity of the plasma membrane and form ER/SR-plasma membrane junctions.

Figure 1.

Schematic mechanism of store-operated Ca2+ entry pathways. ER-endoplasmic reticulum; SR-sarcoplasmic reticulum; PM-plasma membrane; SOCC-store-operated Ca2+ channel;STIM-Stromal interaction molecule.

Diminution of Ca2+ stores also enhances binding of microtubule and end-binding protein to STIM oligomers, which provides guidance toward the plasma membrane (Chen et al., 2013; Honnappa et al., 2009; Tsai et al., 2014).The SOCC Orai proteins translocate to the STIM-containing ER/SR plasma membrane junctions following Ca2+ store depletion and open to mediate Ca2+entry by direct physical interaction between the cytoplasmic C-terminal coiled-coil domain of Orai 1 and the cytoplasmic C-terminal CRAC domain/STIM-Orai activating region of STIM (Chen et al., 2016; Fahrner et al., 2014; Frischauf et al., 2009; Wu et al., 2006). On the other hand, the STIM1 binding proteins, SARAF, golli and ORMDL3, play an important regulatory role in modulating Ca2+entry as well as in the inactivation of Ca2+entry to prevent Ca2+ overload (Albarran et al., 2016a; Carreras-Sureda et al., 2013; Lopez et al., 2016; Palty et al., 2012; Walsh et al., 2010). Two types of SOCC pore-forming subunits including Ca2+ release-activated Ca2+ modulator (CRACM1), Orai 1/2, and transient receptor potential canonical (TRPC) channels have been identified (Desai et al., 2015; Lopez et al., 2016; Smani et al., 2016; Vaeth et al., 2017). However, it has been revealed that the depolarization-induced opening of L-type voltage dependent Ca2+ channels is inhibited by Ca2+ store depletion in a STIM1-dependent pathway (Berridge, 2002; Park et al., 2010; Wang et al., 2010). It may be noted that Orai 1 participate by co-localizing with STIM1 and L-type Ca2+ channels in ER/SR plasma membrane junctions after Ca2+ store depletion (Wang et al., 2010). While Orai, as well as TRPC channels, are opened, L-type Ca2+ channels are inhibited by the ER/SR Ca2+ store depletion by the same signaling mechanism acting as a switch between these two routes of Ca2+ entry. Thus, the STIM protein interacts directly with both SOCC and store-inhibited Ca2+ channels, whereas the Orai protein plays a crucial role in functioning as a SOCC itself (Berridge, 2002; Lee et al., 2010).

3.3 Store-operated Ca2+ channel activators

Any procedure that depletes the ER/SR Ca2+ stores can activate SOCC. Store emptying can be achieved by an increase of IP3 or other Ca2+ releasing signals, resulting in Ca2+ release from these stores. A growing number of studies has supported several such mechanisms, including: IP3 production in the cytosol or the stimulation of IP3 receptors; blockade of the SR Ca2+-ATPase pump (SERCA) using thapsigargin; increase of SR membrane permeability with Ca2+ ionophore (ionomycin); and dialyzation of the cytoplasm with Ca2+ chelators (EGTA or BAPTA) (DeHaven et al., 2009; Gordon et al., 2000; Lemonnier et al., 2006; Parekh and Putney, 2005; Putney, 2010). Mostly known as an inhibitor of SOCC at high concentrations, 2-aminoethyldiphenyl borinate (2-APB) in the 1-20 µM range has been shown to act as an activator of SOCC and enhance the SOCE(Ma et al., 2002; Prakriya and Lewis, 2001). Ionomycin and SERCA pump blockers, which usually cause a rise in cytoplasmic Ca2+ concentration due to Ca2+ store depletion, could also open Ca2+-activated cation channels (Parekh and Putney, 2005).However, accumulating evidence suggests that two direct activators of SOCC - a peptide representing the Orai interacting domain of STIM1(Kawasaki et al., 2009; Muik et al., 2009; Park et al., 2009; Yuan et al., 2009) and the Ca2+ influx factor(Bolotina and Csutora, 2005) isolated from Ca2+ store-depleted cells-are capable of activating SOCC in the absence of store depletion, acting independently of STIM1 (Bolotina, 2008).

Recognizing their involvement in SOCC mechanism, both STIM and Orai proteins, which sense and respond to Ca2+ store depletion, can be modulated. Several agents have been identified to modify STIM-Orai signaling in SOCC (Hogan and Rao, 2015). These include: the ER/SR resident protein STIM-activating enhancer (STIMATE) and the cytosolic protein CRAC regulator 2A (CRACR2A), which stabilize the STIM1-Orai1 signalplex; and septins, which support recruitment and translocation of STIM1 in ER/SR plasma membrane junctions (Albarran et al., 2016b; Jing et al., 2015; Lopez et al., 2016; Sharma et al., 2013; Srikanth et al., 2010; Wilson et al., 2015). In addition, lysophospholipid products of Ca2+ independent phospholipase A2 (iPLA2) has been suggested as an auxiliary component that co-activates and mediates STIM1-Orai 1 interaction (Bolotina, 2008). However, it has been shown that STIM1, through its Orai-activating domain, interacts with and gates Orai channels through a direct protein-protein interaction (Avila-Medina et al., 2018). Also, STIM1, independently of its essential role in SOCC activation, is altered by diverse stimuli such as oxidation, temperature, hypoxia, and acidification (Hooper et al., 2013).

3.4 Store-operated Ca2+ channel antagonists

The discovery of several molecular components of SOCC (Fig. 2) has created the opportunity to develop drugs to, for example, block the pore of the Orai channel or modulate the activity of STIM1 (Putney, 2010). A number of agents that block SOCC have been suggested, either direct channel inhibitors or mechanism-based inhibitors (Table 2) (Chen et al., 2013; Chung et al., 1994; Clementi and Meldolesi, 1996; DeHaven et al., 2008; Franzius et al., 1994; Gregory et al., 2001; Holowka et al., 2014; Hoth and Penner, 1993; Irvine, 1990; Iwasaki et al., 2001; Ohana et al., 2009; Peinelt et al., 2008; Putney, 2001, 2010; Rodland et al., 1997; Smyth et al., 2008; Sweeney et al., 2009; Tian et al., 2016; Xu et al., 2015). Traditionally, ianthanides Gd3+ (gadolinium) and La3+ (lanthanum) were utilized extensively for blocking SOCC (Bird et al., 2008; Broad et al., 1999; Putney, 2001; Xu et al., 2015). STIM1 fails to activate SOCC when microtubule cytoskeleton disconnects from the ER/SR to form the mitotic spindle during mitosis; this mitotic destruction of STIM1 function is linked to specific phosphorylation sites in the C terminus (Smyth et al., 2008). Due to the association of STIM1 with microtubules within the ER/SR, microtubule reorganization with drugs such as nocodazole or colchicine has been proposed for STIM1-induced SOCE blockade.

10.31083/j.rcm.2019.03.522.t0002 Table 2 Pharmacological inhibitors of Store-operated Ca2+ Entry (SOCE)
SOCE Inhibitors Blockade Site References
A. Lanthanides: La3+ (lanthanum) ; Gd3+ (gadolinium) SOCC Orai Hoth and Penner, 1993; Tian et al.,2016; Xu et al., 2015
B. Imidazole compounds: SKF-96365;SKF 96465; econazole; miconazole Thapsigargin mediated SOCC Chung et al.,1994; Franzius et al.,1994; Hoth and Penner, 1993
C. Diphenylboronate compounds: 2-Aminoethyldiphenyl borate (2-APB) : 2-APB analogs- DPB162-AE and DPB163-AE (2-APB derivatives) Translocation of STIM puncta and STIM/Orai binding; Partially Orai channel DeHaven et al., 2008; Gregory et al., 2001; Iwasaki et al., 2001; Peinelt et al., 2008
D. Pyrazolecompounds: Bis (trifluoromethyl) (BTP1, BTP2 and BTP3) SOCE Sweeney et al., 2009
E. ML-9 Myosin light chain kinase (MLCK) Coalescence of STIM1 into puncta Smyth et al., 2008
F. Diethylstilbestrol (DES 18) -a synthetic estrogen agonist SOCE in mast cells and vascular smooth muscle cells Hoth and Penner, 1993; Ohana et al., 2009
G. Carboxyamidotriazole (CAI) Ca2+ dependent inactivation CRAC Rodland et al.,1997
H. RO2959 Human TCR mediated SOCE Chen et al., 2013
I. Linoleic acid: 18-C polyunsaturated fatty acid (PUFA) SOCE by affecting STIM1 oligomerization and subsequent STIM1/ORAI1 coupling Holowka et al., 2014
Figure 2.

Effects of store-operated Ca2+ entry antagonists. ER-endoplasmic reticulum; SR-sarcoplasmic reticulum; PM - Plasma membrane; STIM-Stromal interaction molecule.

For inhibition of the development of STIM1 into puncta as well as ER/SR-plasma membrane translocation, the myosin light chain kinase inhibitor (ML-9) has been suggested (Smyth et al., 2008). 2-aminoethyldiphenyl borinate (2-APB), in the 25-100 µM range transiently blocks the activation of Orai 1-mediated Ca2+ entry (DeHaven et al., 2008; Lis et al., 2007; Schindl et al., 2008; Zhang et al., 2008), as well as the ER/SR-plasma membrane translocation of STIM1 into puncta (DeHaven et al., 2008; Peinelt et al., 2008). This may further slow down the kinetic for Orai 2 blockade (DeHaven et al., 2008; Lis et al., 2007). Series analogs of 2-APB, DPB162-AE and DPB163-AE, have similarly shown their effectiveness as SOCC blockers(Goto et al., 2010). In addition, 3, 5-bis-trifluoromethyl pyrazole derivatives (BTP1, BTP2 and BTP3) in T-cells (Ishikawa et al., 2003; Zitt et al., 2004), SKF-96365 inhibited thapsigargin-induced SOCE in Jurkat T cells (Chung et al., 1994; Tian et al., 2016), and carboxyamidotriazole (CAI) in HEK293 cells (Hussain et al., 2003; Kohn et al., 2001), have been shown to exert SOCC blockade. Recently, linoleic acid, an 18-C polyunsaturated fatty acid (PUFA) commonly known as omega-6 has been reported to inhibit antigen- or thapsigargin-mediated SOCE in mast cells by affecting STIM1 oligomerization and subsequent STIM1/Orai 1 coupling (Holowka et al., 2014). Another important selective Orai 1 inhibitor RO2959 has been suggested to potently inhibit human T-cell receptors (TCR)-mediated SOCE, T-cell proliferation, cytokine production, and gene expression (Chen et al., 2013). SOCC blockers such as 2-APB and SKF-96365 have also been shown to inhibit lysophosphatidic acid-induced increase in intracellular Ca2+ and protein synthesis in vascular smooth muscle (Xu et al., 2005, 2015). Although various SOCC inhibitors (Table 2) including lanthanides and 2-APB have been demonstrated to block transient receptor Ca2+ channels and IP3 receptors, these inhibitors have a limited clinical use due either to their toxic effect or poor specificity (Johnson and Trebak, 2019; Tian et al, 2016).

4.Store-operated Ca2+ Channels in Cardiovascular Function

It is evident that Ca2+ entry through SOCC plays a critical role in regulating cardiovascular function in both health and disease. Since cardiovascular function is mainly determined by the coordinated interaction of cardiac muscle, vascular smooth muscle, and endothelium, this section is focused on the role of SOCC in the function of cardiomyocytes, vascular myocytes, and endothelial cells.

4.1. Role of SOCC in cardiomyocyte function

Several investigators have shown the presence of SOCC and their different components, such as STIM and Orai proteins, in cardiomyocytes, as well as demonstrating their role in Ca2+ entry and the augmentation of ventricular contractility (Bootman and Rietdorf, 2017; Mohl et al., 2011; Rosenberg et al., 2019). Not only are these channels involved in refilling SR Ca2+ stores, but they have also been shown to maintain resting levels of Ca2+ in cardiomyocytes (Huang et al., 2006; Touchberry et al., 2011). Different molecules of SOCC including Orai 1, Orai 3 and STIM1 were found to regulate normal and hypertrophic growth in cardiomyocytes (Ohba et al., 2009; Saliba et al., 2015; Voelkers et al., 2010). While Orai protein deficiency has been shown to lead the development of heart failure (Volkers et al., 2012), the elevation of STIM1 protein in the heart was associated with Ca2+ handling abnormalities and cardiomyopathy (Correll et al., 2015). These observations provide evidence that SOCE affects cardiomyocyte function in both health and disease.

4.2. Role of SOCC in vascular smooth muscle cell function

As in other striated muscles, the presence of SOCC and its components such as STIM and Orai proteins has been observed in different types of smooth muscle cells (Feldman et al., 2017; Golovina et al., 2001; Shibata et al., 2019; Sweeney et al., 2002; Yan et al., 2019). These channels not only promote SOCE but also increase the level of intracellular Ca2+ and refill SR/ER Ca2+ stores in the smooth muscle cells. Hypoxia was found to upregulate SOCC in pulmonary artery smooth muscle cells by increasing the protein expression of STIM1/STIM2 and Orai 1/Orai 2, as well as inducing pulmonary vascular remodeling and vasoconstriction (He et al., 2018; Jernigan et al., 2012). Silencing of STIM1 attenuated the hypoxia-induced pulmonary artery smooth muscle cell proliferation through depression in the SOCE, and accordingly STIM1 was suggested to play an important role in pulmonary hypertension (Hou et al., 2013). On the other hand, an increased level of STIM2 and Orai 2 proteins was shown to contribute in the transition of pulmonary arterial smooth cells from contractile to proliferative phenotypes (Fernandez et al., 2015). Thus, upregulation of SOCC in different types of smooth muscle cells is thought to play a critical role in the development of hypertension.

