Next Article in Journal
Study of Melatonin as Preventive Agent of Gastrointestinal Damage Induced by Sodium Diclofenac
Next Article in Special Issue
Direct Activation of TRPC3 Channels by the Antimalarial Agent Artemisinin
Previous Article in Journal
Parsing the IL-37-Mediated Suppression of Inflammasome Function
Previous Article in Special Issue
TRPC Channels: Dysregulation and Ca2+ Mishandling in Ischemic Heart Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Insights into Activation Mechanisms of Store-Operated TRPC1 Channels in Vascular Smooth Muscle

by
Miguel A. S. Martín-Aragón Baudel
1,
Jian Shi
2,
William A. Large
3 and
Anthony P. Albert
3,*
1
Department of Pharmacology, University of California, Davis, CA 95615, USA
2
LIGHT Laboratories, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds LS2 9JT, UK
3
Vascular Biology Research Centre, Molecular and Clinical Research Institute, St. George’s, University of London, London SW17 0RE, UK
*
Author to whom correspondence should be addressed.
Cells 2020, 9(1), 179; https://doi.org/10.3390/cells9010179
Submission received: 25 November 2019 / Revised: 3 January 2020 / Accepted: 5 January 2020 / Published: 10 January 2020
(This article belongs to the Special Issue TRPC Channels)

Abstract

:
In vascular smooth muscle cells (VMSCs), the stimulation of store-operated channels (SOCs) mediate Ca2+ influx pathways which regulate important cellular functions including contraction, proliferation, migration, and growth that are associated with the development of vascular diseases. It is therefore important that we understand the biophysical, molecular composition, activation pathways, and physiological significance of SOCs in VSMCs as these maybe future therapeutic targets for conditions such as hypertension and atherosclerosis. Archetypal SOCs called calcium release-activated channels (CRACs) are composed of Orai1 proteins and are stimulated by the endo/sarcoplasmic reticulum Ca2+ sensor stromal interaction molecule 1 (STIM1) following store depletion. In contrast, this review focuses on proposals that canonical transient receptor potential (TRPC) channels composed of a heteromeric TRPC1/C5 molecular template, with TRPC1 conferring activation by store depletion, mediate SOCs in native contractile VSMCs. In particular, it summarizes our recent findings which describe a novel activation pathway of these TRPC1-based SOCs, in which protein kinase C (PKC)-dependent TRPC1 phosphorylation and phosphatidylinositol 4,5-bisphosphate (PIP2) are obligatory for channel opening. This PKC- and PIP2-mediated gating mechanism is regulated by the PIP2-binding protein myristoylated alanine-rich C kinase (MARCKS) and is coupled to store depletion by TRPC1-STIM1 interactions which induce Gq/PLCβ1 activity. Interestingly, the biophysical properties and activation mechanisms of TRPC1-based SOCs in native contractile VSMCs are unlikely to involve Orai1.

1. Introduction

In vascular smooth muscle cells (VSMCs), neurotransmitters and hormones such as noradrenaline, adrenaline, angiotensin II (Ang II), and endothelin 1 (ET-1) produce an increase in cytosolic Ca2+ concentration (Ca2+i) (see [1,2,3,4,5,6,7] for reviews on Ca2+ signaling mechanisms in smooth muscle). This rise in Ca2+i is important for initiating vasoconstriction, which determines vascular resistance and blood pressure and ultimately regulates blood flow to our tissues and organs. In addition, an excessive increase in Ca2+i is linked to cell proliferation, migration and growth, phenotypes which are involved in the development of vascular disease. Therefore, understanding cellular pathways which mediate increases in Ca2+i may help to identify future therapeutic targets for conditions such as hypertension and atherosclerosis.
Vasoconstrictors induce a rise in Ca2+i by activating the classical phosphoinositol signaling pathway involving: stimulation of Gq-protein-coupled receptors, phospholipase C (PLC) activity, phosphatidylinositol 4,5-bisphosphate (PIP2) hydrolysis, and inositol 1,4,5-trisphosphate (IP3) and diacylglycerol (DAG) generation [1,2,3,4,5,6,7]. IP3 mediates an increase in Ca2+i by causing the release of Ca2+ from sarcoplasmic reticulum (SR) Ca2+ stores, and this subsequent rise in Ca2+i and DAG-mediated pathways induce Ca2+ influx from the extracellular medium. A component of this Ca2+ influx occurs through activation of voltage-gated Ca2+ channels (VGCCs), but there is also a significant contribution from voltage-independent Ca2+-permeable non-selective cation channels. Stimulation of Ca2+-permeable cation channels are thought to mediate Ca2+ entry pathways through direct Ca2+ influx, and through Na+ influx which leads to depolarization and activation of voltage-gated Ca2+ channels and stimulation of the Na+/Ca2+ exchanger in reverse mode.
There is substantial evidence that Ca2+-permeable non-selective cation channels classified as receptor-operated (ROCs) and store-operated (SOCs) are expressed in VSMCs [1,2]. ROCs are defined as ion channels activated by receptor stimulation independently of IP3-mediated depletion of SR Ca2+ stores, whereas SOCs are activated by pathways coupled to depletion of SR Ca2+ stores but not by the subsequent increase in Ca2+i. These definitions infer that stimulation of Gq-protein-coupled receptors will activate both ROCs and SOCs. In addition, it is proposed that SOCs may be activated by Gq-protein-coupled receptor stimulation independently of store depletion, hence SOCs may also function as ROCs [8]. Together, these findings make it difficult to study ROCs and SOCs independently of each other using macroscopic measurements such as whole-cell patch clamp and Ca2+ signal recordings but possible using single channel recordings [9,10,11]. Moreover, SOCs can be selectively activated by depleting SR Ca2+ stores in the absence of receptor stimulation using SR Ca2+-ATPase inhibitors (e.g., cyclopiazonic acid (CPA) and thapsigargin), and high (e.g., 1,2-bis(o-aminophyoxy)ethane-N,N,N’,N’-tetraacetic acid (BAPTA)) and low affinity (e.g., N,N,N’,N’,-tetrakis(2-pyridmethyl)-1,2-ethnediamine (TPEN)) Ca2+ chelators to passively deplete Ca2+i and SR Ca2+ levels respectively.
This article reviews our current understanding of the biophysical properties, molecular composition, and activation mechanisms of SOCs in VSMCs, in particular, it focuses on recent studies from our laboratory which have described a novel activation pathway of SOCs composed of canonical transient receptor potential 1 (TRPC1) proteins in native contractile VSMCs. In the majority of these studies SOCs were activated by SR Ca2+ ATPase inhibitors or Ca2+ chelators to prevent complications from activation of ROCs, although in the final section we discuss the potential physiological significance of this store-operated activation pathway in vasoconstrictor-induced TRPC1-based SOCs.