4.3. Role of SOCC in endothelial cell function

By virtue of its ability to produce endothelin-1 (ET-1), a potent vasoconstrictor, and nitric oxide (NO), a potent vasodilator, defects in endothelial cell function are known to play an important role in the development of hypertension. Since the formation and release of ET-1 and NO are dependent upon the entry of Ca2+ in the endothelium, various investigators have demonstrated the participation of SOCC and its components in the function of endothelial cells (Giachini et al., 2009; Molnar et al., 2016; Peng et al., 2010; Wilson et al., 2015; Zhou et al., 2016). Hypoxia has been shown to increase SOCE by stimulating SOCC in pulmonary artery endothelial cells, and is known to be involvedin vascular remodeling and hypertension (Fantozzi et al., 2003; Paffett et al., 2007). In fact, reduced SOCE in pulmonary endothelial cells due to chronic hypoxia was found to be due to reduced membrane cholesterol (Zhang et al., 2018a), known to exert a tight control of endothelial cell function (Zhang et al., 2018b). Different interventions have also been demonstrated to profoundly impact upon SOCC in endothelial cells with respect to the production and release of both NO and ET-1 (Adapala et al., 2011; Boittin et al., 2008; Graier et al., 1990; Kaczara et al., 2018; Qu et al., 2017). Both NO and ET-1 have been shown to affect SOCE in smooth muscle cells and are known to be intimately involved in the control of blood pressure (Ansari et al., 2004; Chuang et al., 2012; Clementi, 1998; Jernigan et al., 2006).

5.Store-operated Ca2+ Channels in Hypertension

The physiological and pathophysiological significance of SOCC-associated mechanisms, as well as molecular and cellular pathways for their regulation, are considered to form the basis of SOCE inhibitors as a potential therapy for several human diseases, including hypertension (Avila-Medina et al., 2018; Collins et al., 2013). There is growing evidence to suggest that Ca2+ enters through SOCC in VSMC (Barlow et al., 2006; Domínguez-Rodriguez et al., 2012; Park et al., 2008; Rodríguez-Moyano et al., 2013) and that disorder of these channels are associated with the development of hypertension (Tanwar et al., 2017). The activation of SOCC in VSMC (either by vasoactive agonists or by SERCA inhibition) maintains Ca2+ homeostasis for proper Ca2+ signaling, it is the defective regulation of intracellular Ca2+ that plays a crucial role in the genesis of hypertension (Albert and Large, 2002; Jackson, 2000; Manjarres et al., 2010). It has been suggested that SOCC inhibitors (e.g. rapamycin) may serve as promising drugs for the treatment of hypertension(Houssaini et al., 2013). The beneficial actions of SKF 96365 (SK) on changes in blood pressure, cell proliferation and intracellular Ca2+ have been shown, and the use of a SOCC inhibitor SK in combination with Ca2+ channel antagonist verapamil has been shown to exert additive effects on BP as well as on intracellular Ca2+ (Xu et al., 2005, 2015).

5.1 Store-operated Ca2+ pathways - STIM1/Orai 1 in Hypertension Treatment

Essential regulators of intracellular Ca2+ homeostasis, including STIM and Orai proteins, are key contributors to Ca2+ signaling mechanisms that support their role in cardiovascular disease (Ruhle and Trebak, 2013). The increase in cytosolic Ca2+ via SOCC mediated by STIM and Orai proteins activates a variety of signaling cascades to regulate several cellular functions. Conversely, their dysregulation promotes several pathophysiologies, including atherosclerosis, arterial stenosis, thrombosis, and hypertension (Ruhle and Trebak, 2013; Tanwar et al., 2017). There is a strong correlation between hypertension and enhanced STIM and/or Orai protein expression (Kassan et al., 2016; Pulina et al., 2013; Spinelli and Trebak, 2016; Zhang and Trebak, 2011). It has been proposed that augmented Ca2+ influx through SOCE pathways, STIM1/Orai 1, may contribute in potentiating vascular reactivity (Giachini et al., 2009, 2012) and vascular tone and force generation (Goulopoulou and Webb, 2014; Kitazono et al., 2002; Tanwar et al., 2017) (Fig. 3). The treatment of aortic rings in male spontaneously hypertensive rats with high concentrations of SOCC blockers (either 2-APB and Gd3+, or with STIM1 and Orai1 antibodies) was found to result in a reduction of spontaneous tone and force generation to levels found in normotensive rats. However, it has been observed that higher concentrations of SOCC inhibitors are likely to affect other ion channels, such as ER/SR Ca2+ release channels and pumps, which in turn can result in SOCE inhibition (Cortes et al., 1997; Giachini et al., 2009). A similar study using aortic tissue from rats undergoing chronic ethanol consumption for 30 days showed increases in SOCE, systolic blood pressure, and STIM1 protein expression (Souza Bomfim et al., 2017). Since STIM1 is a key regulator of Ca2+ homeostasis for supporting communications between the ER/SR and the plasma membrane, its upregulation could lead to direct or indirect ER/SR stress, as well as dictating the development of cardiovascular complications in hypertensive conditions. Wild-type mice infused with angiotensin II have been shown to develop hypertension, cardiac hypertrophy, perivascular fibrosis and endothelial dysfunction associated with enhanced STIM1 expression in heart and blood vessels, in study by Kassan et al.(2016) . The same authors reported that STIM1 up-regulation during angiotensin II-induced hypertension was associated with enhanced ER/SR stress through TGF-β and NADPH oxidase-dependent pathways. Accordingly, it has been suggested that smooth muscle STIM1 not only plays a vital role in the pathogenesis of hypertension and associated cardiovascular pathologies, but also signifies a promising therapeutic target in these diseases (Kassan et al., 2016). This is also supported by studies targeting STIM- and/or Orai-mediated SOCE by different inhibitors for the treatment of hypertension (Ruhle and Trebak, 2013).

Figure 3.

Involvement of Store-operated Ca2+ entry in the development of hypertension. STIM-Stromal interaction molecule; ER-endoplasmic reticulum; SR-sarcoplasmic reticulum.

6.Conclusion

Hypertension is a prevalent risk factor of cardiovascular diseases and thus a major cause of mortality worldwide. It is treatable with medications as well as lifestyle changes. Most of the classical antihypertensive agents, including Ca2+ channel antagonists, ameliorate hypertension by lowering intracellular Ca2+ within vascular smooth muscle cells. Although existing medications for hypertension are generally well tolerated, they are known to have side effects. It has been observed that β-blockers may aggravate asthma and decrease heart rate, ACE inhibitors may lead to dry cough, L-type Ca2+ channel antagonists may cause leg swelling, and diuretics may increase urination and leg cramps. Thus, efforts are being made to improve drug therapy for hypertension. A new class of antihypertensive agent for the prevention of Ca2+ entry through SOCC is being developed, but some SOCC inhibitors, including lanthanides, are limited in their clinical use due to either toxicity or poor specificity. The ongoing challenge of developing specific inhibitors of SOCC involving STIM and Orai proteins as targets is to be met only by further developing our understanding of STIM/Orai mechanisms, interactions, and channel gating, with the overarching goal of improving upon currently available drug treatments.

Acknowledgments

Infrastructure support for this project was provided by the St. Boniface Hospital Research Foundation, Winnipeg, Canada.

Conflict of Interest

The authors declare no conflict of interest.

References
[1]
Abdullaev, I. F., Bisaillon, J. M., Potier, M., Gonzalez, J. C., Motiani, R. K. and Trebak, M. (2008) Stim1 and Orai1 mediate CRAC currents and store-operated calcium entry important for endothelial cell proliferation. Circulation Research 103, 1289-1299. 6caef27754b5a0a61069cf74ddb771bfhttp%3A%2F%2Fpubmedcentralcanada.ca%2Fpmcc%2Farticles%2FPMC2682347%2F
[2]
Abernethy, D. R. and Schwartz, J. B. (1999) Calcium-antagonist drugs. New England Journal of Medicine 341, 1447-1457.
[3]
Adapala, R. K., Talasila, P. K., Bratz, I. N., Zhang, D. X., Suzuki, M., Meszaros, J. G. and Thodeti, C. K. (2011) PKCα mediates acetylcholine-induced activation of TRPV4-dependent calcium influx in endothelial cells. American Journal of Physiology-Heart and Circulatory 301, H757-H765. 9220575ec16a8266e9d63ce7808c3f14http%3A%2F%2Feuropepmc.org%2Farticles%2FPMC3302191%2F
[4]
Albarran, L., Lopez, J. J., Amor, N. B., Martin-Cano, F. E., Berna-Erro, A., Smani, T., Salido, G. M. and Rosado, J. A. (2016b) Dynamic interaction of SARAF with STIM1 and Orai1 to modulate store-operated calcium entry. Scientific Reports 6, 244-252. f23054fe097870dc1cc547dd3b3ea373http%3A%2F%2Feuropepmc.org%2Farticles%2FPMC4828706%2F
[5]
Albarran, L., Lopez, J. J., Woodard, G. E., Salido, G. M. and Rosado, J. A. (2016a) Store-operated Ca 2+ entry-associated regulatory factor (SARAF) plays an important role in the regulation of arachidonate-regulated Ca2+ (ARC) channels . Journal of Biological Chemistry 291, 6982-6988. e305997a05a8c2bca84399bb68cfefb6http%3A%2F%2Fpubmedcentralcanada.ca%2Fpmcc%2Farticles%2FPMC4807282%2F
[6]
Albert, A. P. and Large, W. A. (2002) A Ca2+-permeable non-selective cation channel activated by depletion of internal Ca2+ stores in single rabbit portal vein myocytes . Journal of Physiology 538, 717-728.
[7]
Ansari, H. R., Kaddour-Djebbar, I. and Abdel-Latif, A. A. (2004) Involvement of Ca2+ channels in endothelin-1-induced MAP kinase phosphorylation, myosin light chain phosphorylation and contraction in rabbit iris sphincter smooth muscle . Cellular Signalling 16, 609-619.
[8]
Arakawa, N., Sakaue, M., Yokoyama, I., Hashimoto, H., Koyama, Y., Baba, A. and Matsuda, T. (2000) KB-R7943 inhibits store-operated Ca2+ entry in cultured neurons and astrocytes . Biochemical and Biophysical Research Communications 279, 354-357. 7f810cfe0bd9fe98fb92cb61f7e047edhttp%3A%2F%2Fwww.sciencedirect.com%2Fscience%2Farticle%2Fpii%2FS0006291X00939688
[9]
Avila-Medina, J., Mayoral-Gonzalez, I., Dominguez-Rodriguez, A., Gallardo-Castillo, I., Ribas, J., Ordoñez, A., Rosado, J. A. and Smani, T. (2018) The complex role of store operated calcium entry pathways and related proteins in the function of cardiac, skeletal and vascular smooth muscle cells. Frontiers in Physiology 9, 1-14. 965d9276d975e5ac1dc159a7840a36fdhttp%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpmc%2Farticles%2FPMC5872157%2F
[10]
Bangalore, R., Baindur, N., Rutledge, A., Triggle, D. J. and Kass, R. S. (1994) L-type calcium channels: Asymmetrical intramembrane binding domain revealed by variable length, permanently charged 1,4-dihydropyridines. Molecular Pharmacology 46, 660-666. 8ae74e3e7ef23678cf3663acc486c8b8http%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F7969044
[11]
Barlow, C. A., Rose, P., Pulver-Kaste, R. A. and Lounsbury, K. M. (2006) Excitation-transcription coupling in smooth muscle. Journal of Physiology 570, 59-64. 6c9640966102ae26452fcb5f8dc79723http%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F16614312
[12]
Bartekova, M., Ducas , A. and Dhalla, N. S. (2015) Role of vascular endothelium in hypertension, atherosclerosis and peripheral arterial disease. Integrative Diabetes and Cardiovascular Diseases 1, 1-15.
[13]
Bean, B. P. and McDonough, S. I. (2010) Calcium Channels. Chichester, England: John Wiley & Sons Ltd.
[14]
Beevers, G., Lip, G. Y. and O’Brien, E. (2001) ABC of hypertension: The pathophysiology of hypertension. British Medical Journal 322, 912-916. 9692c7e07114835ff71f24331ae9e505http%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F11302910
[15]
Berra-Romani, R., Mazzocco-Spezzia, A., Pulina, M. V. and Golovina, V. A. (2008) Ca2+ handling is altered when arterial myocytes progress from a contractile to a proliferative phenotype in culture . American Journal of Physiology-Cell Physiology 295, C779-C790. aa77e8265497590742c1f9fcf5ecafddhttp%3A%2F%2Fcardiovascres.oxfordjournals.org%2Flookup%2Fijlink%3FlinkType%3DABST%26amp%3BjournalCode%3Dajpcell%26amp%3Bresid%3D295%2F3%2FC779%26amp%3Batom%3D%252Fcardiovascres%252F100%252F2%252F297.atom
[16]
Berridge, M. J., Lipp, P. and Bootman, M. D. (2000) The versatility and universality of calcium signalling. Nature Reviews Molecular Cell Biology 1, 11-21. 7ea7729f1529dbe73469c12a00a9af12http%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F11413485%3Fdopt%3DAbstract
[17]
Berridge, M. J. (2002) The endoplasmic reticulum: a multifunctional signaling organelle. Cell Calcium 32, 235-249. 4941031c331efd303e9d6439705f6cf6http%3A%2F%2Fwww.europepmc.org%2Fabstract%2Fmed%2F12543086
[18]
Bers, D. M. (2008) Calcium cycling and signaling in cardiac myocytes. Annual Review of Physiology 70, 23-49.
[19]
Bhatt, D. L., Kandzari, D. E., O'Neill, W. W., D'Agostino. R., Flack, J. M., Katzen, B. T., Leon, M. B., Liu, M., Mauri, L., Negoita, M., Cohen, S. A., Oparil, S., Rocha-Singh, K., Townsend. R. R. and Bakris, G. L. (2014) A controlled trial of renal denervation for resistant hypertension. New England Journal of Medicine 370, 1393-1401.
[20]
Bird, G. S., DeHaven, W. I., Smyth, J. T. and Putney, J. W. Jr. (2008) Methods for studying store-operated calcium entry. Methods in Enzymology 46, 204-212. 377ea8bea82650fc0ebb3c6546a1ec03http%3A%2F%2Fwww.sciencedirect.com%2Fscience%2Farticle%2Fpii%2FS1046202308001588
[21]
Boittin, F. X., Gribi, F., Serir, K. and Beny, J. L. (2008) Ca 2+-independent PLA2 controls endothelial store-operated Ca2+ entry and vascular tone in intact aorta . American Journal of Physiology-Heart and Circulatory 295, H2466-H2474.
[22]
Bolotina, V. M. and Csutora, P. (2005) CIF and other mysteries of the store-operated Ca 2+-entry pathway . Trends in Biochemical Sciences 30, 378-387.
[23]
Bolotina, V. M. (2008) Orai, STIM1 and iPLA2 beta: a view from a different perspective. Journal of Physiology 586, 3035-3042. 56a65607c0b7dc48d625596ce43add28http%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F18499724
[24]
Bootman, M., D. and Rietdorf , K. (2017) Tissue Specificity: Store-operated Ca 2+ entry in cardiac myocytes . Advances in Experimental Medicine and Biology 993, 363-387. 666a6bd42fd3a67e5227082a7c25096ahttp%3A%2F%2Flink.springer.com%2Fchapter%2F10.1007%2F978-3-319-57732-6_19
[25]
Bose, D. D. (2017) Store-operated calcium entry channels: potential role in cardiac. In, Malhotra A. and Shivani Soni (eds). Emerging Applications, Perspectives, and Discoveries in Cardiovascular Research (pp. 53-72). Hershey, Pennsylvania, USA: IGI Global Disseminator of Knowledge.
[26]
Broad, L. M., Cannon, T. R and Taylor, C. W. (1999) A non-capacitative pathway activated by arachidonic acid is the major Ca 2+ entry mechanism in rat A7r5 smooth muscle cells stimulated with low concentrations of vasopressin . Journal of Physiology 517, 121-134. 09eeea99e5344ad2822dd060c90a4455http%3A%2F%2Feuropepmc.org%2Fabstract%2FMED%2F10226154
[27]
Cahalan, M. D. (2009) STIMulating store-operated Ca 2+ entry . Nature Cell Biology 11, 669-677. 250eddc41ed0fbb986e443cd931bcb66http%3A%2F%2Fcardiovascres.oxfordjournals.org%2Flookup%2Fexternal-ref%3Faccess_num%3D19488056%26amp%3Blink_type%3DMED%26amp%3Batom%3D%252Fcardiovascres%252F93%252F2%252F360.atom
[28]
Cain, A. E and Khalil, R. A. (2002) Pathophysiology of essential hypertension: role of the pump, the vessel, and the kidney. Seminars in Nephrology 22, 3-16. 1c89764ac49c0c1bb34cd7aa804fcbc9http%3A%2F%2Feuropepmc.org%2Fabstract%2FMED%2F11785064
[29]
Carafoli , E. (2003) The calcium-signalling saga: tap water and protein crystals. Nature Reviews Molecular Cell Biology 4, 326-332. 84755775bd7f2e4f7ac09279f0ab5713http%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F12671655
[30]
Carreras-Sureda, G., Cantero-Recasens, F., Rubio-Moscardo, K., Kiefer, K., Peinelt, C., Niemeyer, B. A., Valverde, M. A. and Vicente, R. (2013) ORMDL3 modulates store-operated calcium entry and lymphocyte activation. Human Molecular Genetics 22, 519-530. 8617c5e0edbc8e5fc260ba101187080ehttp%3A%2F%2Fhmg.oxfordjournals.org%2Fcontent%2F22%2F3%2F519.full
[31]
Carretero, O. A and Oparil , S. (2000) Essential hypertension. Part I: definition and etiology. Circulation 101, 329-335. ad59f321b8fbcb6f423222fb69a14ffdhttp%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F10645931%3Fdopt%3DAbstract
[32]
Chaturvedi , N. (2003) Ethnic differences in cardiovascular disease. Heart 89, 681-686. 15d5dcd4023ae83b03104367fde33dedhttp%3A%2F%2Fheart.bmj.com%2Fcontent%2F89%2F6%2F681
[33]
Chen, G., Panicker, S., Lau, K. Y., Apparsundaram, S., Patel, V. A., Chen, S. L., Soto, R., Jung, J. K., Ravindran, P., Okuhara, D., Bohnert, G., Che, Q., Rao, P. E., Allard, J. D., Badi, L., Bitter, H. M., Nunn, P. A., Narula, S. K. and DeMartino, J. A. (2013) Characterization of a novel CRAC inhibitor that potently blocks human T cell activation and effector functions. Molecular Immunology 54, 355-367. 5db9ffbc14eb9b9d8f77b989e50185a5http%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F23357789
[34]
Chen, Y. F., Hsu, K. F and Shen, M. R. ( 2016) The store-operated Ca 2+ entry-mediated signaling is important for cancer spread . Biochimica et Biophysica Acta 863, 1427-1435. eaae001a17557d63f2d9caeee3ba3974http%3A%2F%2Feuropepmc.org%2Fabstract%2FMED%2F26643254
[35]
Chen, Y. T., Chen, Y. F., Chiu, W. T., Liu, K. Y., Liu, Y. L., Chang, J. Y., Chang, H. C. and Shen, M. R. (2013) Microtubule-associated histone deacetylase 6 supports the calcium store sensor STIM1 in mediating malignant cell behaviors. Cancer Research 73, 4500-4509. d3872c12c44b24f7e9875f456b5ea68ahttp%3A%2F%2Feuropepmc.org%2Fabstract%2Fmed%2F23698468
[36]
Chiong, J. R., Aronow, W. S., Khan, I. A., Nair, C. K., Vijayaraghavan, K., Dart, R. A., Behrenbeck, T. R. and Geraci, S. A. (2008) Secondary hypertension: current diagnosis and treatment. International Journal of Cardiology 124, 6-21. 5b611f3389df01562d2eb9c95292a2d4http%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F17462751
[37]
Chobanian, A. V., Bakris, G. L., Black, H. R., Cushman, W. C., Green, L. A., Izzo, J. L. Jr., Jones, D. W., Materson, B. J., Oparil, S., Wright, J. T. Jr. and Roccella, E. J. (2003) The seventh report of the joint national committee on prevention, detection, evaluation, and treatment of high blood pressure: The JNC 7 report. Journal of the American Medical Association 289, 2560-2572.
[38]
Chuang, T. Y., Au, L. C., Wang, L. C., Ho, L. T., Yang, D. M. and Juan, C. C. (2012) Potential effect of resistin on the ET-1-increased reactions of blood pressure in rats and Ca2+ signaling in vascular smooth muscle cells . Journal of Cellular Physiology 7, 1610-1618.
[39]
Chung, S. C., Mcdonald, T. V and Gardner , P. (1994) Inhibition by SK&F 96365 of Ca2+ current, IL-2 production and activation in T lymphocytes . British Journal of Pharmacology 113, 861-868.
[40]