2. Biophysical Properties and Molecular Composition of SOCs in VSMCs

It is well-established that archetypal SOCs, termed calcium release-activated channels (CRACs), are characterized by high Ca2+ permeabilities (PCa2+:PNa+ > 1000:1), pronounced inward rectification with reversal potentials (Erev) greater than +50 mV, unitary conductances in the fS range, and are composed of Orai1 proteins (see [12] for review on properties and functions of Orai channels and their associated activation mechanisms). The fundamental activation mechanism of Orai1-based CRACs is also clearly outlined, with an essential role for stromal interaction molecule 1 (STIM1) which senses Ca2+ levels within ER/SR Ca2+ stores and following store depletion undergoes oligmerisation and translocation to cytosolic surface of the plasma membrane where it interacts with Orai1 to induce channel assembly and gating.
However, it is apparent that many cell types express SOCs with very different characteristics to Orai1-based CRACs such as much lower Ca2+ permeabilities, relatively linear or outward rectification, and considerably larger unitary conductances (see [13] for review on role of TRPC proteins in mediated SOCs). Given the properties of these SOCs, they are unlikely to be composed of Orai proteins. The molecular composition of these SOCs is controversial but there is increasing evidence that they are formed by the TRPC family of Ca2+-permeable non-selective cation channels (TRPC1–C7) [13]. These TRPC-based SOCs are likely to form a diverse group of channels with differing expression, properties, and functions since TRPC subunits form heterotetrameric structures [7,13]. There is further controversy over whether TRPC proteins form SOCs due to limited understanding, unlike with Orai proteins, of how store depletion couples to channel activation that is an essential requirement when defining SOCs. These issues are beginning to be unraveled by findings which suggest that store depletion activates TRPC-based SOCs through diverse STIM1-mediated processes including direct interactions between TRPC and Orai1 proteins, activation of Orai1-based CRACs as a prerequisite for TRPC1 opening, and direct interactions between TRPC and STIM1 [13,14]. For example, in overexpression studies it is proposed that direct interactions between STIM1 and TRPC channels govern activation [13,14,15,16,17,18], whereas elegant studies in salivary glands have shown that activation of TRPC1-based SOCs require prior activation of Orai1-based CRACs to induce insertion of TRPC1 subunits into the plasma membrane, with both activation of Orai1-based CRACs and TRPC1-based SOCs coupled to store depletion by STIM1 (see [19] for review on activation mechanisms of TRPC1-based SOCs in salivary glands). The studies highlighted in this review indicate that activation of STIM1-mediated TRPC1-based SOCs in native contractile VSMCs involves a Gq/PLCβ1 pathway and interactions between protein kinase C (PKC) activity and PIP2 which do not require Orai1.
The molecular composition and activation mechanisms of SOCs in VSMCs reflects the controversies highlighted above as there is significant evidence to indicate that both Orai1-based CRACs and TRPC-based SOCs are present in VSMCs, but that these channels are differentially expressed according to cell phenotype. In freshly isolated VSMCs or primary cultured VSMCs maintained in low serum, conditions in which VSMCs retain their native contractile phenotype, evidence suggests that SOCs have biophysical properties which resemble TRPC-based SOCs and not Orai1-based CRACs with relatively linear, S-shaped, or slightly outward rectification, low Ca2+ permeabilities (PCa2+:PNa+ of 1–50), and unitary conductances of 2–3 pS and 2–8 pS in 1.5 mM and 0 mM external Ca2+ respectively [1,2,20,21,22,23,24,25,26,27,28,29]. Moreover, studies using an array of different techniques such as blocking anti-TRPC antibodies (e.g., T1E3), TRPC1−/− mice, molecular knockdown (e.g., siRNA), and pharmacology (e.g., the selective TRPC1/C4/C5 inhibitor Pico145) [30,31] indicate that these SOCs are composed of a heteromeric TRPC1/C5 molecular template, which can also involve other TRPC subunits depending on the vascular bed [7,20,21,22,23,24,25,26,27,28,29,32,33]. As such, these studies show that TRPC1/C5-based SOCs are functionally expressed in pial arterioles and mesentery artery VSMCs, whereas TRPC1/C5/C6- and TRPC1/C5/C7-based SOCs are reported in coronary artery and portal vein VSMCs respectively. It is predicted that differences in the molecular compositions of these TRPC-based SOCs are reflected in the different properties of TRPC-based SOCs such as rectification, Ca2+ permeability and unitary conductance. For example, inclusion of TRPC6 within the TRPC1/C5 template produces biophysical properties which indicate a decreased Ca2+ permeability [22], whereas the presence of TRPC7 produces a facilitatory action of IP3 on TRPC1-based SOCs [22,34]. Studies using TRPC1−/− VSMCs indicate that TRPC1 is essential for conferring activation by store depletion, and therefore these TRPC1/C5 channel templates are termed TRPC1-based SOCs [25]. These findings fit with the idea that TRPC1 subunits do not form a functional homotetrameric channel [13]. Several studies have indicated that Orai1 proteins are unlikely to be involved in composing SOCs in VSMCs with a native contractile phenotype, with low expression of Orai1 proteins present in these cells ([35,36] and see [37,38] for review of Orai1 proteins in vascular smooth muscle) and biophysical properties of TRPC1-based SOCs and their STIM1-mediated activation mechanisms unaffected in freshly isolated and primary cultured VSMCs from Orai1−/− mice [29].
In contrast to native contractile VSMCs, long-term cultured VSMCs maintained in high serum, conditions that induce a non-contractile synthetic phenotype with proliferative, migrative and growth characteristics, express whole-cell SOCs and store-operated Ca2+ entry with similar properties to Orai1-based CRACs such as pronounced inward rectification, inhibition by a Orai1 selective inhibitor, and inhibition by knockdown of Orai1 and STIM1 protein levels but not by knockdown of TRPC1, TRPC4 or TRPC6 proteins [36,37,38,39,40,41,42]. In addition, store-operated whole-cell currents and Ca2+ entry are unaffected in cultured VSMCs maintained in high serum conditions from TRPC1−/− mice [43] further suggesting the involvement of Orai1-based CRACs in these VSMCs. In contrast, TRPC1 has also been proposed to be involved in regulating SOCs present in synthetic VSMCs, perhaps by regulating Orai1-based CRACs, and it is suggested that increase in the expression and function of TRPC1 may be an important trigger in the development of the cellular switch from a contractile to synthetic phenotype [37,38].
These complexities mean that when studying SOCs in VSMCs it is essential to clearly define cell phenotype of VSMCs. In this regard, when investigating SOCs in native contractile VSMCs it is advantageous to use freshly isolated cells whenever possible, and when required only use primary culture cells maintained in low serum conditions for as short a time as possible.

3. Activation Mechanisms of TRPC1-Based SOCs in Native Contractile VSMCs

The following sections discuss our recent findings that TRPC1-based SOCs in freshly isolated and primary cultured VSMCs with a native contractile phenotype exhibit a complex activation mechanism, in which interactions between PKC-dependent phosphorylation of TRPC1 and phosphatidylinositol 4,5-bisphosphate (PIP2) regulated by myristoylated alanine-rich C kinase (MARCKS) are obligatory gating partners, with these mechanisms coupled to store depletion by a novel STIM1-mediated Gq-PLCβ1 pathway.

3.1. PKC Activity and PIP2 have Obligatory Roles in Activation of TRPC1-based SOCs

There are several lines of evidence which indicate that PKC activity and PIP2 are important in activating TRPC1-based SOCs. The stimulation of whole-cell and single channel TRPC1-based SOCs is significantly reduced by PKC inhibitors and anti-PIP2 antibodies [22,25,44]. In support of these results, PKC activators (diacylglycerol analogues, phorbol esters, PKC catalytic subunits) and exogenous application of diC8-PIP2 (a water-soluble form of PIP2) stimulate TRPC1-based SOCs in WT but not in TRPC1−/− VSMCs [22,25,44]. Interestingly, PKC-stimulated activation of TRPC1-based SOCs are prevented by anti-PIP2 antibodies and pharmacological agents such as wortmannin to deplete PIP2 levels, whereas diC8-PIP2-activated TRPC1-based SOCs are reduced by PKC inhibitors [44]. Moreover, store depletion induces PKC-dependent phosphorylation of TRPC1 and increases association between PIP2 and TRPC1 [26,27,28,44]. Together, these results indicate that interactions between PKC activity and PIP2 have obligatory roles in activation of TRPC1-based SOCs; PKC cannot activate TRPC1-based SOCs without PIP2, and vice versa. This supports the hypothesis that PIP2 is the activating ligand of TRPC1-based SOCs and that store-operated PKC-dependent phosphorylation of TRPC1 is required for this opening mechanism to occur.
These proposed roles of PKC and PIP2 on TRPC1-based SOCs are different to their roles in the activation of TRPC3/C6/C7-based ROCs in native contractile VSMCs (see [23,24] for comprehensive reviews of activation mechanisms of TRPC channels). It is well established that this subgroup of TRPC channels are activated by receptor-mediated generation of DAG which leads to channel opening via PKC-independent mechanisms, with PKC causing channel inhibition. In addition, the role of PIP2 on TRPC3/C6/C7-based ROCs is unclear with both inhibitory and excitatory actions proposed [45,46]. These findings further indicate that TRPC1-based SOCs and TRPC3/C6/C7-based ROCs form distinct channel structures with differing activation mechanisms, and likely distinct functions in VSMCs.
Important omissions from our understanding are what PKC isoform(s) is involved, which amino acid(s) within TRPC1 protein structure is phosphorylated, and how a PKC-dependent phosphorylation process alters interactions between TRPC1 and PIP2. The PKC family comprises of at least 11 serine/threonine kinases divided into three groups according to their basic structure and activation requirements: conventional PKC isoforms (α, βI, βII and γ) require both Ca2+ and diacylglycerol (DAG), novel PKC isoforms (δ, ε, η and θ) require DAG but are Ca2+-insensitive, and atypical PKC isoforms (ζ, ι and λ) are activated by lipid mediators such as phosphatidylserine and do not require Ca2+ or DAG (see [47] for review of the role of PKC isoforms in vascular smooth muscle). Our results indicate that stimulation of TRPC1-based SOCs and PKC-dependent phosphorylation of TRPC1 by store depletion requires PLCβ1 activity, and that DAG analogues activate TRPC1-based SOCs through a PKC-dependent mechanism [9,23,24,27]. Moreover, we have shown that TRPC1-based SOCs are activated by store depleting agents which are likely to increase (e.g., CPA), decrease (e.g., the high affinity cell-impermeable and -permeable Ca2+ chelators BAPTA and BAPTA-AM), or produce little change in Ca2+i (e.g., the low affinity cell-permeable Ca2+ chelator TPEN). These findings suggest that the PKC isoform involved is likely to require DAG but is Ca2+-insensitive, which are characteristics of the novel group of PKC isoforms. Our preliminary data indicate that PKCδ is the most highly expressed novel PKC isoform in VSMCs, and that selective PKCδ inhibitory peptides and knockdown of PKCδ using morpholino sequences prevent activation of TRPC1-based SOCs [31].
Interestingly, the predication of PKCδ-dependent phosphorylation sites within the TRPC1 sequence using GPS 3.0 reveals five intracellular serine residues, with Ser619 and Ser752 at the C-terminal domain of potential significance as both these sites are close to a known PIP2-binding domain [48]. It is therefore possible that PKCδ-dependent phosphorylation of these sites increases PIP2 affinity leading to increased binding to TRPC1 and channel opening. Previous studies have highlighted that protein kinase A (PKA), protein kinase G (PKG), and calmodulin kinase II (CaMKII) have inhibitory actions on TRPC1-based SOCs in VSMCs [49,50,51], and it may be that phosphorylation of serine/threonine amino acids by these kinases reduce TRPC1 and PIP2 interactions by lowering PIP2 affinity. Similar roles for kinase activities in modulating lipid-protein interactions are well-established in the regulation of different K+ channel subtypes [52].

3.2. Interactions Between PKC Activity and PIP2 are Regulated by MARCKS

An important question is how PKC-dependent phosphorylation regulates PIP2 gating of TRPC1-based SOCs when physiological activators of SOCs involve stimulation of Gq-coupled receptors which drive PLC activity and PIP2 hydrolysis. To provide answers to this question, we investigated the role of MARCKS in activation of TRPC1-based SOCs. MARCKS is a membrane-bound PIP2-binding protein that acts a PIP2 buffer, which can provide a distinct pool of PIP2 at the plasma membrane that is protected from breakdown by PLC thus enabling this phospholipid to be released in a coordinated manner into the local environment [53,54]. Using a combination of electrophysiological, co-immunoprecipitation, and PIP2-binding dot-blot assays, we showed that in unstimulated conditions MARCKS is bound to TRPC1, with PIP2 predominately associated with MARCKS and not TRPC1 [26]. Upon stimulation by store depleting agents MARCKS dissociates from TRPC1, which leads to PIP2 being released and associated with TRPC1 to cause channel opening. Importantly, both dissociation of MARCKS from TRPC1 and redistribution of PIP2 from MARCKS to TRPC1 are stimulated by phorbol esters and prevented by PKC inhibitors indicating that PKC activity is central to these mechanisms. It will be important to ascertain how receptor stimulation and store-depleting agents couple to these actions of MARCKS. A potential idea is that stimulation of CaM is involved, as this Ca2+-binding protein is known to bind to MARCKS and to cause dissociation from the plasma membrane and PIP2 release [53,55]. Furthermore, CaM is known to activate TRPC1-based SOCs in native contractile VSMCs [50].

3.3. Store Depletion Activates a Gq-PLCβ1 Pathway Involved in Activation of TRPC1-Based SOCs

For a channel to be defined as store-operated, it is essential to understand how store depletion is coupled to channel opening. Therefore, a central question surrounding the activation of TRPC1-based SOCs is how store depletion couples to PKC-dependent phosphorylation of TRPC1 and opening by PIP2? A crucial finding was the discovery that store depletion activates a Gq-PLCβ1 pathway which drives the PKC-dependent phosphorylation of TRPC1 [27]. Our studies showed that the G-protein inhibitor GDP-β-S, anti-Gq antibodies (but not anti-Gi), the PLC inhibitor U73122, and PLCβ1 shRNA inhibited activation of TRPC1-based SOCs. In addition, store-operated PKC-dependent phosphorylation of TRPC1 was inhibited by U73122 and PLCβ1 shRNA. A significant finding was that store depleting agents induced PLC activity measured using GFP-PLCδ1-PH, a fluorescent biosensor for PIP2 and IP3 ([56,57,58] and reviewed in [59]) in primary cultured VSMCs which was prevented by a PLC inhibitor and PLCβ1 shRNA. In support of these findings, co-immunoprecipitation and proximity ligation assays (PLA) demonstrated that store depletion induced interactions between TRPC1, Gq and PLCβ1 at the plasma membrane.