Clementi , E. and Meldolesi , J. (1996) Pharmacological and functional properties of voltage-independent Ca 2+ channels . Cell Calcium 19, 269-279. 3ad8b57eaefdf859ad871ed6ffe64b8fhttp%3A%2F%2Fwww.sciencedirect.com%2Fscience%2Farticle%2Fpii%2FS0143416096900688
[41]
Clementi, E. ( 1998) Role of nitric oxide and its intracellular signalling pathways in the control of Ca 2+ homeostasis . Biochemical Pharmacology 55, 713-718. f9881bafb9a90db8f4967a1db3e8a7adhttp%3A%2F%2Fwww.sciencedirect.com%2Fscience%2Farticle%2Fpii%2FS0006295297003754
[42]
Collins, H. E., Zhu-Mauldin, X., Marchase, R. B. and Chatham, J. C. (2013) STIM1/Orai1-mediated SOCE: current perspectives and potential roles in cardiac function and pathology. American Journal of Physiology-Heart and Circulatory 305, H446-H458. eb72bd380dc8d23a99a95fbade784e6fhttp%3A%2F%2Fcardiovascres.oxfordjournals.org%2Flookup%2Fijlink%3FlinkType%3DABST%26amp%3BjournalCode%3Dajpheart%26amp%3Bresid%3D305%2F4%2FH446%26amp%3Batom%3D%252Fcardiovascres%252F111%252F1%252F94.atom
[43]
Colovina, V. A. (1999) Cell proliferation in associated with enhanced capacitative Ca2+ entry in human arterial myocytes . American Journal of Physiology-Cell Physiology 277, C343-C349. 4062888cdefca119f728837491deb974http%3A%2F%2Feuropepmc.org%2Fabstract%2FMED%2F10444411
[44]
Correll, R. N., Goonasekera, S. A., van Berlo, J. H., Burr, A. R., Accornero, F., Zhang, H., Makarewich, C. A., York, A. J., Sargent, M. A., Chen, X., Houser, S. R. and Molkentin, J. D. (2015) STIM1 elevation in the heart results in aberrant Ca²⁺ handling and cardiomyopathy. Journal of Molecular and Cellular Cardiology 87, 38-47. 1192fd7f2befa0f9bf60bbcd7ea46a4bhttp%3A%2F%2Fpubmedcentralcanada.ca%2Fpmcc%2Farticles%2FPMC4637225%2F
[45]
, S. F., Lemos, V. S and Stoclet, J. C. (1997) Alterations in calcium stores in aortic myocytes from spontaneously hypertensive rats. Hypertension 29, 1322-1328. 84003245531fd935e0ff7365fbdedc49http%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F9180636
[46]
DeHaven, W. I., Jones, B. F., Petranka, J. G., Smyth, J. T., Tomita, T., Bird, G. S. and Putney, J. W. Jr. (2009) TRPC channels function independently of STIM1 and Orai1. Journal of Physiology 587, 2275-2298. 93d195bbb7ed4f6ab6d85ca96567625fhttp%3A%2F%2Feuropepmc.org%2Fabstract%2FMED%2F19332491
[47]
DeHaven, W. I., Smyth, J. T., Boyles, R. R., Bird, G. S. and Putney, J. W. Jr. (2008) Complex actions of 2-aminoethyldiphenyl borate on store-operated calcium entry. Journal of Biological Chemistry 283, 9265-19273. 6a9d2d5bf2c166c68462fe6e501cd96ahttp%3A%2F%2Feuropepmc.org%2Farticles%2FPMC2443677
[48]
Desai, P. N., Zhang, X., Wu, S., Janoshazi, A., Bolimuntha, S., Putney, J. W. and Trebak, M. (2015) Multiple types of calcium channels arising from alternative translation initiation of the Orai1 message. Science Signaling 8, 1-26. f0c08da74c625be1916a8f9210c8fe7bhttp%3A%2F%2Feuropepmc.org%2Farticles%2FPMC4583604%2F
[49]
Dhalla, N. S., Pierce, G. N., Panagia, V., Singal, P. K. and Beamish, R. E. (1982) Calcium movements in relation to heart function. Basic Research in Cardiology 77, 117-139. 49666951ffd84310cddaf3e3fa85b32bhttp%3A%2F%2Fcardiovascres.oxfordjournals.org%2Flookup%2Fexternal-ref%3Faccess_num%3D10.1007%2FBF01908167%26amp%3Blink_type%3DDOI
[50]
Dhalla, N. S., Ziegelhoffer , A. and Harrow, J. A. (1977) Regulatory role of membrane systems in heart function. Canadian Journal of Physiology and Pharmacology 55, 1211-1234. 27bcee62ea37a29901a6fd8bcbd510a0http%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F23210
[51]
Dilmac, N., Hilliard , N. and Hockerman, G. H. (2003) Molecular determinants of Ca2+ potentiation of diltiazem block and Ca 2+-dependent inactivation in the pore region of cav1.2 . Molecular Pharmacology 64, 491-501.
[52]
Dominguez-Rodriguez, A., Diaz, I., Rodriguez-Moyano, M., Calderon-Sanchez, E., Rosado, J. A., Ordonez, A. and Smani, T. (2012) Urotensin-II signaling mechanism in rat coronary artery: role of STIM1 and Orai1-dependent store operated calcium influx in vasoconstriction. Arteriosclerosis, Thrombosis, and Vascular Biology 32, 1325-1332. f2dc5d6e9ce70f2443e4d769d36d099fhttp%3A%2F%2Fcardiovascres.oxfordjournals.org%2Flookup%2Fijlink%3FlinkType%3DABST%26amp%3BjournalCode%3Datvbaha%26amp%3Bresid%3D32%2F5%2F1325%26amp%3Batom%3D%252Fcardiovascres%252F100%252F2%252F297.atom
[53]
Dorans, K. S., Mills, K. T., Liu, Y. and He, J. (2018) Trends in prevalence and control of hypertension according to the 2017 American College of Cardiology/American Heart Association (ACC/AHA) Guideline. Journal of the American Heart Association 7, 1-11. 80d7c8e758864254751c3701f5e56b0ehttp%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpmc%2Farticles%2FPMC6015372%2F
[54]
Eid, A. H., El-Yazbi, A. F., Zouein, F., Arredouani, A., Ouhtit, A., Rahman, M. M., Zayed, H., Pintus, G. and Abou-Saleh, H. (2018) Inositol 1,4,5-trisphosphate receptors in hypertension. Frontiers in Physiology 9, 1-12. 5a354d2de479b4d494cdf913165f2c85http%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpmc%2Farticles%2FPMC6071574%2F
[55]
Fahrner, M., Derler, I., Jardin, I. and Romanin, C. 1. (2013) The STIM1/Orai signaling machinery. Channels 7, 330-343. c3302bca27c4597df31b280fe5d12ef6http%3A%2F%2Fpubmedcentralcanada.ca%2Fpmcc%2Farticles%2FPMC3913757%2F
[56]
Fahrner, M., Muik, M., Schindl, R., Butorac, C., Stathopulos, P., Zheng, L., Jardin, I., Ikura, M. and Romanin, C. (2014) A coiled-coil clamp controls both conformation and clustering of stromal interaction molecule 1 (STIM1). Journal of Biological Chemistry 289, 33231-33244. f1de3541609654048044380cf5a4a49chttp%3A%2F%2Feuropepmc.org%2Fabstract%2Fmed%2F25342749
[57]
Fantozzi, I., Zhang, S., Platoshyn, O., Remillard, C. V., Cowling, R. T. and Yuan, J. X. (2003) Hypoxia increases AP-1 binding activity by enhancing capacitative Ca2+ entry in human pulmonary artery endothelial cells . American Journal of Physiology-Lung Cellular and Molecular Physiology 285, L1233-L1245. 95cd7abbec4f9f133c87641f1007ae2dhttp%3A%2F%2Fcardiovascres.oxfordjournals.org%2Flookup%2Fijlink%3FlinkType%3DABST%26amp%3BjournalCode%3Dajplung%26amp%3Bresid%3D285%2F6%2FL1233%26amp%3Batom%3D%252Fcardiovascres%252F91%252F4%252F677.atom
[58]
Feldman, C. H., Grotegut, C. A and Rosenberg, P. B. (2017) The role of STIM1 and SOCE in smooth muscle contractility. Cell Calcium 63, 60-65. dca7e78faa3405ee86bba6ac024e4e19http%3A%2F%2Feuropepmc.org%2Fabstract%2FMED%2F28372809
[59]
Fernandez, R. A., Wan, J., Song, S., Smith, K. A., Gu, Y., Tauseef, M., Tang, H., Makino, A., Mehta, D. and Yuan, J. X. (2015) Upregulated expression of STIM2, TRPC6, and Orai2 contributes to the transition of pulmonary arterial smooth muscle cells from a contractile to proliferative phenotype. American Journal of Physiology-Cell Physiology 308, C581-C593.
[60]
Fleckenstein , A. (1977) Specific pharmacology of calcium in myocardium, cardiac pacemakers, and vascular smooth muscle. Annual Review of Pharmacology and Toxicology 17, 149-166. 457af83d8376a3b6ac4adc0ad0e2fd9bhttp%3A%2F%2Fwww.annualreviews.org%2Fdoi%2Fabs%2F10.1146%2Fannurev.pa.17.040177.001053
[61]
Forouzanfar, M. H., Liu, P., Roth, G. A., Ng, M., Biryukov, S., Marczak, L., Alexander, L., Estep, K., Hassen Abate, K., Akinyemiju, T. F., et al. (2017) Global burden of hypertension and systolic blood pressure of at least 110 to 115 mm Hg, 1990-2015. Journal of the American Medical Association 317, 165-182. 596e13f9076d2e1843d5e5cfc11fb8a7http%3A%2F%2Fbmjopen.bmj.com%2Flookup%2Fexternal-ref%3Faccess_num%3Ddoi%3A10.1001%2Fjama.2016.19043%26amp%3Blink_type%3DDOI
[62]
Franzius, D., Hoth , M. and Penner , R. (1994) Non-specific effects of calcium entry antagonists in mast cells. Pflugers Archiv: European Journal of Physiology 428, 433-438. 4d677fddc7d41346cf0e4a7696739d5fhttp%3A%2F%2Feuropepmc.org%2Fabstract%2FMED%2F7838664
[63]
Freichel, M., Suh, S. H., Pfeifer, A., Schweig, U., Trost, C., Weissgerber, P., Biel, M., Philipp, S., Freise, D., Droogmans, G., Hofmann, F., Flockerzi, V. and Nilius, B. (2001) Lack of an endothelial store-operated Ca2+ current impairs agonist-dependent vasorelaxation in TRP4-/- mice . Nature Cell Biology 3, 121-127. 94ac72fb82115d499ec99f3b0ed17f2bhttp%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F11175743
[64]
Frischauf, I., Muik, M., Derler, I., Bergsmann, J., Fahrner, M., Schindl, R., Groschner, K. and Romanin, C. (2009) Molecular determinants of the coupling between STIM1 and Orai channels, differential activation of Orai1-3 channels by a STIM1 coiled-coil mutant. Journal of Biological Chemistry 284, 21696-21706. 178f4fec49acdb383822c8a3104cc57ahttp%3A%2F%2Feuropepmc.org%2Farticles%2FPMC2755892%2F
[65]
Fritze, O., Romero, B., Schleicher, M., Jacob, M. P., Oh, D. Y., Starcher, B., Schenke-Layland, K., Bujan, J. and Stock, U. A. (2012) Age-related changes in the elastic tissue of the human aorta. Journal of vascular Research 49, 77-86. 8acac5ec856c2c68c7f5d13f3d0ef4b8http%3A%2F%2Feuropepmc.org%2Fabstract%2FMED%2F2178615
[66]
Gates, P. E., Strain, W. D and Shore, A. C. (2009) Human endothelial function and microvascular ageing. Experimental Physiology 94, 311-316. d7998f367a5dc2f2c383f9a168437b82http%3A%2F%2Fnew.med.wanfangdata.com.cn%2FPaper%2FDetail%3Fid%3DPeriodicalPaper_JJ026010006
[67]
Giachini, F. R., Chiao, C. W., Carneiro, F. S., Lima, V. V., Carneiro, Z. N., Dorrance, A. M., Tostes, R. C. and Webb, R. C. (2009) Increased activation of stromal interaction molecule-1/Orai-1 in aorta from hypertensive rats: a novel insight into vascular dysfunction. Hypertension 53, 409-416. e7bb50728d2d0fb9b113566f9a9a2031http%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F19075091
[68]
Giachini, F. R., Lima, V. V., Filgueira, F. P., Dorrance, A. M., Carvalho, M. H., Fortes, Z. B., Webb, R. C. and Tostes, R. C. (2012) STIM1/Orai1 contributes to sex differences in vascular responses to calcium in spontaneously hypertensive rats. Clinical Science 122, 215-226.
[69]
Giachini, F. R., Webb, R. C and Tostes, R. C. (2009) STIM and Orai proteins: players in sexual differences in hypertension-associated vascular dysfunction. Clinical Science 118, 391-396.
[70]
Godfraind , T. (2017) Discovery and development of calcium channel blockers. Frontiers in Pharmacology 8, 1-25.
[71]
Golovina, V. A., Platoshyn, O., Bailey, C. L., Wang, J., Limsuwan, A., Sweeney, M., Rubin, L. J. and Yuan, J. X. (2001) Upregulated TRP and enhanced capacitative Ca 2+ entry in human pulmonary artery myocytes during proliferation . American Journal of Physiology-Heart and Circulatory 280, H746-H755. 0784c3682d09828fe444b93f0454e779http%3A%2F%2Fcardiovascres.oxfordjournals.org%2Flookup%2Fijlink%3FlinkType%3DABST%26amp%3BjournalCode%3Dajpheart%26amp%3Bresid%3D280%2F2%2FH746%26amp%3Batom%3D%252Fcardiovascres%252F89%252F2%252F282.atom
[72]
Gong, L., Zhang, W., Zhu, Y., Zhu, J., Kong, D., Page, V., Ghadirian, P., LeLorier, J. and Hamet, P. (1996) Shanghai trial of nifedipine in the elderly (STONE). Journal of Hypertension 14, 1237-1245. 79b9319cc113bca18bf6a3e82d3afd6fhttp%3A%2F%2Fwww.sciencedirect.com%2Fscience%2Farticle%2Fpii%2F089570619682139X
[73]
Gordon, A. M., Homsher , E. and Regnier , M. (2000) Regulation of contraction in striated muscle. Physiological Reviews 80, 853-924. df9825968f195d30814f56c601cf8a47http%3A%2F%2Fjmg.bmj.com%2Flookup%2Fijlink%3FlinkType%3DABST%26amp%3BjournalCode%3Dphysrev%26amp%3Bresid%3D80%2F2%2F853%26amp%3Batom%3D
[74]
Goto, J, Suzuki, A. Z., Ozaki, S., Matsumoto, N., Nakamura, T., Ebisui, E., Fleig, A., Penner, R. and Mikoshiba, K. (2010) Two novel 2-aminoethyl diphenylborinate (2-APB) analogues differentially activate and inhibit store-operated Ca2+ entry via STIM proteins . Cell Calcium 47, 1-10. ad7f4502b30271dc1fab19e21e5e1f1chttp%3A%2F%2Feuropepmc.org%2Farticles%2FPMC2905153
[75]
Goulopoulou , S. and Webb, R. C. (2014) Symphony of vascular contraction: how smooth muscle cells lose harmony to signal increased vascular resistance in hypertension. Hypertension 63, 33-39. 95d3ba31770c38be718b9e30c83210e7http%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F24470463
[76]
Graier, W. F., Schmidt , K. and Kukovetz, W. R. . (1990) Effect of sodium fluoride on cytosolic free Ca2+ concentrations and cGMP-levels in endothelial cells . Cellular Signalling 2, 369-375. d8c2e1dd66852843ecc6c0cac44668b6http%3A%2F%2Feuropepmc.org%2Fabstract%2Fmed%2F2174691
[77]
Green, D. J., Swart, A., Exterkate, A., Naylor, L. H., Black, M. A., Cable, N. T. and Thijssen, D. H. (2010) Impact of age, sex and exercise on brachial and popliteal artery remodelling in humans. Atherosclerosis 210, 525-530.
[78]
Gregory, R. B., Rychkov , G. and Barritt, G. J. (2001) Evidence that 2-aminoethyl diphenylborate is a novel inhibitor of store-operated Ca 2+ channels in liver cells and acts through a mechanism which does not involve inositol trisphosphate receptors . Biochemical Journal 354, 285-290.
[79]
Groschner, K., Shrestha , N. and Fameli , N. (2017) Cardiovascular and hemostatic disorders, SOCE in cardiovascular cells- Emerging targets for therapeutic intervention. Advances in Experimental Medicine and Biology 993, 473-503. 471eae20789032d3fc2f04aa621f1778http%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F28900929
[80]
Guibert, C., Ducret , T. and Savineau, J. P. (2008) Voltage independent calcium influx in smooth muscle. Progress in Biophysics and Molecular Biology 98, 10-23. 491d053850188c5feca1c13667d4fbefhttp%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F18614208
[81]
Harada, K., Nomura, M., Nishikado, A., Uehara, K., Nakaya, Y. and Ito, S. (2003) Clinical efficacy of efonidipine hydrochloride, a T-type calcium channel inhibitor, on sympathetic activities. Circulation Journal 67, 139-145. 19ae5deb862722614a5bb2784caabfcehttp%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F12547996
[82]
He, X., Song, S., Ayon, R. J., Black, S. M., Makino, A., Wier, W. G., Zang, W. J. and Yuan, J. X. (2018) Hypoxia selectively upregulates cation channels and increases cytosolic [Ca2+] in pulmonary, but not coronary, arterial smooth muscle cells . American Journal of Physiology-Cell Physiology 314, C504-C517. f29b34b4160b99a68fa23ebe8bc27736http%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F29351410
[83]
Hockerman, G. H., Peterson, B. Z., Johnson, B. D. and Catterall, W. A. (1997) Molecular determinants of drug binding and action on L-type calcium channels. Annual Review of Pharmacology and Toxicology 37, 361-396. d6690a4bea142ead5902afcb6ecc6f91http%3A%2F%2Feuropace.oxfordjournals.org%2Fexternal-ref%3Faccess_num%3D9131258%26amp%3Blink_type%3DMED
[84]
Hofmann, F., Lacinova , L. and Klugbauer , N. (1999) Voltage-dependent calcium channels: From structure to function. Reviews of Physiology, Biochemistry and Pharmacology 139, 33-87. 65a33cb3a0b8cedf90f76c87163f5348http%3A%2F%2Fonlinelibrary.wiley.com%2Fresolve%2Freference%2FPMED%3Fid%3D10453692
[85]
Hogan, P. G. and Rao, A. (2015) Store-operated calcium entry: mechanisms and modulation. Biochemical and Biophysical Research Communications 460, 40-49. 53f537f888e92d81a77982d2b433487ehttp%3A%2F%2Feuropepmc.org%2Fabstract%2FMED%2F25998732
[86]
Holowka, D., Korzeniowski, M. K., Bryant, K. L. and Baird, B. (2014) Polyunsaturated fatty acids inhibit stimulated coupling between the ER Ca2+ sensor STIM1 and the Ca2+ channel protein Orai1 in a process that correlates with inhibition of stimulated STIM1 oligomerization . Biochimica et Biophysica Acta 1841, 1210-1216. 5339ba96cdc227bfa8d1ab1de507e363http%3A%2F%2Feuropepmc.org%2Fabstract%2Fmed%2F24769339
[87]
Honnappa, S., Gouveia, S. M., Weisbrich, A., Damberger, F. F., Bhavesh, N. S., Jawhari, H., Grigoriev, I., van Rijssel, F. J., Buey, R. M., Lawera, A., Jelesarov, I., Winkler, F. K., Wuthrich, K., Akhmanova, A. and Steinmetz, M. O. (2009) An EB1-binding motif acts as a microtubule tip localization signal. Cell 138, 366-376. 07e007fb30d9fef9e196651ddd6c6fa4http%3A%2F%2Fwww.cell.com%2Fabstract%2FS0092-8674%2809%2900638-2
[88]
Hooper, R., Samakai, E., Kedra, J. and Soboloff, J. (2013) Multifaceted roles of STIM proteins. Pflugers Archiv: European Journal of Physiology 465, 1383-1396.
[89]
Hoth , M. and Penner , R. (1993) Calcium release-activated calcium current in rat mast cells. Journal of Physiology 465, 359-386. 9c4d1d72dc9cd3094f9d5b9429cd5432http%3A%2F%2Feuropepmc.org%2Fabstract%2Fmed%2F8229840
[90]
Hou, X., Chen, J., Luo, Y., Liu, F., Xu, G. andGao, Y. (2013) Silencing of STIM1 attenuates hypoxia-induced PASMCs proliferation via inhibition of the SOC/Ca2+/NFAT pathway . Respiratory Research 14, 1-10. 5e5c07b6cc4f3b11e36327ec161bec30http%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F23289723
[91]
Houssaini, A., Abid, S., Mouraret, N., Wan, F., Rideau, D., Saker, M., Marcos, E., Tissot, C. M., Dubois-Rande, J. L., Amsellem, V. and Adnot, S. (2013) Rapamycin reverses pulmonary artery smooth muscle cell proliferation in pulmonary hypertension. American Journal of Respiratory Cell and Molecular Biology 48, 568-577.
[92]
Huang, Y., Lu, M. Q., Li, H., Xu, C., Yi, S. H. and Chen, G. H. (2006) Occurrence of cGMP/nitric oxide-sensitive store-operated calcium entry in fibroblasts and its effect on matrix metalloproteinase secretion. World Journal of Gastroenterology 12, 5483-5489. e603be34f0994bd9e088d4d0e95d8caehttp%3A%2F%2Feuropepmc.org%2Farticles%2FPMC4088230
[93]
Hulot, J. S., Fauconnier, J., Ramanujam, D., Chaanine, A., Aubart, F., Sassi, Y., Merkle, S., Cazorla, O., Ouille, A., Dupuis, M., Hadri, L., Jeong, D., Muhlstedt, S., Schmitt, J., Braun, A., Benard, L., Saliba, Y., Laggerbauer, B., Nieswandt, B., Lacampagne, A., Hajjar, R. J., Lompre, A. M. and Engelhardt, S. (2011) Critical role for stromal interaction molecule 1 in cardiac hypertrophy. Circulation 124, 796-805.
[94]
Hussain, M. M., Kotz, H., Minasian, L., Premkumar, A., Sarosy, G., Reed, E., Zhai, S., Steinberg, S. M., Raggio, M., Oliver, V. K., Figg, W. D. and Kohn, E. C. (2003) Phase II trial of carboxyamidotriazole in patients with relapsed epithelial ovarian cancer. Journal of Clinical Oncology 21, 4356-4363. 46f95fdd58b42c7a07a2fd07f8c6071chttp%3A%2F%2Feuropepmc.org%2Fabstract%2FMED%2F14645425
[95]
Inoue, R., Jensen, L. J., Shi, J., Morita, H., Nishida, M., Honda, A. and Ito, Y. (2006) Transient receptor potential channels in cardiovascular function and disease. Circulation Research 99, 119-131. 7f02ae7bc8bf5ce1cfda29ff67f27f0ehttp%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F16857972
[96]
Irvine, R. F. (1990) ‘Quantal’ Ca 2+ release and the control of Ca 2+ entry by inositol phosphates-a possible mechanism . Federation of European Biochemical Societies Letters 263, 5-9. f9e4d1c227cf99a5522c1f68eb424116http%3A%2F%2Fwww.tandfonline.com%2Fservlet%2Flinkout%3Fsuffix%3DCIT0013%26amp%3Bdbid%3D8%26amp%3Bdoi%3D10.3109%252F10799899509045222%26amp%3Bkey%3D2185036
[97]
Ishikawa, J., Ohga, K., Yoshino, T., Takezawa, R., Ichikawa, A., Kubota, H. and Yamada, T. (2003) A pyrazole derivative, YM-58483, potently inhibits store-operated sustained Ca 2+ influx and IL-2 production in T lymphocytes . Journal of Immunology 170, 4441-4449. f6e252bdd10da741d645615c58301d66http%3A%2F%2Fonlinelibrary.wiley.com%2Fresolve%2Freference%2FPMED%3Fid%3D12707319
[98]
Iwasaki, H., Mori, Y., Hara, Y., Uchida, K., Zhou, H. and Mikoshiba, K. (2001) 2-aminoethoxydiphenyl borate (2-APB) inhibits capacitative calcium entry independently of the function of inositol 1,4,5-trisphosphate receptors. Receptors and Channels 7, 429-439. b40e2264549a3d60d7cb7d5ad8264631http%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F11918346%2F
[99]
Jackson, W. F. (2000) Ion channels and vascular tone. Hypertension 35, 173-178.
[100]
Jernigan, N. L., Broughton, B. R., Walker, B. R. and Resta, T. C. (2006) Impaired NO-dependent inhibition of store- and receptor-operated calcium entry in pulmonary vascular smooth muscle after chronic hypoxia. American Journal of Physiology-Lung Cellular and Molecular Physiology 290, L517-L525. 16c645d4580037e9fd7695e165cd2720http%3A%2F%2Feuropepmc.org%2Fabstract%2FMED%2F16243900
[101]
Jernigan, N. L., Herbert, L. M., Walker, B. R. and Resta, T. C. (2012) Chronic hypoxia upregulates pulmonary arterial ASIC1: A novel mechanism of enhanced store-operated Ca2+ entry and receptor-dependent vasoconstriction . American Journal of Physiology-Cell Physiology 302, C931-C940. 3ad9e30f9ccfd0f7cc77a23d2ce3a2e3http%3A%2F%2Feuropepmc.org%2Farticles%2FPMC3311238%2F
[102]
Jing, J., He, L., Sun, A., Quintana, A., Ding, Y., Ma, G., Tan, P., Liang, X., Zheng, X., Chen, L., Shi, X., Zhang, S. L., Zhong, L., Huang, Y., Dong, M. Q., Walker, C. L., Hogan, P. G., Wang, Y. and Zhou, Y. (2015) Proteomic mapping of ER-PM junctions identifies STIMATE as a regulator of Ca2+ influx . Nature Cell Biology 17, 1339-1347.
[103]
Johnson , M. and Trebak , M. (2019) ORAI channels in cellular remodeling of cardiorespiratory disease. Cell Calcium 79, 1-10
[104]
Kaczara, P., Proniewski, B., Lovejoy, C., Kus, K., Motterlini, R., Abramov, A. Y. and Chlopicki, S. (2018) CORM-401 induces calcium signalling, NO increase and activation of pentose phosphate pathway in endothelial cells. Federation of European Biochemical Societies Journal 285, 1346-1358. 8d313ae2ad5ec135aec07ce9f11c40c1http%3A%2F%2Fonlinelibrary.wiley.com%2Fdoi%2F10.1111%2Ffebs.14411%2Ffull
[105]
Karlstad, J., Sun , Y. and Singh, B. B. (2012) Ca 2+ signaling: an outlook on the characterization of Ca2+ channels and their importance in cellular functions . Advances in Experimental Medicine and Biology 740, 143-157. fc37073c3e01b84b45b1d11cafa73ef3http%3A%2F%2Flink.springer.com%2Fchapter%2F10.1007%252F978-94-007-2888-2_6
[106]
Kassan, M., Ait-Aissa, K., Radwan, E., Mali, V., Haddox, S., Gabani., M., Zhang, W., Belmadani, S., Irani, K., Trebak, M. and Matrougui, K. (2016) Essential role of smooth muscle STIM1 in hypertension and cardiovascular dysfunction. Arteriosclerosis, Thrombosis, and Vascular Biology 36, 1900-1909. 9f482ceed16c8fbda29c6b58b244986fhttp%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F27470514
[107]
Kawasaki, T., Lange , I. and Feske , S. (2009) A minimal regulatory domain in the C terminus of STIM1 binds to and activates ORAI1 CRAC channels. Biochemical and Biophysical Research Communications 385, 49-54. 62df58da145c36ca834324beeecbfae3http%3A%2F%2Fwww.sciencedirect.com%2Fscience%2Farticle%2Fpii%2FS0006291X09009085
[108]
Khoury, S. R and Ratchford, E. V. (2018) Hypertension. Vascular Medicine 23, 293-297.
[109]
Kitazono, T., Ago, T., Kamouchi, M., Santa, N., Ooboshi, H., Fujishima, M. and Ibayashi, S. (2002) Increased activity of calcium channels and Rho-associated kinase in the basilar artery during chronic hypertension in vivo. Journal of Hypertension 20, 879-884. 51d642a0e2405c81c96ce2282ef69648http%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F12011648
[110]