3.4. STIM1 Couples Store Depletion to Gq-PLCβ1 Activity to Stimulate TRPC1 SOCs

Next, we investigated how store depletion is coupled to the formation and activation of this Gq/PLCβ1/PKC/TRPC1 signal transduction pathway. An obvious candidate was STIM1, since along with its classically described role in activating Orai1-based CRACs it has been implicated in activating TRPC-based SOCs (see [13] for review). Moreover, it is known that that STIM1 has diverse cellular partners including ion channels [12,13,15,16,17,18,60,61], SR and plasma membrane ATPases [62,63], and adenylate cyclase [64] and therefore it seemed a reasonable idea that STIM1 may couple to Gq/PLCβ1 activity.
We demonstrated that whole-cell and single channel TRPC1-based SOCs and store-operated PLCβ1 activity were inhibited by shRNA STIM1 and were absent in TRPC1−/− VSMCs [28]. In addition, store-operated PKC-dependent phosphorylation of TRPC1 was greatly reduced by shRNA STIM1. Moreover, STIM1 was required for store-operated interactions between Gq, PLCβ1 and TRPC1, and TRPC1 was essential for store-operated interactions between Gq, PLCβ1, and STIM1. These findings provide strong evidence that STIM1 is an essential molecule in activation of TRPC1-based SOCs, and importantly it indicates that store-operated STIM1-TRPC1 interactions (measured using PLA assays which imply that these interactions occur within 40 nm) form the structural basis for stimulation of the Gq/PLCβ1 pathway required for PKC-dependent TRPC1 phosphorylation and channel opening by PIP2.
As expected, store depletion induced translocation of STIM1 from the cytosol to the plasma membrane where it formed discrete puncta and interactions with TRPC1 using immunocytochemical and PLA techniques, which were not dependent on downstream molecules in the signal pathway such as PLCβ1 [28]. Interestingly, in the absence of TRPC1, store depletion still induced translocation of STIM1 from cytosol to the plasma membrane, but STIM1 formed an even distribution throughout the plasma membrane and not discrete puncta. This suggests that TRPC1 may be essential for coordinating the response of STIM1 to store depletion.
An interpretation from these studies is that STIM1-TRPC1 interactions, possibly involving other unknown molecules, act as a cellular activator of Gq subunits, and that this leads to interactions with, and activation of, PLCβ1. In essence STIM1-TRPC1 interactions behave like Gq-coupled receptors or guanine exchange factors (GEFs). In the future, it will be important to investigate structural interactions between STIM1 and TRPC1 and discover where Gq subunits binds. Initial work might focus on the CRAC-activating (CAD) and polybasic domains which have been linked to binding and activation of TRPC1 by protein-protein interactions and electrostatic interactions respectively [13,15,16,17,18].
Interestingly, differential blocking effects of N-terminal and C-terminal anti-STIM1 antibodies on TRPC1-based SOCs using whole-cell and inside-out patch clamp recordings may suggest that store-operated STIM1/TRPC1 interactions lead to STIM1 acting as a transmembrane protein at the plasma membrane [28]. In this configuration, the N-terminal domain of STIM1 is now present at the extracellular surface of the plasma membrane. Perhaps this orientation of STIM1-TRPC1 interactions has implications for association with Gq subunits in native contractile VSMCs. A similar role for STIM1 has been previously described in mediating store-operated conductances [65]. However, it seems unlikely that the source of STIM1 involved in activating TRPC1-based SOCs resides at the plasma membrane, as with activation of arachidonic acid-regulated channels (ARC) composed of Orai1/Orai3 subunits [66], since our data indicates that store depletion induces translocation of STIM1 from the cytosol to the plasma membrane.

3.5. Activation Mechanisms of TRPC1-Based SOCs are Independent of Orai1

There is substantial evidence that TRPC1, Orai1 and STIM1 have important roles in mediating store-operated Ca2+ entry pathways in VSMCs with a synthetic phenotype, although it is unclear whether Orai1 and TRPC1 interact together or form separate STIM1-mediated ionic mechanisms (see earlier). In addition, it is unclear whether Orai1 is required for activation of TRPC1-based SOCs in native contractile VSMCs. In our recent study, we showed that the properties of store-operated whole-cell and single channel currents in freshly isolated and primary cultured native contractile WT and Orai1−/− VSMCs were similar to previously described TRPC1-based SOCs, and that store-operated STIM1-mediated PLCβ1 activity and STIM1-TRPC1 interactions were unaffected in Orai1−/− cells [29]. These findings provide significant evidence that Orai1 proteins are not required for the molecular composition or activation mechanisms of TRPC1-based SOCs in contractile VSMCs. These results support the findings that Orai1 expression is very low in native contractile VSMCs [35,36,37,38]. A caveat is that Orai proteins are composed of three subtypes, Orai1-3 [12], and therefore Orai2 and Orai3 may be involved in TRPC1-based SOCs in native contractile VSMCs. However, whole-cell store-operated conductances with the distinct characteristics of Orai proteins, such as such inward rectification, and reversal potential greater than +50 mV have not been identified in native contractile VSMCs from WT, Orai1−/− or TRPC1−/− preparations [25,29].
It should be noted that other studies using pharmacological and molecular techniques have implicated Orai1 and Ca2+-independent phospholipase A2 (iPLA2) in store-operated conductances in native and primary cultured contractile VSMCs [67]. These studies propose that store depletion linked to STIM1-mediated release of a calcium influx factor (CIF) from the SR activates Orai1 through an iPLA2-mediated mechanism involving production of lysophospholipids (see [68] for review). In addition, Orai1 has been proposed alongside TRPC1 to regulate store-operated vascular contractility through interactions with voltage-gated Ca2+ channels ([69] and reviewed in [70]). The differences between these findings to those described above are unclear and may reflect differences between SOCs in different vascular beds and perhaps different cell isolation or culturing conditions. However, before these ideas can be truly accepted it will be essential to resolve the identity of CIF and understand why proposed Orai1-sensitive currents in contractile VSMCs have such a linear rectification [67]. For example, do contractile VSMCs express an Orai1 splice variant with very different properties to established Orai-based CRACs? In addition, a difficult finding to explain is why store-operated conductances are absent in native contractile VSMCs from TRPC1−/− mice [25]. Moreover, involvement of iPLA2 has been inferred in many studies using the proposed inhibitor bromoenol lactone (BEL), which has also been shown to block heteromeric TRPC1/C5, TRPC5, and TRPC6 channels and VGCCs [71].

4. Physiological Significance of TRPC1-Based SOCs Activation Pathway

To avoid complications from receptor-operated pathways, most studies use agents that selectively deplete SR Ca2+ stores (e.g., CPA, BAPTA) instead of receptor stimulation to study SOCs (see Introduction). However, it is important to examine whether proposed store-operated mechanisms are involved in activation of SOCs by physiological receptor stimulation. This is particularly important for TRPC1-based SOCs in native contractile VSMCs as these channels also behave as ROCs, and so may be activated by distinct store-independent and -dependent pathways [1,2]. Previous studies have shown that noradrenaline, Ang II and ET-1-activated ETB receptor stimulation induce TRPC1-based SOCs via a PLC-mediated pathway that requires PKC activity and PIP2 for channel opening in portal vein, mesenteric artery and coronary artery VSMCs respectively [8,9,10,11,23,72]. To investigate whether these receptor-mediated activation pathways of TRPC1-based SOCs might involve our proposed store-operated STIM1/Gq/PLCβ1 pathway, we showed that noradrenaline-activated TRPC1-based SOCs were greatly reduced by knockdown of PLCβ1 and STIM1 and that noradrenaline also induced interactions between STIM1 and TRPC1, Gq, and PLCβ1 [27,28]. Moreover, noradrenaline induced PKC-dependent phosphorylation of TRPC1 and activation of TRPC1-based SOCs by regulating MARCKS-TRPC1-PIP2 interactions similar to those produced by store depleting agents [26]. These findings highlight that physiological receptor stimulation is likely to activate TRPC1-based SOCs through the store-operated STIM1/Gq/PLCβ1/PKC pathway and also via a store-independent pathway involving a Gq/PLC/PKC pathway. It may be that receptor stimulation switches between these two modes to activate TRPC1-based SOCs according to concentration of the physiological agonist. For example, higher vasoconstrictor concentrations may be more likely to produce substantial IP3-mediated depletion of SR Ca2+ stores and activation of TRPC1-based SOCs by store depletion.
It is likely that certain receptors only activate TRPC1-based SOCs via store-independent pathways. In native contractile coronary artery VSMCs, ET-1-activated ETA receptor stimulation induces TRPC1-based SOCs through a Gβγ-protein-phosphoinositol 3-kinase (PI3K)-mediated pathway, with PIP3 formation thought to both induce PKC activity and act as the channel activating ligand; a pathway which is unlikely to involve IP3-mediated depletion of SR Ca2+ stores [11,72]. Therefore, TRPC1-based SOCs in coronary artery VSMCs are activated by ETA and ETB receptor stimulation through separate store-independent and -dependent pathways respectively, both requiring obligatory roles of PKC activity and phospholipids [11,66].
Interestingly, the knockdown of PLCβ1 and STIM1 had little effect on noradrenaline-stimulated PLC activity measured using the GFP-PLCδ-PH biosensor, although it was inhibited by U73122 a general PLC isoform inhibitor [27,28]. These findings support significant physiological relevance to our results, as it suggests that stimulation of Gq-protein-coupled receptors and IP3-mediated depletion of SR Ca2+ stores are likely to activate two different Gq-PLC pathways mediated by distinct PLC isoforms. To test this hypothesis, it would be important to identify the PLC isoform linked to α1-adrenoceptor stimulation. The critical role of MARCKS in stimulation of TRPC1-based SOCs is further highlighted when considering the physiological significance of our proposed activation pathway. Stimulation of Gq-protein-coupled receptors and IP3-mediated depletion of SR Ca2+ stores will both induce Gq-PLC activities leading to PIP2 hydrolysis. Therefore, MARCKS provides the essential role of a buffering a separate pool of PIP2, which is protected from PLC-mediated hydrolysis enabling the phospholipid to be available for channel opening.

5. Summary

This review highlights recent findings that SOCs in native contractile VSMCs are mediated by TRPC1-based SOCs, which have very different biophysical properties to the well-characterized channels formed by Orai proteins, including archetypal Orai1-based CRACs. Moreover, Figure 1 describes a novel activation pathway for these TRPC1-based SOCs proposed from our recent studies, in which SR Ca2+ store depletion by receptor-induced IP3-mediated generation or store depleting agents such as CPA and BAPTA stimulate STIM1 to translocate to the plasma membrane where it interacts with TRPC1 subunits to form a receptor for Gq G-protein subunits which induces PLCβ1 activity, production of DAG, and PKC activity. PKC-dependent phosphorylation of TRPC1 increases PIP2 binding and channel opening. The interactions between TRPC1 and PIP2 are regulated by MARCKS, a plasma membrane PIP2-binding protein bound to TRPC1 at rest, which upon store depletion dissociates from TRPC1 and releases PIP2 into the local environment where the phospholipid acts as the gating ligand. Figure 1 also depicts our current understanding of how receptor stimulation by vasoconstrictors activates TRPC1-based SOCs in contractile VSMCs by store-independent pathways involving PKC activity and phospholipids.

Author Contributions

All authors were involved in production of this manuscript and have discussed the final version of the manuscript.

Funding

This work was supported by Biotechnology and Biological Sciences Research Council grants (BB/J007226/1 and BB/M018350/1 to APA).

Conflicts of Interest

To the best of our knowledge, the authors declare no conflict of interest.