Kohn, E. C., Reed, E., Sarosy, G. A., Minasian, L., Bauer, K. S., Bostick-Bruton, F., Kulpa, V., Fuse, E., Tompkins, A., Noone, M., Goldspiel, B., Pluda, J., Figg, W. D. and Liotta, L. A. (2001) A phase I trial of carboxyamido-triazole and paclitaxel for relapsed solid tumors: potential efficacy of the combination and demonstration of pharmacokinetic interaction. Clinical Cancer Research 7, 1600-1609.
[111]
Kuramoto , K. (1999) National intervention cooperative study in elderly hypertensives study group. Randomized doubleblind comparison of a calcium antagonist and a diuretic in elderly hypertensives. Hypertension 34, 1129-1133.
[112]
Kurokawa, J., Adachi-Akahane , S. and Nagao , T. (1997) 1,5-benzothiazepine binding domain is located on the extracellular side of the cardiac L-type Ca2+ channel . Molecular Pharmacology 51, 262-268. 17e9ed2829dda74ab10413f1f2c7332fhttp%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F9203631
[113]
Lambert, M., Capuano, V., Olschewsk, A., Sabourin, J., Nagaraj, C., Girerd, B., Weatherald, J., Humbert, M. and Antigny, F. (2018) Ion channels in pulmonary hypertension: A therapeutic interest. International Journal of Molecular Sciences 19, 1-49.
[114]
Lawes, C. M., Vander Hoorn, . . and Rodgers , A. (2008) Global burden of blood-pressure-related disease, 2001. Lancet 371, 1513-1518. b004dc3f8793b23b1a1f1e1c78880425http%3A%2F%2Fstatic.www.bmj.com%2Flookup%2Fexternal-ref%3Faccess_num%3D18456100%26amp%3Blink_type%3DMED%26amp%3Batom%3D%252Fbmj%252F342%252Fbmj.d442.atom
[115]
Lee, K. P., Yuan, J. P., Hong, J. H., So, I., Worley, P. F. and Muallem, S. (2010) An endoplasmic reticulum/plasma membrane junction: STIM1/Orai1/ TRPCs. Federation of European Biochemical Societies Letters 584, 2022-2027. 80369739549faa2d7d5ab419143dd5c4http%3A%2F%2Fonlinelibrary.wiley.com%2Fdoi%2F10.1016%2Fj.febslet.2009.11.078%2Ffull
[116]
Lemonnier, L., Trebak, M., Lievremont, J. P., Bird, G. S. and Putney, J. W. Jr. (2006) Protection of TRPC7 cation channels from calcium inhibition by closely associated SERCA pumps. American Societies for Experimental Biology Journal 20, 503-505. 06a9faf5e84e3503c11569bb928d6667http%3A%2F%2Fcardiovascres.oxfordjournals.org%2Flookup%2Fijlink%3FlinkType%3DABST%26amp%3BjournalCode%3Dfasebj%26amp%3Bresid%3D05-4714fjev1%26amp%3Batom%3D%252Fcardiovascres%252F92%252F2%252F189.atom
[117]
Leung, F. P., Yung, L. M., Yao, X., Laher, I. and Huang, Y. (2008) Store-operated calcium entry in vascular smooth muscle. British Journal of Pharmacology 153, 846-857. eab2088ce9a9810f19fc514b3396dec3http%3A%2F%2Fcpfd.cnki.com.cn%2FArticle%2FCPFDTOTAL-ZGYS200607001100.htm
[118]
Leung, Y. M and Kwan, C. Y. (1999) Current perspectives in the pharmacological studies of store-operated Ca2+ entry blockers . Japanese Journal of Pharmacology 81, 253-258. c32b98770a5ae8ae90748fc4778264c5http%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F10622212
[119]
Lewington, S., Clarke, R., Qizilbash, N., Peto, R. and Collins, R. (2002) Age-specific relevance of usual blood pressure to vascular mortality: A meta-analysis of individual data for one million adults in 61 prospective studies. Lancet 360, 1903-1913. dd7ea30724172e45d0fc6aca5b5298afhttp%3A%2F%2Fwww.sciencedirect.com%2Fscience%2Farticle%2Fpii%2FS0140673602119118
[120]
Lewis, R. S. (2011) Store-operated calcium channels: new perspectives on mechanism and function. Cold Spring Harbor Perspectives in Biology 3, 1-24. cab29cfa923d34f2dbcf36abcaeb719chttp%3A%2F%2Fabbs.oxfordjournals.org%2Fcgi%2Fijlink%3FlinkType%3DABST%26amp%3BjournalCode%3Dcshperspect%26amp%3Bresid%3D3%2F12%2Fa003970
[121]
Liou, J., Kim, M. L., Heo, W. D., Jones, J. T., Myers, J. W., Ferrell, J. E. Jr. and Meyer, T. (2005) STIM is a Ca 2+ sensor essential for Ca2+-store-depletion-triggered Ca 2+ influx . Current Biology 15, 1235-1241.
[122]
Lis, A., Peinelt, C., Beck, A., Parvez, S., Monteilh-Zoller, M., Fleig, A. and Penner, R. (2007) CRACM1, CRACM2, and CRACM3 are store-operated Ca2+ channels with distinct functional properties . Current Biology 17, 794-800. ce7a26e5894728a790b676c41e69ec29http%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F17442569
[123]
Liu, L., Wang, J. G., Gong, L., Liu, G. and Staessen, J. A. (1998) Comparison of active treatment and placebo in older chinese patients with isolated systolic hypertension. Systolic hypertension in China (Syst-China) collaborative group. Journal of Hypertension 16, 1823-1829. 1e850f6d025577fc324cb4d2fbc00a84http%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F9869017%3Fdopt%3DAbstract
[124]
Lopez, J. J., Albarran, L., Gomez, L. J., Smani, T., Salido, G. M. and Rosado, J. A. (2016) Molecular modulators of store-operated calcium entry. Biochimica et Biophysica Acta 1863, 2037-2043. 04dcdc612526f681d649c7451638abcbhttp%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F27130253
[125]
Luo, X., Hojayev, B., Jiang, N., Wang, Z. V., Tandan, S., Rakalin, A., Rothermel, B. A., Gillette, T. G. and Hill, J. A. (2012) STIM1-dependent store-operated Ca2+ entry is required for pathological cardiac hypertrophy . Journal of Molecular and Cellular Cardiology 52, 136-147. 46605157e2741938eeda237d0817fd92http%3A%2F%2Fpubmedcentralcanada.ca%2Fpmcc%2Farticles%2FPMC3247164%2F
[126]
Lur, G., Haynes, L. P., Prior, I. A., Gerasimenko, O. V., Feske, S., Petersen, O. H., Burgoyne, R. D. and Tepikin, A. V. (2009) Ribosome-free terminals of rough ER allow formation of STIM1 puncta and segregation of STIM1 from IP3 receptors. Current Biology 19, 1648-1653. 7b0c549f77b1e75a9b03bc1344e26210http%3A%2F%2Fwww.sciencedirect.com%2Fscience%2Farticle%2Fpii%2FS0960982209016029
[127]
Ma, H. T., Venkatachalam, K., Parys, J. B. and Gill, D. L. (2002) Modification of store-operated channel coupling and inositol trisphosphate receptor function by 2-aminoethoxydiphenyl borate in DT40 lymphocytes. Journal of Biological Chemistry 277, 6915-6922 2a7b5ffa363b5049b0fa796ffc782cf2http%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F11741932
[128]
Manjarres, I. M., Rodriguez-Garcia, A., Alonso, M. T. and Garcia-Sancho, J. (2010) The sarco/endoplasmic reticulum Ca2+ ATPase (SERCA) is the third element in capacitative calcium entry . Cell Calcium 47, 412-418. 21d268a733210bff7ed72ed32b3b34a5http%3A%2F%2Fwww.sciencedirect.com%2Fscience%2Farticle%2Fpii%2FS014341601000045X
[129]
Manolis, A. J., Kallistratos, M. S., Doumas, M., Pagoni, S. and Poulimenos, L. (2015) Recent advances in the management of resistant hypertension. Faculty of 1000 Prime Reports 7, 03. 5acddf1ca6075c97df90826bccee2929http%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F25705386
[130]
McFadzean , I. and Gibson , A. (2002) The developing relationship between receptor-operated and store-operated calcium channels in smooth muscle. British Journal of Pharmacology 135, 1-13 3fc7ec34e545ad4235668404ffc94bd7http%3A%2F%2Feuropepmc.org%2Farticles%2FPMC1573126
[131]
Mohl, M. C., Iismaa, S. E., Xiao, X. H., Friedrich, O., Wagner, S., Nikolova-Krstevski, V., Wu, J., Yu, Z. Y., Feneley, M., Fatkin, D., Allen, D. G. and Graham, R. M. (2011) Regulation of murine cardiac contractility by activation of α(1A)-adrenergic receptor-operated Ca2+ entry . Cardiovascular Research 91, 310-319. 3a2b9d7327662618459cae7697d34136http%3A%2F%2Fcardiovascres.oxfordjournals.org%2Fcontent%2F91%2F2%2F310
[132]
Molnar, T., Yarishkin, O., Iuso, A., Barabas, P., Jones, B., Marc, R. E., Phuong, T. T. and Krizaj, D. (2016) Store-operated calcium entry in Müller glia is controlled by synergistic activation of TRPC and Orai channels. Journal of Neuroscience 36, 3184-3198. dd297bb506357a2f7e3e6c6c121f670ehttp%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F26985029
[133]
Mrowka, R. (2019) Recent advances in hypertension research. Acta Physiologica 226, e13295.
[134]
Muik, M., Fahrner, M., Derler, I., Schindl, R., Bergsmann, J., Frischauf, I., Groschner, K. and Romanin, C. (2009) A cytosolic homomerization and a modulatory domain within STIM1 C terminus determine coupling to ORAI1 channels. Journal of Biological Chemistry 284, 8421-8426. a6bfffce8d78677afb26db429d1600behttp%3A%2F%2Feuropepmc.org%2Farticles%2FPMC2659200
[135]
Muik, M., Fahrner, M., Schindl, R., Stathopulos, P., Frischauf, I., Derler, I., Plenk, P., Lackner, B., Groschner, K., Ikura, M. and Romanin, C. (2011) STIM1 couples to ORAI1 via an intramolecular transition into an extended conformation European Molecular Biology Organization Journal 30, 1678-1689. 9dff92e3437dd193d7eb851f6797011dhttp%3A%2F%2Fwww.nature.com%2Femboj%2Fjournal%2Fv30%2Fn9%2Fabs%2Femboj201179a.html
[136]
Nakayama, H., Wilkin, B. J., Bodi, I. and Molkentin, J. D. (2006) Calcineurin-dependent cardiomyopathy is activated by TRPC in the adult mouse heart. Federation of American Societies for Experimental Biology Journal 20, 1660-1670. 132a72ac890a8e31d0c4f4b0d8dd7e66http%3A%2F%2Fbioinformatics.oxfordjournals.org%2Fcgi%2Fijlink%3FlinkType%3DABST%26amp%3BjournalCode%3Dfasebj%26amp%3Bresid%3D20%2F10%2F1660
[137]
Ohana, L., Newell, E. W., Stanley, E. F. and Schlichter, L. C. (2009) The Ca2+ release-activated Ca2+ current (ICRAC) mediates storeoperated Ca2+ entry in rat microglia . Channels 3, 129-139.
[138]
Ohba, T., Watanabe, H., Murakami, M., Sato, T., Ono, K. and Ito, H. (2009) Essential role of STIM1 in the development of cardiomyocyte hypertrophy. Biochemical and Biophysical Research Communications 389, 172-176. 76d0dfb6b6cb4497c987bfe63c61debehttp%3A%2F%2Fnew.med.wanfangdata.com.cn%2FPaper%2FDetail%3Fid%3DPeriodicalPaper_JJ0216263058
[139]
Oparil, S., Amin Zaman, M. and Calhoun, D. A., (2003) Pathogenesis of hypertension. Annals of Internal Medicine 139, 761-776.
[140]
Ozawa, Y., Hayashi , K. and Kobori , H. (2006) New generation calcium channel blockers in hypertensive treatment. Current Hypertension Reviews 2, 103-111.
[141]
Paffett, M. L., Naik, J. S., Resta, T. C. and Walker, B. R. (2007) Reduced store-operated Ca2+ entry in pulmonary endothelial cells from chronically hypoxic rats . American Journal of Physiology-Lung Cellular and Molecular Physiology 293, L1135-L1142. 7cfa80ecb6e64f549099e6dd71ae8abbhttp%3A%2F%2Feuropepmc.org%2Fabstract%2Fmed%2F17693482
[142]
Palty, R., Raveh, A., Kaminsky, I., Meller, R. and Reuveny, E. (2012) SARAF inactivates the store operated calcium entry machinery to prevent excess calcium refilling. Cell 149, 425-438. 3d84b4ec9406201307b69c85199d387bhttp%3A%2F%2Feuropepmc.org%2Fabstract%2FMED%2F22464749
[143]
Pang, Y., Hunton, D. L., Bounelis, P. and Marchase, R. B. (2002) Hyperglycemia inhibits capacitative calcium entry and hypertrophy in neonatal cardiomyocytes. Diabetes 51, 3461-3467. a9d854495a39c3c4bc4f6a4244282923http%3A%2F%2Feuropepmc.org%2Fabstract%2FMED%2F12453900
[144]
Parekh, A. B and Putney, J. W. (2005) Store-operated calcium channels. Physiological Reviews 85, 757-810.
[145]
Park, K. M., Trucillo, M., Serban, N., Cohen, R. A. and Bolotina, V. M. (2008) Role of iPLA2 and store-operated channels in agonist-induced Ca 2+ influx and constriction in cerebral, mesenteric, and carotid arteries . American Journal of Physiology-Heart and Circulatory 294, H1183-H1187. 0c96cbc46227fc6e089a48164d1feff4http%3A%2F%2Feuropepmc.org%2Fabstract%2FMED%2F18156193
[146]
Park, C. Y., Hoover, P. J ., Mullins, F. M., Bachhawat, P., Covington, E. D., Raunser, S., Walz, T., Garcia, K. C., Dolmetsch, R. E. and Lewis, R. S. (2009) STIM1 clusters and activates CRAC channels via direct binding of a cytosolic domain to Orai1. Cell 136, 876-890. 0f1e6169e4c45fe279c3c59abe15ee1dhttp%3A%2F%2Fwww.cell.com%2Fabstract%2FS0092-8674%2809%2900152-4%3Fscript%3Dtrue