References

  1. Albert, A.P.; Large, W.A. Store-operated Ca2+-permeable non-selective cation channels in smooth muscle cells. Cell Calcium 2003, 33, 345–356. [Google Scholar] [CrossRef]
  2. Albert, A.P.; Saleh, S.N.; Peppiatt-Wildman, C.M.; Large, W.A. Multiple activation mechanisms of store-operated TRPC channels in smooth muscle cells. J. Physiol. 2007, 583, 25–36. [Google Scholar] [CrossRef] [PubMed]
  3. Abramowitz, J.; Birnbaumer, L. Physiology and pathophysiology of canonical transient receptor potential channels. FASEB J. 2009, 23, 297–328. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Beech, D.J. Characteristics of transient receptor potential canonical calcium-permeable channels and their relevance to vascular physiology and disease. Circ. J. 2013, 77, 570–579. [Google Scholar] [CrossRef] [Green Version]
  5. Earley, S.; Brayden, J.E. Transient receptor potential channels in the vasculature. Physiol. Rev. 2015, 95, 645–690. [Google Scholar] [CrossRef] [Green Version]
  6. Large, W.A. Receptor-operated Ca2+-permeable nonselective cation channels in vascular smooth muscle: A physiologic perspective. J. Cardiovasc. Electrophysiol. 2002, 13, 493–501. [Google Scholar] [CrossRef]
  7. Albert, A.P.; Saleh, S.N.; Large, W.A. Identification of canonical transient receptor potential (TRPC) channel proteins in native vascular smooth muscle cells. Curr. Med. Chem. 2009, 16, 1158–1165. [Google Scholar] [CrossRef]
  8. Albert, A.P.; Large, W.A. Activation of store-operated channels by noradrenaline via protein kinase C in rabbit portal vein myocytes. J. Physiol. 2002, 544, 113–125. [Google Scholar] [CrossRef]
  9. Saleh, S.N.; Albert, A.P.; Peppiatt, C.M.; Large, W.A. Angiotensin II activates two cation conductances with distinct TRPC1 and TRPC6 channel properties in rabbit mesenteric artery myocytes. J. Physiol. 2006, 577, 479–495. [Google Scholar] [CrossRef]
  10. Shi, J.; Ju, M.; Saleh, S.N.; Albert, A.P.; Large, W.A. TRPC6 channels stimulated by angiotensin II are inhibited by TRPC1/C5 channel activity through a Ca2+- and PKC-dependent mechanism in native vascular myocytes. J. Physiol. 2010, 588, 3671–3682. [Google Scholar] [CrossRef]
  11. Shi, J.; Ju, M.; Large, W.A.; Albert, A.P. Pharmacological profile of phosphatidylinositol 3-kinases and related phosphatidylinositols mediating endothelin(A) receptor-operated native TRPC channels in rabbit coronary artery myocytes. Br. J. Pharmacol. 2012, 166, 2161–2175. [Google Scholar] [CrossRef] [Green Version]
  12. Prakriya, M.; Lewis, R.S. Store-operated calcium channels. Physiol. Rev. 2015, 95, 1383–1436. [Google Scholar] [CrossRef] [Green Version]
  13. Cheng, K.T.; Ong, H.L.; Liu, X.; Ambudkar, I.S. Contribution and regulation of TRPC channels in store-operated Ca2+ entry. Curr. Top. Membr. 2013, 71, 149–179. [Google Scholar]
  14. Liao, Y.; Abramowitz, J.; Birnbaumer, L. The TRPC family of TRP channels: Roles inferred (mostly) from knockout mice and relationship to ORAI proteins. Handb. Exp. Pharmacol. 2014, 223, 1055–1075. [Google Scholar]
  15. Worley, P.F.; Zeng, W.; Huang, G.N.; Yuan, J.P.; Kim, J.Y.; Lee, M.G.; Muallem, S. TRPC channels as STIM1-regulated store-operated channels. Cell Calcium 2007, 42, 205–211. [Google Scholar] [CrossRef] [Green Version]
  16. Yuan, J.P.; Kim, M.S.; Zeng, W.; Shin, D.M.; Huang, G.; Worley, P.F.; Muallem, S. TRPC channels as STIM1-regulated SOCs. Channels 2009, 3, 221–225. [Google Scholar] [CrossRef] [Green Version]
  17. Lee, K.P.; Choi, S.; Hong, J.H.; Ahuja, M.; Graham, S.; Ma, R.; So, I.; Shin, D.M.; Muallem, S.; Yuan, J.P. Molecular determinants mediating gating of transient receptor potential canonical (TRPC) channels by stromal interaction molecule 1 (STIM1). J. Biol. Chem. 2014, 289, 6372–6382. [Google Scholar] [CrossRef] [Green Version]
  18. Asanov, A.; Sampieri, A.; Moreno, C.; Pacheco, J.; Salgado, A.; Sherry, R.; Vaca, L. Combined single channel and single molecule detection identifies subunit composition of STIM1-activated transient receptor potential canonical (TRPC) channels. Cell Calcium 2015, 57, 1–13. [Google Scholar] [CrossRef]
  19. Ambudkar, I.S.; de Souza, L.B.; Ong, H.L. TRPC1, Orai1, and STIM1 in SOCE: Friends in tight spaces. Cell Calcium 2017, 63, 33–39. [Google Scholar] [CrossRef] [Green Version]
  20. Trepakova, E.S.; Gericke, M.; Hirakawa, Y.; Weisbrod, R.M.; Cohen, R.A.; Bolotina, V.M. Properties of a native cation channel activated by Ca2+ store depletion in vascular smooth muscle cells. J. Biol. Chem. 2001, 276, 7782–7790. [Google Scholar] [CrossRef] [Green Version]
  21. Albert, A.P.; Large, W.A. A Ca2+-permeable non-selective cation channel activated by depletion of internal Ca2+ stores in single rabbit portal vein myocytes. J. Physiol. 2002, 538, 717–728. [Google Scholar] [CrossRef] [PubMed]
  22. Saleh, S.N.; Albert, A.P.; Peppiatt-Wildman, C.M.; Large, W.A. Diverse properties of store-operated TRPC channels activated by protein kinase C in vascular myocytes. J. Physiol. 2008, 586, 2463–2476. [Google Scholar] [CrossRef]
  23. Large, W.A.; Saleh, S.N.; Albert, A.P. Role of phosphoinositol 4,5-bisphosphate and diacylycerol in regulating native TRPC channel proteins in vascular smooth muscle. Cell Calcium 2009, 45, 574–582. [Google Scholar] [CrossRef]
  24. Albert, A.P. Gating mechanisms of canonical transient receptor potential channel proteins: Role of phosphoinositols and diacylglycerol. Adv. Exp. Med. Biol. 2011, 704, 391–411. [Google Scholar]
  25. Shi, J.; Ju, M.; Abramowitz, J.; Large, W.A.; Birnbaumer, L.; Albert, A.P. TRPC1 proteins confer PKC and phosphoinositol activation on native heteromeric TRPC1/C5 channels in vascular smooth muscle: Comparative study of wild-type and TRPC1−/− mice. FASEB J. 2012, 26, 409–419. [Google Scholar] [CrossRef] [Green Version]
  26. Shi, J.; Birnbaumer, L.; Large, W.A.; Albert, A.P. Myristoylated alanine-rich C kinase substrate coordinates native TRPC1 channel activation by phosphatidylinositol 4,5-bisphosphate and protein kinase C in vascular smooth muscle. FASEB J. 2014, 28, 244–255. [Google Scholar] [CrossRef] [Green Version]
  27. Shi, J.; Miralles, F.; Birnbaumer, L.; Large, W.A.; Albert, A.P. Store depletion induces Gαq-mediated PLCβ1 activity to stimulate TRPC1 channels in vascular smooth muscle cells. FASEB J. 2016, 30, 702–715. [Google Scholar] [CrossRef]
  28. Shi, J.; Miralles, F.; Birnbaumer, L.; Large, W.A.; Albert, A.P. Store-operated interactions between plasmalemmal STIM1 and TRPC1 proteins stimulate PLCβ1 to induce TRPC1 channel activation in vascular smooth muscle cells. J. Physiol. 2017, 595, 1039–1058. [Google Scholar] [CrossRef]
  29. Shi, J.; Miralles, F.; Kinet, J.P.; Birnbaumer, L.; Large, W.A.; Albert, A.P. Evidence that Orai1 does not contribute to store-operated TRPC1 channels in vascular smooth muscle cells. Channels 2017, 11, 329–339. [Google Scholar] [CrossRef] [Green Version]
  30. Rubaiy, H.N.; Ludlow, M.J.; Henrot, M.; Gaunt, H.J.; Miteva, K.; Cheung, S.Y.; Tanahashi, Y.; Hamzah, N.; Musialowski, K.E.; Blythe, N.M.; et al. Picomolar, selective, and subtype-specific small-molecule inhibition of TRPC1/4/5 channels. J. Biol. Chem. 2017, 292, 8158–8173. [Google Scholar] [CrossRef] [Green Version]
  31. Martin-Aragon Baudel, M.; Shi, J.; Jahan, K.S.; Large, W.A.; Albert, A.P. Obligatory role for PKCδ in PIP2-mediated activation of store-operated TRPC1 channels in vascular smooth muscle cells (manuscript in preparation). (manuscript in preparation).
  32. Xu, S.Z.; Beech, D.J. TrpC1 is a membrane-spanning subunit of store-operated Ca2+ channels in native vascular smooth muscle cells. Circ. Res. 2001, 88, 84–87. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Xu, S.Z.; Boulay, G.; Flemming, R.; Beech, D.J. E3-targeted anti-TRPC5 antibody inhibits store-operated calcium entry in freshly isolated pial arterioles. Am. J. Physiol. Heart Circ. Physiol. 2006, 291, H2653–H2659. [Google Scholar] [CrossRef] [Green Version]
  34. Liu, M.; Albert, A.P.; Large, W.A. Facilitatory effect of Ins(1,4,5)P3 on store-operated Ca2+-permeable cation channels in rabbit portal vein myocytes. J. Physiol. 2005, 566, 161–171. [Google Scholar] [CrossRef]
  35. Berra-Romani, R.; Mazzocco-Spezzia, A.; Pulina, M.V.; Golovina, V.A. Ca2+ handling is altered when arterial myocytes progress from a contractile to a proliferative phenotype in culture. Am. J. Physiol. Cell Physiol. 2008, 295, C779–C790. [Google Scholar] [CrossRef] [Green Version]
  36. Potier, M.; Gonzalez, J.C.; Motiani, R.K.; Abdullaev, I.F.; Bisaillon, J.M.; Singer, H.A.; Trebak, M. Evidence for STIM1- and Orai1-dependent store-operated calcium influx through ICRAC in vascular smooth muscle cells: Role in proliferation and migration. FASEB J. 2009, 23, 2425–2437. [Google Scholar] [CrossRef] [Green Version]
  37. Beech, D.J. Orai1 calcium channels in the vasculature. Pflug. Arch. 2012, 463, 635–647. [Google Scholar] [CrossRef] [Green Version]
  38. Trebak, M. STIM/Orai signalling complexes in vascular smooth muscle. J. Physiol. 2012, 590, 4201–4208. [Google Scholar] [CrossRef]
  39. Li, J.; Sukumar, P.; Milligan, C.J.; Kumar, B.; Ma, Z.Y.; Munsch, C.M.; Jiang, L.H.; Porter, K.E.; Beech, D.J. Interactions, functions, and independence of plasma membrane STIM1 and TRPC1 in vascular smooth muscle cells. Circ. Res. 2008, 103, 97–104. [Google Scholar] [CrossRef] [Green Version]
  40. Ng, L.C.; O′Neill, K.G.; French, D.; Airey, J.A.; Singer, C.A.; Tian, H.; Shen, X.M.; Hume, J.R. TRPC1 and STIM1 mediate capacitative Ca2+ entry in mouse pulmonary arterial smooth muscle cells. J. Physiol. 2009, 587, 2429–2442. [Google Scholar] [CrossRef]
  41. Ng, L.C.; Ramduny, D.; Airey, J.A.; Singer, C.A.; Keller, P.S.; Shen, X.M.; Tian, H.; Valencik, M.; Hume, J.R. Orai1 interacts with STIM1 and mediates capacitative Ca2+ entry in mouse pulmonary arterial smooth muscle cells. Am. J. Physiol. Cell Physiol. 2010, 299, C1079–C1090. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Li, J.; McKeown, L.; Ojelabi, O.; Stacey, M.; Foster, R.; O′Regan, D.; Porter, K.E.; Beech, D.J. Nanomolar potency and selectivity of a Ca²⁺ release-activated Ca²⁺ channel inhibitor against store-operated Ca²⁺ entry and migration of vascular smooth muscle cells. Br. J. Pharmacol. 2011, 164, 382–393. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Dietrich, A.; Kalwa, H.; Storch, U.; Mederos y Schnitzler, M.; Salannova, B.; Pinkenburg, O.; Dubrovska, G.; Essin, K.; Gollasch, M.; Birnbaumer, L.; et al. Pressure-induced and store-operated cation influx in vascular smooth muscle cells is independent of TRPC1. Pflug. Arch. 2007, 455, 465–477. [Google Scholar] [CrossRef] [PubMed]
  44. Saleh, S.N.; Albert, A.P.; Large, W.A. Obligatory role for phosphatidylinositol 4,5-bisphosphate in activation of native TRPC1 store-operated channels in vascular myocytes. J. Physiol. 2009, 587, 531–540. [Google Scholar] [CrossRef] [Green Version]
  45. Albert, A.P.; Saleh, S.N.; Large, W.A. Inhibition of native TRPC6 channel activity by phosphatidylinositol 4,5-bisphosphate in mesenteric artery myocytes. J. Physiol. 2008, 586, 3087–3095. [Google Scholar] [CrossRef] [Green Version]
  46. Imai, Y.; Itsuki, K.; Okamura, Y.; Inoue, R.; Mori, M.X. A self-limiting regulation of vasoconstrictor-activated TRPC3/C6/C7 channels coupled to PI(4,5)P2-diacylglycerol signalling. J. Physiol. 2012, 590, 1101–1119. [Google Scholar] [CrossRef] [Green Version]
  47. Salamanca, D.A.; Khalil, R.A. Protein kinase C isoforms as specific targets for modulation of vascular smooth muscle function in hypertension. Biochem. Pharmacol. 2005, 70, 1537–1547. [Google Scholar] [CrossRef] [Green Version]
  48. Kwon, Y.; Hofmann, T.; Montell, C. Integration of phosphoinositide- and calmodulin-mediated regulation of TRPC6. Mol. Cell 2007, 25, 491–503. [Google Scholar] [CrossRef] [Green Version]
  49. Liu, M.; Large, W.A.; Albert, A.P. Stimulation of beta-adrenoceptors inhibits store-operated channel currents via a cAMP-dependent protein kinase mechanism in rabbit portal vein myocytes. J. Physiol. 2005, 562, 395–406. [Google Scholar] [CrossRef]
  50. Albert, A.P.; Liu, M.; Large, W.A. Dual effect of calmodulin on store-operated Ca2+ -permeable cation channels in rabbit portal vein myocytes. Br. J. Pharmacol. 2006, 148, 1001–1011. [Google Scholar] [CrossRef]
  51. Chen, I.S.; Dai, Z.K.; Welsh, D.G.; Chen, I.J.; Wu, B.N. Protein kinases modulate store-operated channels in pulmonary artery smooth muscle cells. J. Biomed. Sci. 2011, 18, 2. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Logothetis, D.E.; Petrou, V.I.; Zhang, M.; Mahajan, R.; Meng, X.Y.; Adney, S.K.; Cui, M.; Baki, L. Phosphoinositide control of membrane protein function: A frontier led by studies on ion channels. Annu. Rev. Physiol. 2015, 77, 81–104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. McLaughlin, S.; Wang, J.; Gambhir, A.; Murray, D. PIP2 and proteins: Interactions, organization, and information flow. Annu. Rev. Biophys. Biomol. Struct. 2002, 31, 151–175. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Gamper, N.; Shapiro, M.S. Target-specific PIP2 signalling: How might it work? J. Physiol. 2007, 582, 967–975. [Google Scholar] [CrossRef] [PubMed]
  55. Gallant, C.; You, J.Y.; Sasaki, Y.; Grabarek, Z.; Morgan, K.G. MARCKS is a major PKC-dependent regulator of calmodulin targeting in smooth muscle. J. Cell Sci. 2005, 118, 3595–3605. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Quinn, K.V.; Behe, P.; Tinker, A. Monitoring changes in membrane phosphatidyylinositol 4,5-bisphoshate in living cells using a domain from the transcription factor tubby. J. Physiol. 2008, 586, 2855–2871. [Google Scholar] [CrossRef] [PubMed]
  57. Balla, T.; Vamai, P. Visualization of cellular phosphoinositide pools with GFP-fused protein-domains. Curr. Protoc. Cell. Biol. 2009, 42. [Google Scholar] [CrossRef] [Green Version]
  58. Szentpetery, Z.; Balla, A.; Kim, Y.J.; Lemmon, M.A.; Balla, T. Live cell imaging with protein domains vapable of recognizing phosphatidylinositol 4,5-bisphosphate; a comparative study. BMC Cell Biol. 2009, 10, 67. [Google Scholar] [CrossRef] [Green Version]