[147]
Park, C. Y., Shcheglovitov , A. and Dolmetsch , R. (2010) The CRAC channel activator STIM1 binds and inhibits L-type voltage-gated calcium channels. Science 330, 101-105. f7e4dc646822e383eef0d2945e0ff24dhttp%3A%2F%2Fwww.jstor.org%2Fstable%2F40802978
[148]
Peinelt, C., Lis, A., Beck, A., Fleig, A. and Penner, R. (2008) 2-APB directly facilitates and indirectly inhibits STIM1-dependent gating of CRAC channels. Journal of Physiology 586, 3061-3073.
[149]
Peng, G., Lu, W., Li, X., Chen, Y., Zhong, N., Ran, P. and Wang, J. (2010) Expression of store-operated Ca 2+ entry and transient receptor potential canonical and vanilloid-related proteins in rat distal pulmonary venous smooth muscle . American Journal of Physiology-Lung Cellular and Molecular Physiology 299, L621-L630. a799a1e3bd4c6f9c614fe6d879d908a9http%3A%2F%2Feuropepmc.org%2Fabstract%2FMED%2F20693314
[150]
Potier, M., Gonzalez, J. C., Motiani, R. K., Abdullaev, I. F., Bisaillon, J. M., Singer, H. A. and Trebak, M. (2009) Evidence for STIM1- and Orai1-dependent store-operated calcium influx through ICRAC in vascular smooth muscle cells: role in proliferation and migration. Federation of American Societies for Experimental Biology Journal 23, 2425-2437. 8014effa8bb26fb44c58b2d8385bf566http%3A%2F%2Feuropepmc.org%2Fabstract%2Fmed%2F19364762
[151]
Prakriya , M. and Lewis, R. S. (2001) Potentiation and inhibition of Ca 2+ release-activated Ca 2+ channels by 2-aminoethyldiphenyl borate (2-APB) occurs independently of IP3 receptors . Journal of Physiology 536, 3-19. f1a59766df11538d54ba707c32b5dd3chttp%3A%2F%2Fonlinelibrary.wiley.com%2Fdoi%2F10.1111%2Fj.1469-7793.2001.t01-1-00003.x%2Fabstract
[152]
Prakriya , M. and Lewis, R. S. (2015) Store-operated calcium channels. Physiological Reviews 95, 1383-1436.
[153]
Pulina, M. V., Zulian, A., Baryshnikov, S. G., Linde, C. I., Karashima, E., Hamlyn, J. M., Ferrari, P., Blaustein, M. P. and Golovina, V. A. (2013) Cross talk between plasma membrane Na+/Ca 2+ exchanger-1 and TRPC/Orai-containing channels: key players in arterial hypertension . Advances in Experimental Medicine and Biology 961, 365-374. 1ffe6d9b09079885bf4e215ecf249086http%3A%2F%2Fpubmedcentralcanada.ca%2Fpmcc%2Farticles%2FPMC3857394%2F
[154]
Putney, J. W. (2009) Capacitative calcium entry: from concept to molecules. Immunological Reviews 231, 10-22. c91f9227aae02128b273a4e438791942http%3A%2F%2Fonlinelibrary.wiley.com%2Fdoi%2F10.1111%2Fj.1600-065X.2009.00810.x%2Ffull
[155]
Putney, J. W. (1986) A model for receptor-regulated calcium entry. Cell Calcium 7, 1-12. 5c48b284bd768d467b02bd3e46dbd314http%3A%2F%2Fwww.sciencedirect.com%2Fscience%2Farticle%2Fpii%2F0143416086900266
[156]
Putney, J. W. (1990) Capacitative calcium entry revisited. Cell Calcium 11, 611-624.
[157]
Putney, J. W. (2018) Forms and functions of store-operated calcium entry mediators, STIM and Orai. Advances in Biological Regulation 68, 88-96. ae1306db85b115319d4016855403ac42http%3A%2F%2Feuropepmc.org%2Fabstract%2FMED%2F29217255
[158]
Putney, J. W. (2001) Pharmacology of capacitative calcium entry. Molecular Interventions 1, 84-94. 4563121fc9efd763a5eee4955857f324http%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F14993328
[159]
Putney, J. W. (2010) Pharmacology of store-operated calcium channels. Molecular Interventions 10, 209-218. 1bb7fb7630ce220ed9fe8388b131d4d3http%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F20729487
[160]
Putney, J. W. (2011) The physiological function of store-operated calcium entry. Neurochemical Research 36, 1157-1165. 01a3dfd7b5ae2ac2b533a767f3cd15cbhttp%3A%2F%2Feuropepmc.org%2Farticles%2FPMC3112236%2F
[161]
Qu, Y. Y., Wang, L. M., Zhong, H., Liu, Y. M., Tang, N., Zhu, L. P., He, F. and Hu, Q. H. (2017) TRPC1 stimulates calcium-sensing receptor-induced store-operated Ca2+ entry and nitric oxide production in endothelial cells . Molecular Medicine Reports 16, 4613-4619. bc1f40be385b536e0dcfde37835f3801http%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F28791397
[162]
Remuzzi, G., Scheppati , A. and Ruggenenti , P. (2002) Clinical practice. Nephropathy in patients with type 2 diabetes. New England Journal of Medicine 346, 1145-1151. 20cf3904614ce1a4e1bbb96a00d92f2fhttp%3A%2F%2Fwww.bmj.com%2Flookup%2Fexternal-ref%3Faccess_num%3D11948275%26amp%3Blink_type%3DMED%26amp%3Batom%3D%252Fbmj%252F346%252Fbmj.e8525.atom
[163]
Rodland, K. D., Wersto, R. P., Hobson, S. and Kohn, E. C. (1997) Thapsigargin-induced gene expression in nonexcitable cells is dependent on calcium influx. Molecular Endocrinology 11, 281-291. 683045c1add16580665680f175d7209ahttp%3A%2F%2Feuropepmc.org%2Fabstract%2FMED%2F9058375
[164]
Rodriguez-Moyano, M., Diaz, I., Dionisio, N., Zhang, X., Avila-Medina, J., Calderon-Sanchez, E., Trebak, M., Rosado, J. A., Ordonez, A. and Smani, T. (2013) Urotensin-II promotes vascular smooth muscle cell proliferation through store-operated calcium entry and EGFR transactivation. Cardiovascular Research 100, 297-306. 1fe165547942797147e32bb69718abe4http%3A%2F%2Fcardiovascres.oxfordjournals.org%2Fcontent%2F100%2F2%2F297
[165]
Rosenberg, P., Katz , D. and Bryson , V. (2019) SOCE and STIM1 signaling in the heart: Timing and location matter. Cell Calcium 77, 20-28.
[166]
Ruhle , B. and Trebak , M. (2013) Emerging roles for native Orai Ca2+ channels in cardiovascular disease . Current Topics in Membranes 71, 209-235. c25ac972b00a604e27622670524d9c0ahttp%3A%2F%2Feuropepmc.org%2Fabstract%2Fmed%2F23890117
[167]
Saliba, Y., Keck, M., Marchand, A., Atassi, F., Ouille, A., Cazorla, O., Trebak, M., Pavoine, C., Lacampagne, A., Hulot, J. S., Fares, N., Fauconnier, J. and Lompre, A. M. (2015) Emergence of Orai3 activity during cardiac hypertrophy. Cardiovascular Research 105, 248-259.
[168]
Schindl, R., Bergsmann, J., Frischauf, I., Derler, I., Fahrner, M., Muik, M., Fritsch, R., Groschner, K. and Romanin, C. (2008) 2-Aminoethoxydiphenyl borate alters selectivity of Orai3 channels by increasing their pore size. Journal of Biological Chemistry 283, 20261-20267. e6f86f2aa9239c1293ea8b54b4c38784http%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F18499656
[169]
Scultety , S. and Tamaskovits , E. (1991) Effect of Ca 2+ antagonists on isolated rabbit detrusor muscle . Acta Physiologica Hungarica 77, 269-278. b5ac8147a5a97c283713b650e5a7d267http%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F1755331
[170]
Sharma, S., Quintana, A., Findlay, G. M., Mettlen, M., Baust, B., Jain, M., Nilsson, R., Rao, A.and Hogan, P. G. (2013) An siRNA screen for NFAT activation identifies septins as coordinators of store-operated Ca2+ entry . Nature 499, 238-242. 7572f4156ddb90c7e4f4785557528764http%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F23792561
[171]
Shaw, P. J and Feske , S. (2012) Physiological and pathophysiological functions of SOCE in the immune system. Frontiers in Bioscience 4, 2253-2268. 31f5d90d588451ec320de898d565c73ehttp%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F%3Fterm%3D22202035
[172]
Shibata, A., Uchida, K., Kodo, K., Miyauchi, T., Mikoshiba, K., Takahashi, T. and Yamagishi, H. (2019) Type 2 inositol 1,4,5-trisphosphate receptor inhibits the progression of pulmonary arterial hypertension via calcium signaling and apoptosis. Heart Vessels 34, 724-734. f94efdbd12b078629184209e65be63fdhttp%3A%2F%2Flink.springer.com%2Farticle%2F10.1007%2Fs00380-018-1304-4
[173]
Smani, T., Domínguez-Rodriguez, A., Callejo-Garcia, P., Rosado, J. A. and Avila-Medina, J. (2016) Phospholipase A2 as a molecular determinant of store-operated calcium entry. Advances in Experimental Medicine and Biology 898, 111-131. d906ce2e95a8e7f2cc7022a991a828f6http%3A%2F%2Feuropepmc.org%2Fabstract%2Fmed%2F27161227
[174]
Smyth, J. T., DeHaven, W. I., Bird, G. S. and Putney, J. W. Jr. (2008) Ca 2+-store dependent and-independent reversal of Stim1 localization and function . Journal of Cell Science 121, 762-772.
[175]
Soboloff, J., Rothberg, B. S., Madesh, M. and Gill, D. L. (2012) STIM proteins: dynamic calcium signal transducers. Nature Reviews Molecular Cell Biology 13, 549-565. 6a34e670c25511d6aca08f1194bc3cachttp%3A%2F%2Fpubmedcentralcanada.ca%2Fpmcc%2Farticles%2FPMC3458427%2F
[176]
Souza Bomfim, G. H., Mendez-Lopez, I., Arranz-Tagarro, J. A., Ferraz Carbonel, A. A., Roman-Campos, D., Padin, J. F., Garcia, A. G., Jurkiewicz, A. and Jurkiewicz, N. H. (2017) Functional upregulation of STIM-1/Orai-1-mediated Store-Operated Ca 2+ contributing to the hypertension development elicited by chronic EtOH consumption . Current Vascular Pharmacology 15, 265-281. 6ec00d467d451b4c6713394ac0ac316bhttp%3A%2F%2Feuropepmc.org%2Fabstract%2FMED%2F28155613
[177]
Spinelli, A. M and Trebak , M. (2016) Orai channel-mediated Ca2+ signals in vascular and airway smooth muscle . American Journal of Physiology-Cell Physiology 310, C402-C413. 22eaafeaf682301a7fb225dbe808bcf6http%3A%2F%2Feuropepmc.org%2Fabstract%2FMED%2F26718630
[178]
Srikanth, S., Jung, H. J., Kim, K. D., Souda, P., Whitelegge, J. and Gwack, Y. (2010) A novel EF-hand protein, CRACR2A, is a cytosolic Ca2+ sensor that stabilizes CRAC channels in T cells . Nature Cell Biology 12, 436-446. 3f0d1b8f6e0d48bb51a343d72054ef29http%3A%2F%2Feuropepmc.org%2Fabstract%2FMED%2F20418871
[179]
Staessen, J. A., Fagard, R., Thijs, L., Celis, H., Arabidze, G. G., Birkenhager, W. H., Bulpitt, C. J., de Leeuw, P. W., Dollery, C. T., Fletcher, A. E., Forette, F., Leonetti, G., Nachev, C., O'Brien, E. T., Rosenfeld, J., Rodicio, J. L., Tuomilehto, J. and Zanchetti, A. (1997) Randomised double-blind comparison of placebo and active treatment for older patients with isolated systolic hypertension. The systolic hypertension in Europe (Syst-Eur) trial investigators. Lancet 350, 757-764. 8e2630ee7a2af9cd7d7822b7ec6ede38http%3A%2F%2Feuropepmc.org%2Fabstract%2FMED%2F9297994
[180]
Stanaway, J. D., Afshin, A., Gakidou, E., Lim, S. S., Abate, D., Abate, K. H., Abbafati, C., Abbasi, N., Abbastabar, H., Abd-Allah, F., et al. (2018) Global, regional, and national comparative risk assessment of 84 behavioural, environmental and occupational, and metabolic risks or clusters of risks for 195 countries and territories, 1990-2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet 392, 1923-1994. e3bb0b668e50fbfb9606a43f38f36c85http%3A%2F%2Flra.le.ac.uk%2Fbitstream%2F2381%2F42126%2F6%2F1-s2.0-S0140673617323668-main.pdf
[181]
Stiber, J., Hawkins, A., Zhang, Z. S., Wang, S., Burch, J., Graham, V., Ward, C. C., Seth, M., Finch, E., Malouf, N., Williams, R. S., Eu, J. P. and Rosenberg, P. (2008) STIM1 signalling controls store-operated calcium entry required for development and contractile function in skeletal muscle. Nature Cell Biology 10, 688-697. 44d06f057a82fab4b2ca8f8b7d58837dhttp%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F18488020
[182]
Sweeney, M., Yu, Y., Platoshyn, O., Zhang, S., McDaniel, S. S. and Yuan, J. X. (2002) Inhibition of endogenous TRP1 decreases capacitative Ca2+ entry and attenuates pulmonary artery smooth muscle cell proliferation . American Journal of Physiology-Lung Cellular and Molecular Physiology 283, L144-L155. 4fa0ed13a0043ecb15811b395187166ehttp%3A%2F%2Fcardiovascres.oxfordjournals.org%2Flookup%2Fijlink%3FlinkType%3DABST%26amp%3BjournalCode%3Dajplung%26amp%3Bresid%3D283%2F1%2FL144%26amp%3Batom%3D%252Fcardiovascres%252F81%252F4%252F660.atom
[183]
Sweeney, Z. K., Minatti, A., Button, D. C. and Patrick, S. (2009) Small molecule inhibitors of store-operated calcium entry. ChemMedChem 4, 706-718.
[184]
Takahara, A., Sugiyama, A., Satoh, Y., Nakamura, Y. and Hashimoto, K. (2004) Cardiovascular effects of an L/N-type Ca2+ channel blocker cilnidipine assessed in the chronic atrioventricular conduction block dogs . Journal of Pharmacological Sciences 96, 219-223. 0c75ab92aca0a0ac10831c9c1ffa26ffhttp%3A%2F%2Feuropepmc.org%2Fabstract%2Fmed%2F15492462
[185]
Tanwar, J., Trebak , M. and Motiani, R. K. (2017) Cardiovascular and hemostatic disorders: Role of STIM and orai proteins in vascular disorders. Advances in Experimental Medicine and Biology 993, 425-452. e518917a367f16acb611c0ee4bab28behttp%3A%2F%2Flink.springer.com%2Fchapter%2F10.1007%2F978-3-319-57732-6_22
[186]
Tian, C., Du, L., Zhou, Y. and Li, M. (2016) Store-operated CRAC channel inhibitors: opportunities and challenges. Future Medicinal Chemistry 8, 817-832. fff224f0bfc11f7a9ddb5ba0d58859e4http%3A%2F%2Fpubmedcentralcanada.ca%2Fpmcc%2Farticles%2FPMC5558521%2F
[187]
Tocci, G., Battistoni, A., Passerini, J., Musumeci, M. B., Francia, P., Ferrucci, A. and Volpe, M. (2015) Calcium channel blockers and hypertension. Journal of Cardiovascular Pharmacology and Therapeutics 20, 121-130. 6063ca2b96fcacc23d3a77c1ecac4222http%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F25398848
[188]
Touchberry, C. D., Elmore, C. J., Nguyen, T. M., Andresen, J. J., Zhao, X., Orange, M., Weisleder, N., Brotto, M., Claycomb, W. C. and Wacker, M. J. (2011) Store-operated calcium entry is present in HL-1 cardiomyocytes and contributes to resting calcium. Biochemical and Biophysical Research Communications 416, 45-50. a4d3f14ca3d5fd5ef4528b22b249e499http%3A%2F%2Fpubmedcentralcanada.ca%2Fpmcc%2Farticles%2FPMC3237803%2F
[189]
Trepakova, E. S., Csutora, P., Hunton, D. L., Marchase, R. B., Cohen, R. A. and Bolotina, V. M. (2000) Calcium influx factor directly activates store-operated cation channels in vascular smooth muscle cells. Journal of Biological Chemistry 275, 26158-26163. 4068ce109aa7cfb245538be36366dba8http%3A%2F%2Feuropepmc.org%2Fabstract%2Fmed%2F10851243
[190]
Tsai, F. C., Seki, A., Yang, H. W., Hayer, A., Carrasco, S., Malmersjo, S. and Meyer, T. (2014) A polarized Ca2+, diacylglycerol and STIM1 signalling system regulates directed cell migration . Nature Cell Biology 16, 133-144. 62f48cb6a6eacc0b5637a687ad27199ahttp%3A%2F%2Fpubmedcentralcanada.ca%2Fpmcc%2Farticles%2Fpmid%2F24463606
[191]
Vaeth., M, Yang, J., Yamashita., M., Zee, I., Eckstein, M., Knosp, C., Kaufmann, U., Karoly, J. P., Lacruz, R. S., Flockerzi, V., Kacskovics, I., Prakriya, M. and Feske, S. (2017) ORAI 2 modulates store-operated calciumentry and T cell-mediated immunity. Nature Communications 8, 1-17. 0c8f2edca1c5cb933d4cf33e6d76ca10http%3A%2F%2Feuropepmc.org%2Fabstract%2FMED%2F28294127
[192]
Van den, M. I., Scholten, R., Cable, N. T., Hopman, M. T., Green, D. J. and Thijssen, D. H. (2012) Impact of age and sex on carotid and peripheral arterial wall thickness in humans. Acta Physiologica 206, 220-228.
[193]
Voelkers, M., Salz, M., Herzog, N., Frank, D., Dolatabadi, N., Frey, N., Gude, N., Friedrich, O., Koch, W. J., Katus, H. A., Sussman, M. A. and Most, P. (2010) Orai1 and Stim1 regulate normal and hypertrophic growth in cardiomyocytes. Journal of Molecular and Cellular Cardiology 48, 1329-1334. d50394bacb8c9e22acc9d6d8fd234500http%3A%2F%2Feuropepmc.org%2Fabstract%2FMED%2F20138887
[194]
Volkers, M., Dolatabadi, N., Gude, N., Most, P., Sussman, M. A. and Hassel, D. (2012) Orai1 deficiency leads to heart failure and skeletal myopathy in zebrafish. Journal of Cell Science 125, 287-294. e1d4d202eb4f55fe9aa5e52caf29a833http%3A%2F%2Feuropepmc.org%2Fabstract%2Fmed%2F22302996
[195]
Walsh, C. M., Doherty, M. K., Tepikin, A. V. and Burgoyne, R. D. (2010) Evidence for an interaction between Golli and STIM1 in store-operated calcium entry. Biochemical Journal 430, 453-460. 242014163cec40c8a9bb40fc1e2d0a1ahttp%3A%2F%2Fnew.med.wanfangdata.com.cn%2FPaper%2FDetail%3Fid%3DPeriodicalPaper_JJ0216386172
[196]
Wang, X., Wang, G., Lemos, J. R. and Treistman, S. N. (1994) Ethanol directly modulates gating of a dihydropyridine-sensitive Ca 2+ channel in neurohypophysial terminals . Journal of Neuroscience 14, 5453-5460. 1fbadced298844a885950f10ba1533e0http%3A%2F%2Fonlinelibrary.wiley.com%2Fresolve%2Freference%2FPMED%3Fid%3D7521910
[197]
Wang, Y., Deng, X., Mancarella, S., Hendron, E., Eguchi, S., Soboloff, J., Tang, X. D. and Gill, D. L. (2010) The calcium store sensor, STIM1, reciprocally controls Orai and CaV1.2 channels. Science 330, 105-109. 3849fcddd1923e383db39f0233de5b3chttp%3A%2F%2Feuropepmc.org%2Farticles%2FPMC3601900%2F
[198]
Watanabe, H., Murakami, M., Ohba, T., Takahashi, Y. and Ito, H. (2008) TRP channel and cardiovascular disease. Pharmacology and Therapeutics 118, 337-351. 054c7dc01e75475cce38b3010794e3c9http%3A%2F%2Fcardiovascres.oxfordjournals.org%2Flookup%2Fexternal-ref%3Faccess_num%3D18508125%26amp%3Blink_type%3DMED%26amp%3Batom%3D%252Fcardiovascres%252F91%252F3%252F492.atom
[199]
Wei, Y. C., George, N. I., Chang, C. W. and Hicks, K. A. (2017) Assessing sex differences in the risk of cardiovascular disease and mortality per increment in systolic blood pressure: a systematic review and meta-analysis of follow-up studies in the United States. PLoS One 12, 1-15.
[200]
Wellman, G. C., Cartin, L., Eckman, D. M., Stevenson, A. S., Saundry, C. M., Lederer, W. J. and Nelson, M. T. (2001) Membrane depolarization, elevated Ca2+ entry, and gene expression in cerebral arteries of hypertensive rats . American Journal of Physiology-Heart and Circulatory 281, H2559-H2567. 1aa1c5e650e921138af24134acaaa06ehttp%3A%2F%2Fwww.europepmc.org%2Fabstract%2FMED%2F11709423
[201]
Whelton, P. K., Carey, R. M., Aronow, W. S., Casey, D. E., Collins, K. J., Himmelfarb, C. D., DePalma, S. M., Gidding, S., Jamerson, K. A., Jones, D. W., et al. (2018) 2017 Guideline for the prevention, detection, evaluation, and management of high blood pressure in adults: executive summary: A report of the American College of Cardiology/American Heart Association task force on clinical practice guidelines. Hypertension 71, 1269-1324.
[202]
Wilson, L. A., McKeown, L., Tumova, S., Li, J. and Beech, D. J. (2015) Expression of a long variat of CRACR2A that belongs to the Rab GTPase protein family in endothelial cells. Biochemical and Biophysical Research Communications 456, 398-402. 77f0373d5577b04fb5d19821aa3ae03chttp%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F25475730
[203]
Wilson, P. C., Fitzgibbon, W. R., Garrett, S. M., Jaffa, A. A., Luttrel, L. M., Brands, M. W. and El-Shewy, H. M. (2015) Inhibition of Sphingosine kinase 1 ameliorates Angiotensin II-induced hypertension and inhibits transmembrane calcium entry via store-operated calcium channel. Molecular Endocrinology 29, 896-908. 11cfe8c0e030b19cb7e52f7b38392777http%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F25871850
[204]
Wu., M., Buchanan, J., Luik, R. M. and Lewis, R. S. (2006) Ca 2+ store depletion causes STIM1 to accumulate in ER regions closely associated with the plasma membrane . Journal of Cell Biology 174, 803-813. 2d724cc0efc919478c520f9768cd900bhttp%3A%2F%2Fpubmedcentralcanada.ca%2Farticlerender.cgi%3Faccid%3DPMC2064335
[205]
Xu, Y. J., Elimban , V. and Dhalla, N. S. (2015) Reduction of blood pressure by store-operated calcium channel blockers. Journal of Cellular and Molecular Medicine 19, 2763-2770. c54b10ed2c194f6bf9bc9dff411fbfechttp%3A%2F%2Fpubmedcentralcanada.ca%2Fpmcc%2Farticles%2FPMC4687696%2F
[206]
Xu, Y. J., Saini, H. K., Cheema, S. K., Dhalla, N. S. (2005) Mechanisms of lysophosphatidic acid-induced increase in intracellular calcium in vascular smooth muscle cells. Cell Calcium 38, 569-579. e5ac1566dfd719de4e7cf45128a6759bhttp%3A%2F%2Fwww.europepmc.org%2Fabstract%2FMED%2F16216324
[207]
Yan, F. R., Zhu, Z. L., Mu, Y. P., Zhuang, X. L., Lin, D. C., Wu, Z. J., Gui, L. X. and Lin, M. J. (2019) Increased caveolin-1 expression enhances the receptor-operated Ca2+ entry in the aorta of two-kidney, one-clip hypertensive rats . Experimental Physiology 104, 932-945.
[208]
Yang, S., Zhang, J. J and Huang, X. Y. (2009) Orai1 and STIM1 are critical for breast tumor cell migration and metastasis. Cancer Cell 15, 124-134. 443f0de8ec41f233d2b35c289ff6e426http%3A%2F%2Fwww.sciencedirect.com%2Fscience%2Farticle%2Fpii%2FS1535610808004388
[209]
Yang, X., Jin, H., Cai, X. and Shen, Y. (2012) Structural and mechanistic insights into the activation of Stromal interaction molecule 1 (STIM1). Proceedings of the National Academy of Sciences of the United States of America 109, 5657-5662. 9775b94cdbee22444585df65c2264153http%3A%2F%2Fcpfd.cnki.com.cn%2FArticle%2FCPFDTOTAL-IGSS201208001181.htm
[210]
Yuan, J. P., Zeng, W., Dorwart, M. R., Choi, Y. J., Worley, P. F. and Muallem, S. (2009) SOAR and the polybasic STIM1 domains gate and regulate Orai channels. Nature Cell Biology 11, 337-343. 92c84297186c5dcb070088183db501bahttp%3A%2F%2Fwww.nature.com%2Fncb%2Fjournal%2Fv11%2Fn3%2Fabs%2Fncb1842.html
[211]
Zhang, B., Naik, J. S., Jernigan, N. L., Walker, B. R. and Resta, T. C. (2018 a) Reduced membrane cholesterol after chronic hypoxia limits Orai1-mediated pulmonary endothelial Ca 2+ entry . American Journal of Physiology-Heart and Circulatory 314, H359-H369. e552a4ed261d45561d3bd47344162608http%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F29101179
[212]
Zhang, B., Paffett, M. L., Naik, J. S., Jernigan, N. L., Walker, B. R. and Resta, T. C. (2018 b) Cholesterol Regulation of Pulmonary Endothelial Calcium Homeostasis. Current Topics in Membranes 82, 53-91.
[213]
Zhang, S. L., Kozak, J. A., Jiang, W., Yeromin, A. V., Chen, J., Yu, Y., Penna, A., Shen, W., Chi, V. and Cahalan, M. D. (2008) Store-dependent and-independent modes regulating Ca2+ release-activated Ca2+ channel activity of human Orai1 and Orai3 . Journal of Biological Chemistry 283, 17662-17671. f6907fd87530f5847455102a9d674153http%3A%2F%2Feuropepmc.org%2Farticles%2FPMC2427323%2F
[214]
Zhang, W. and Trebak, M. (2011) STIM1 and Orai1: novel targets for vascular diseases. Science China Life 54, 780-785. 9c33de68debf0cc22b03bcef414828a0http%3A%2F%2Flink.springer.com%2Farticle%2F10.1007%2Fs11427-011-4206-6
[215]
Zhou, C., Townsley, M. I., Alexeyev, M., Voelkel, N. F. and Stevens, T. (2016) Endothelial hyperpermeability in severe pulmonary arterial hypertension: role of store-operated calcium entry. American Journal of Physiology-Lung Cellular and Molecular Physiology 311, L560-L569. cd3e7e09d6ad1f562bfc39d28d635b48http%3A%2F%2Fwww.ncbi.nlm.nih.gov%2Fpubmed%2F27422996
[216]
Zhou, Y., Meraner, P., Kwon, H. T., Machnes, D., Oh-hora, M., Zimmer, J., Huang, Y., Stura, A., Rao, A. and Hogan, P. G. (2010) STIM1 gates the store-operated calcium channel ORAI1 in vitro. Nature Structural and Molecular Biology 17, 112-116. 2302d3e5f7182799ffd7f15294fd026fhttp%3A%2F%2Fwww.nature.com%2Fnsmb%2Fjournal%2Fv17%2Fn1%2Fabs%2Fnsmb.1724.html
[217]
Zitt, C., Strauss, B., Schwarz, E. C., Spaeth, N., Rast, G., Hatzelmann, A. and Hoth, M. (2004) Potent inhibition of Ca 2+ release-activated Ca 2+ channels and T-lymphocyte activation by the pyrazole derivative BTP2 . Journal of Biological Chemistry 279, 12427-12437. 7c8cee49b77c42826d313c295b5cdb78http%3A%2F%2Feuropepmc.org%2Fabstract%2Fmed%2F14718545
Share
Back to top