  59. Balla, T. Phosphoinositides: Tiny lipids with giant impact on cell regulation. Physiol. Rev. 2013, 93, 1019–1137. [Google Scholar] [CrossRef]
  60. Park, C.Y.; Shcheglovitov, A.; Dolmetsch, R. The CRAC channel activator binds and inhibits L-type voltage-gated calcium channels. Science 2010, 330, 101–105. [Google Scholar] [CrossRef]
  61. Wang, Y.; Deng, X.; Mancarella, S.; Hendron, E.; Eguchi, S.; Soboloff, J.; Tang, X.D.; Gill, D.L. The calcium store sensor, STIM1, reciprocally controls Orai1 and CaV1.2 channels. Science 2010, 330, 105–109. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Jousset, H.; Rrieden, M.; Demaurex, N. STIM1 knockdown reveals that store-operated Ca2+ channels located close to sarco/endoplasmic Ca2+-ATPases (SECRA) pumps silently refull the endoplasmic tecticulum. J. Biol. Chem. 2007, 282, 11456–11464. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Ritchie, M.F.; Samakai, E.; Soboloff, J. STIM1 is required for attenuation of PMCA-mediated Ca2+ clearance during T-cell activation. EMBO J. 2012, 31, 1123–1133. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Lefkimmiatis, K.; Srikanthan, M.; Maiellaro, I.; Moyer, M.P.; Curci, S.; Hofer, A.M. Store-operated cyclic AMP signalling mediated by STIM1. Nat. Cell Biol. 2009, 11, 433–442. [Google Scholar] [CrossRef] [PubMed]
  65. Spassova, M.A.; Soboloff, J.; He, L.P.; Xu, W.; Dziadek, M.A.; Gill, D.L. STIM1 has a plasma membrane role in the activation of store-operated Ca2+ channels. Proc. Natl. Acad. Sci. USA 2006, 103, 4040–4045. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Thompson, J.L.; Shuttleworth, T.J. Exploring the unique features of the ARC channel a store-operated Orai channel. Channels 2013, 7, 364–373. [Google Scholar] [CrossRef] [Green Version]
  67. Yang, B.; Gwozdz, T.; Dutko-Gwozdz, J.; Bolotina, V.M. Orai1 and Ca2+-independent phospholipase A2 are required for store-operated Icat-SOC current, Ca2+ entry, and proliferation of primary vascular smooth muscle cells. Am. J. Physiol. Cell Physiol. 2012, 302, C748–C756. [Google Scholar] [CrossRef] [Green Version]
  68. Bolotina, V.M. Orai, STIM1 and iPLA2beta: A view from a different perspective. J. Physiol. 2008, 586, 3035–3042. [Google Scholar] [CrossRef]
  69. Avila-Medina, J.; Calderón-Sánchez, E.; González-Rodríguez, P.; Monje-Quiroga, F.; Rosado, J.A.; Castellano, A.; Ordonez, A.; Smani, T. Orai1 and TRPC1 Proteins Co-localize with CaV1.2 Channels to Form a Signal Complex in Vascular Smooth Muscle Cells. J. Biol. Chem. 2016, 291, 21148–21159. [Google Scholar] [CrossRef] [Green Version]
  70. Avila-Medina, J.; Mayoral-Gonzalez, I.; Dominguez-Rodriguez, A.; Gallardo-Castillo, I.; Ribas, J.; Ordonez, A.; Rosado, J.A.; Smani, T. The Complex Role of Store Operated Calcium Entry Pathways and Related Proteins in the Function of Cardiac, Skeletal and Vascular Smooth Muscle Cells. Front. Physiol. 2018, 9, 257. [Google Scholar] [CrossRef]
  71. Chakraborty, S.; Berwick, Z.C.; Bartlett, P.J.; Kumar, S.; Thomas, A.P.; Sturek, M.; Tune, J.D.; Obukhov, A.G. Bromoenol lactone inhibits voltage-gated Ca2+ and transient receptor potential canonical channels. J. Pharmacol. Exp. Ther. 2011, 339, 329–340. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Saleh, S.N.; Albert, A.P.; Large, W.A. Activation of native TRPC1/C5/C6 channels by endothelin-1 is mediated by both PIP3 and PIP2 in rabbit coronary artery myocytes. J. Physiol. 2009, 587, 5361–5375. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Proposed model of TRPC1-based store-operated channels (SOCs) in native contractile vascular smooth muscle cells (VSMCs). A, Receptor stimulation of distinct Gq-PLC and Gβγ-PI3K-PIP3 pathways lead to PKC-dependent phosphorylation of TRPC1 and channel opening by PIP2 and PIP3 respectively. Local levels of PIP2 involved in channel activation is controlled by MARCKS (M). B, Store depletion by receptor-mediated IP3 generation and store depleting agents such as CPA, BAPTA and TPEN activate TRPC1-based SOCs through a STIM1-TRPC1-mediated pathway. In unstimulated cells, SR Ca2+ stores are full and TRPC1-based SOCs are in a closed state. In this configuration, channels are associated with MARCKS which buffers local PIP2 levels and do not interact with Gq, PLCβ1 or STIM1. Following SR Ca2+ depletion, STIM1 (red) is activated and translocates from the SR to the plasma membrane where it interacts with TRPC1. Formation of STIM1-TRPC1 interactions enable binding and activation of Gq and PLCβ1 activity, PIP2 hydrolysis, DAG generation, stimulation PKCδ and PKC-dependent phosphorylation of TRPC1. This leads to dissociation of MARCKS from TRPC1 and release of PIP2 (previously protected from PIP2 hydrolysis) into the local environment where it acts as the activating ligand.
Figure 1. Proposed model of TRPC1-based store-operated channels (SOCs) in native contractile vascular smooth muscle cells (VSMCs). A, Receptor stimulation of distinct Gq-PLC and Gβγ-PI3K-PIP3 pathways lead to PKC-dependent phosphorylation of TRPC1 and channel opening by PIP2 and PIP3 respectively. Local levels of PIP2 involved in channel activation is controlled by MARCKS (M). B, Store depletion by receptor-mediated IP3 generation and store depleting agents such as CPA, BAPTA and TPEN activate TRPC1-based SOCs through a STIM1-TRPC1-mediated pathway. In unstimulated cells, SR Ca2+ stores are full and TRPC1-based SOCs are in a closed state. In this configuration, channels are associated with MARCKS which buffers local PIP2 levels and do not interact with Gq, PLCβ1 or STIM1. Following SR Ca2+ depletion, STIM1 (red) is activated and translocates from the SR to the plasma membrane where it interacts with TRPC1. Formation of STIM1-TRPC1 interactions enable binding and activation of Gq and PLCβ1 activity, PIP2 hydrolysis, DAG generation, stimulation PKCδ and PKC-dependent phosphorylation of TRPC1. This leads to dissociation of MARCKS from TRPC1 and release of PIP2 (previously protected from PIP2 hydrolysis) into the local environment where it acts as the activating ligand.
Cells 09 00179 g001

Share and Cite

MDPI and ACS Style

Martín-Aragón Baudel, M.A.S.; Shi, J.; Large, W.A.; Albert, A.P. Insights into Activation Mechanisms of Store-Operated TRPC1 Channels in Vascular Smooth Muscle. Cells 2020, 9, 179. https://doi.org/10.3390/cells9010179

AMA Style

Martín-Aragón Baudel MAS, Shi J, Large WA, Albert AP. Insights into Activation Mechanisms of Store-Operated TRPC1 Channels in Vascular Smooth Muscle. Cells. 2020; 9(1):179. https://doi.org/10.3390/cells9010179

Chicago/Turabian Style

Martín-Aragón Baudel, Miguel A. S., Jian Shi, William A. Large, and Anthony P. Albert. 2020. "Insights into Activation Mechanisms of Store-Operated TRPC1 Channels in Vascular Smooth Muscle" Cells 9, no. 1: 179. https://doi.org/10.3390/cells9010179

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop