European Journal of Cell Biology 101 (2022) 151208 Contents lists available at ScienceDirect European Journal of Cell Biology journal homepage: www.elsevier.com/locate/ejcb Review Metabolic regulation and dysregulation of endothelial small conductance calcium activated potassium channels Shawn Kant, Frank Sellke, Jun Feng * Division of Cardiothoracic Surgery, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI, USA A R T I C L E I N F O A B S T R A C T Keywords: EDHF Endothelial dysfunction SK channel Diabetes Hypertension Ischemia-reperfusion The vascular endothelium is an important regulator of vascular reactivity and preserves the balance between vasoconstrictor and vasodilator tone during normal physiologic conditions. Example endothelial-derived vaso­ constrictors include endothelin-1 and thromboxane A2; example vasodilators include nitric oxide and prosta­ cyclin. A growing body of evidence points to the existence of a non-nitric oxide, non-prostacyclin endotheliumderived vasodilatory factor of currently unclear identity, often referred to as endothelium-derived hyper­ polarizing factor (EDHF). Recent research testifies to the significance of EDHF in endothelium-dependent vascular smooth muscle relaxation. Special emphasis has been placed on the role of small conductance calcium-activated potassium channels (SK) in facilitating the endothelial and vascular responses to EDHF across the microcirculation, including coronary, mesenteric, and pulmonary vascular beds. Meanwhile, decreased ac­ tivity of endothelial SK channel activity has been implicated in the pathology of a variety of disease states that alter the balance between vasodilator and vasoconstrictor tone. Hence the primary goal of this review is to characterize the physiology of endothelial SK channels in the microvasculature under normal and pathological conditions. Themes of regulation and dysregulation of SK channel activity through the action of protein kinases, reactive oxygen species, and byproducts of intermediary metabolism provide unifying principles to tie together vascular pathology in altered metabolic states ranging from hypertension to diabetes, to ischemia-reperfusion. A comprehensive understanding of SK channel pathophysiology may provide a foundation for development of new therapeutics targeting SK channels, particularly SK channel potentiators, that may have widespread application for many chronic disease states. 1. Introduction Consisting of a single layer of cells lining the intima of blood vessels, the vascular endothelium influences multiple important physiological processes, including metabolism, vascular permeability, vascular tone, hemostasis, and inflammation. Preservation of balance, be it between thrombosis and fibrinolysis, between promotion and inhibition of leukocyte extravasation, and most importantly, between vasodilation and vasoconstriction, is a key principle that unifies the many functions of the vascular endothelium (Krüger-Genge et al., 2019). With respect to regulation of vascular tone, the endothelium produces a variety of short and long-acting factors that have important effects on vascular smooth muscle. Notable endothelium-derived vasoconstrictors include endothelin-1 and thromboxane A-2 (Kvietys and Granger, 2014). Notable endothelium-derived vasodilators include nitric oxide (NO), prostacyclin (PGI2) and endothelium-derived hyperpolarizing factor (EDHF), Most vasoconstrictors, like endothelin-1, act on cell surface receptors that promote vascular smooth muscle depolarization. For example, endothelin binding to ETA receptors on vascular smooth muscle cells activates a phospholipase C-protein kinase C driven pathway that ulti­ mately elevates intracellular calcium levels (through release from sarcoplasmic reticulum stores) to trigger smooth muscle contraction (Kowalczyk et al., 2014). In contrast, most endothelium-derived vaso­ dilators act either by (1) inhibiting depolarization-induced smooth muscle contraction or (2) inducing endothelial and vascular smooth muscle hyperpolarization. For an example of (1), consider NO. NO produced by endothelial nitric oxide synthase (eNOS) stimulates vascular smooth muscle guanylyl cyclase, which converts GTP to cyclic GMP (cGMP) (Chen et al., 2008). cGMP activates protein kinase G, which can promote closure of vascular smooth muscle membrane voltage gated calcium channels and promote calcium uptake by * Correspondence to: Cardiothoracic Surgery Research Laboratory, Rhode Island Hospital, 1 Hoppin Street, Coro West Room 5229, Providence, RI 02903, USA. E-mail address: jfeng@lifespan.org (J. Feng). https://doi.org/10.1016/j.ejcb.2022.151208 Received 27 December 2021; Received in revised form 6 February 2022; Accepted 7 February 2022 Available online 8 February 2022 0171-9335/© 2022 Published by Elsevier GmbH. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/). S. Kant et al. European Journal of Cell Biology 101 (2022) 151208 BK channels (KCa1.1) (Kshatri et al., 2018; Yang et al., 2015; Lee and Cui, 2010). sarcoplasmic reticulum calcium-ATPases (SERCA) (Chen et al., 2008; Cohen et al., 1999). Reduced intracellular calcium levels prevent calcium-induced smooth muscle contraction during depolarization. PGI2 acts in a similar fashion by activating adenylyl cyclase, causing increased con­ version of ATP to cyclic AMP (cAMP), after which cAMP activates pro­ tein kinase A (PKA). PKA promotes sarcoplasmic reticulum calcium uptake and inhibits myosin light chain kinase via phosphorylation, leading to smooth muscle relaxation and vasodilation (Majed and Khali, 2012). For an example of (2), consider EDHF. Although its specific identity remains a matter of intense debate, extensive research has confirmed the presence of a non-NO, non-PGI2 inducer of vasodilation that acts on vascular beds by increasing potassium conductance through opening of endothelial and smooth muscle potassium channels (Bryan et al., 2005; Busse et al., 2002; Félétou and Vanhoutte, 2006; Feletou and Vanhoutte, 2007). Application of potassium channel agonists to blood vessels mimics the effects of EDHF, and pharmacological blockade of multiple distinct types of potassium channels, in particular calcium-activated potassium channels, prevents non-NO, non-PGI2 dependent vasodila­ tion (Grgic et al., 2009). Efflux of potassium along its electrochemical gradient results in cell membrane hyperpolarization that can be communicated from endothelial cells to vascular smooth muscle cells via gap junctions; this has been called the “myoendothelial coupling” hypothesis (Yamamoto et al., 1999; Sandow et al., 2004). Alternatively, EDHF may stimulate inward rectifier potassium channels (KIR) and the sodium potassium ATPase on vascular smooth muscle cells which could also promote membrane hyperpolarization (Coleman et al., 2004). Altogether, most putative explanations of the action of EDHF include, in some capacity, the activation of endothelial calciumactivated potassium channels (KCa) as critical mediators of the final vasodilatory pathway. Two broad categories of calcium-activated po­ tassium channels have been identified: small/intermediate conductance (SK/IK) and large conductance (BK) calcium activated potassium channels. SK channel-mediated vasodilation plays significant roles across many vascular beds, most notably in the microcirculation. This review will examine the specific roles of calcium-activated potassium channels in endothelial function, with particular emphasis on the SK and IK channels that are most heavily implicated in endothelium-dependent vasodilation. We will consider aspects of normal physiology and phar­ macology before turning to pathophysiology, more specifically the dysregulation of SK/IK channels in conditions that alter metabolic ho­ meostasis, such as diabetes, hypertension, and hypoxia-reperfusion injury. 2.2. Endothelial localization of calcium-activated potassium channels Extensive research has confirmed that SK and IK channels are constitutively expressed on vascular endothelial cells in humans and a variety of animals (Félétou, 2009; Dalsgaard et al., 2009; Steffensen et al., 2017; Bychkov et al., 2002; Burnham et al., 2002). Of the different subtypes of SK channels, KCa2.3 (SK3) and KCa3.1 (IK) are particularly abundant in endothelia (Kohler et al., 1996; Sandow et al., 2006; Ber­ tuccio et al., 2018; Dora et al., 2008). More specifically, KCa2.3 and KCa3.1 have been found in rat and pig carotid arteries, rat mesenteric arteries, human small pulmonary arteries, and human coronary arteries (Takai et al., 2013; Kroigaard et al., 2012; Burnham et al., 2002; Sandow et al., 2006; Dora et al., 2008; Köhler et al., 2001; Wulff and Köhler, 2013; Liu et al., 2015). BK channels have been definitively identified in the vascular smooth muscle of arteries and arterioles, where they contribute to vasodilation and relaxation (Yang et al., 2011; Dopico et al., 2018; Wu and Marx, 2010). There, BK channels trigger a negative feedback loop in response to depolarization induced calcium influx and smooth muscle contrac­ tion, thereby helping regulate the degree of myogenic tone (Dopico et al., 2018). However, when it comes to endothelial cells, the presence of BK channels becomes more complicated. Although most studies have not found BK channels in the vascular endothelium, some electrophys­ iology and molecular biology experiments have detected BK channels in certain human endothelial cell lines, more specifically umbilical and microvascular endothelia (Grgic et al., 2005; Papassotiriou et al., 2000; Zyrianova et al., 2021). Nonetheless, most work to date has focused on SK and IK channels in the context of endothelial function, given their confirmed presence; hence, the focus of this review. 2.3. Structure and activation of SK and IK channels Having completed an exploration of the localization of endothelial SK channels in the circulation, we will move on to discuss some prin­ ciples governing their molecular structure. The general structure of SK channels resembles voltage-gated potassium channels, insofar as the framework is a tetrameric assembly (Nam et al., 2017). Detailed analysis from protein purification assays show that the SK tetramer is symmetric, with a length of approximately 95 angstroms and a width of 120 ang­ stroms in the plane of the plasma membrane (Lee and MacKinnon, 2018). Different genes encode the SK alpha subunits for the various SK channel subtypes. KCNN1, KCNN2, and KCNN3 genes encode the SK alpha subunits of SK1, SK2, and SK3 respectively (Kohler, 1996). Each SK alpha subunit consists of six hydrophobic transmembrane alpha he­ lical domains (S1-S6). A pore forming p-loop between the S5 and S6 domains governs the potassium selectivity of the overall channel (Fig. 1) (Nam et al., 2017). Moreover, the pore-forming regions of the alpha subunits always face the channel center. Amino (N) and carboxy (C) terminal domains of the alpha subunits are cytosolic, and calmodulin binding domains lie at the proximal C terminal end of S6. An important feature that distinguishes SK and IK channels from BK channels is their voltage-independence, as they do not possess any voltage sensor domains. Instead, the chief regulator of SK and IK channel activity is calcium, acting through a calmodulin-dependent mechanism. Therefore, it should not be surprising that many SK channels are located in close proximity to intracellular calcium stores and cell membrane calcium channels (Stocker, 2004; Adelman et al., 2012). Note that the lack of voltage gating suggests that SK channels will not inactivate at negative membrane potentials. Thus, if left open without modulation, they will facilitate a hyperpolarization response that moves cell mem­ brane potential towards the potassium equilibrium potential. Anywhere from two to eight calmodulin molecules can bind to any one SK channel (Halling et al., 2014). Meanwhile, only one calmodulin 2. Structure, Localization, and Function of SK channels 2.1. Categories calcium-activated potassium channels The first part of this review will present the localization and funda­ mental cellular/molecular characteristics of endothelial SK channels. We begin with a general overview of the different subcategories of SK channels. The fundamental role of calcium-activated potassium chan­ nels is to couple changes in intracellular calcium concentrations to cell membrane hyperpolarization. This in turn allows for a physiological “brake” on runaway excitation that might otherwise occur in the pres­ ence of persistently elevated intracellular calcium. As mentioned before, there are two broad families of calcium-activated potassium channels: the small/intermediate conductance (SK/IK) and the large conductance (BK) channels (Wei et al., 2005). Within the SK/IK family, four subtypes of channels have been identified: SK1 (KCa2.1), SK2 (KCa2.2), SK3 (KCa2.3), and SK4 (KCa3.1). The latter, SK4, is sometimes referred to as IK1 due to its higher unitary conductance (up to 40–45 pS) relative to SK1–3 (all in the range of 5–15 pS) (Maylie et al., 2004; Grgic et al., 2009; Kshatri et al., 2018). However, all the SK/IK channels exhibit lower unitary conductance when compared to the 100–300 pS range of 2 S. Kant et al. European Journal of Cell Biology 101 (2022) 151208 Fig. 1. General SK Channel structure. Four pore-forming alpha subunits form the SK channel complex (right). Each alpha subunit consists of 6 transmembrane domains (S1-S6) (left), with a pore forming loop (P-loop) between S5 and S6. S4 is an important calcium sensing domain. Note N and C termini, with a calmodulin (CaM) binding domain at the C terminus of each alpha subunit. Created with BioRender.com. can bind to each SK subunit, resulting in a maximum of four bound calmodulins per tetramer (Lee and MacKinnon, 2018). Curiously, ex­ periments aiming to purify SK channels from human cells also copurified calmodulin in the presence and absence of calcium, suggesting that calcium is not necessary per se for calmodulin-SK channel binding (which appears to be constitutive). Indeed, research shows that calcium binding to the N-lobes of calmodulin triggers a structural change in the bound calmodulin-SK channel-calmodulin-binding-domain conforma­ tion (Lee and MacKinnon, 2018). This conformational shift alters the arrangement of the alpha helices that form the SK channel pore, leading to channel opening and permitting the flow of potassium. 2000). Therefore, even though some studies (as mentioned earlier) al­ lude to the presence of endothelial BK channels, results such as those produced by Kohler et al. support the notion that BK channels do not have a significant role in endothelial hyperpolarization. Turning to genetic knockout models, deletion of the gene encoding IK channels in mice results in significantly impaired KCa currents and impaired hyperpolarization in the carotid endothelium (Si et al., 2006). In addition, deletion of endothelial SK3 channel genes in mice produces impaired endothelium-derived hyperpolarization, impaired vaso­ relaxation, and altered calcium dynamics (Yap et al., 2016a, 2016b). Furthermore, treatment with doxycycline, shown to alter KCa2.3 channel expression and activity, also virtually abolishes KCa currents in mouse models (Grgic et al., 2009). On the other hand, overexpression of SK3 in transgenic mouse models produces significantly elevated KCa currents and vascular hyperpolarizing responses which result in mark­ edly reduced myogenic tone (Taylor et al., 2003). 2.4. Function of SK and IK channels in endothelium-dependent hyperpolarization 2.4.1. Pharmacological and genetic studies The second part of this review will explore the physiologic function of endothelial SK channels in the human body and their associated signaling pathways in normal/healthy conditions. We begin by consid­ ering an assortment of pharmacologic and genetic studies that have examined the effects of altered SK channel activity and expression on endothelial hyperpolarization. First, application of highly selective KCa3.1 inhibitors 1-[(2-chlorophenyl) diphenylmethyl]-1H-pyrazole (TRAM-34) and 2-(2-chlorophenyl)-2,2-diphenylacetonitrile (TRAM39) abolish IK-mediated potassium currents in response to acetylcholine in rat carotid endothelia (Eichler et al., 2003). Moreover, application of TRAM-34 alone or TRAM-39 plus SK3 channel inhibitor apamin blocks nitric oxide and prostacyclin independent vasodilation (Eichler et al., 2003). These findings have been replicated in the context of human coronary artery endothelial cells and the human skeletal muscle microvasculature (Liu et al., 2015, 2008). In addition, application of the selective SK/IK channel activator NS309 increases endothelial SK/IK currents and promotes arteriolar vasodilation in the coronary and skeletal microvasculature (Liu et al., 2015, 2008). It is also worthwhile to note that application of iberiotoxin, a selective KCa1.1 (BK) channel blocker, appears to have little, if any, effect on endothelial hyperpo­ larization when tested in human mesenteric arteries (Köhler et al., 2.4.2. EDHF and SK/IK channel activity In the introduction, we introduced the concept of EDHF, a NOindependent, PGI2-independent vasodilator that acts in a variety of vascular beds through activation of SK and IK channels. (Félétou, 2009; Garland et al., 2011). While the specific identity of EDHF remains un­ clear, several potential candidates, such as potassium itself, ananda­ mide, hydrogen peroxide, C-natriuretic peptide, and epoxyeicosatrienoic acids (EETs), have been proposed based on current theories regarding the mechanism of EDHF-driven vascular smooth muscle relaxation (Bellien et al., 2008). A synthesis of the existing literature results in the following proposal (Goto et al., 2018; Bellien et al., 2008; Luksha et al., 2004; Luksha et al., 2009; Ozkor and Quyyumi, 2011). Essentially, endothelium specific agonists, such as acetylcholine, bradykinin, and substance P, bind to receptors on the vascular endothelium, mobilize release of calcium from intracellular stores into the cytosol, and potentially induce synthesis of EDHF. Acetylcholine acts on endothelial cells via muscarinic M1 or M3 re­ ceptors, which are G-protein coupled receptors (Tangsucharit et al., 2016; Walch et al., 2001; Furchtott and Furchgott and Zawadzki, 1980). Binding of acetylcholine to M1 or M3 receptors leads to activation of Gq 3 S. Kant et al. European Journal of Cell Biology 101 (2022) 151208 and phospholipase-C (PLC) mediated signal transduction cascade. PLC cleaves plasma membrane bound PIP2 to IP3 and DAG; IP3 moves through the cytosol to the smooth endoplasmic reticulum and induces release of stored calcium into the cytosol. Note, however, that M3 re­ ceptor activity on endothelial cells also stimulates endothelial nitric oxide synthase in addition to promoting elevated intracellular calcium; hence acetylcholine may also promote non-EDHF mediated vasorelaxation. Bradykinin stimulates endothelial bradykinin B2 receptors, which increase free cytosolic calcium levels and promotes release of prosta­ cyclin, NO, and potentially EDHF (Gryglewski et al., 2002; Hornig and Drexler, 1997). Like acetylcholine, bradykinin receptor activation in­ creases cytosolic calcium through a PLC-mediated pathway. Two addi­ tional points are worth noting. First, among the all the various endothelial agonists, bradykinin has been shown to be a particularly potent stimulator of intracellular calcium release. Two, like acetylcho­ line, bradykinin also promotes non-EDHF pathways of vasorelaxation (e. g., through endothelial nitric oxide synthase stimulation) (Hornig and Drexler, 1997). Meanwhile, substance P mobilizes intracellular calcium stores and promotes calcium influx into endothelial cells, potentially through action of endothelial neurokinin 1 receptors, although the precise details of this process remain murky (Greeno et al., 1993; Sharma and Davis, 1995; Kuroiwa et al., 1995). Akin to bradykinin and acetylcholine, it is also possible that besides increasing intracellular calcium, substance P promotes synthesis of a separate EDHF substance. Vessel wall shear stress due to blood flow impinging on the endo­ thelium, and the resulting mechanical stretch, can also trigger endo­ thelial signaling pathways, including laminar shear-stress induced endothelial nitric oxide synthase phosphorylation and activation, stim­ ulation of COX-2 synthesis of PGI2, and EDHF-mediated vasodilation (Lu and Kassab, 2011; Bellien et al., 2008). The combination of elevated intracellular calcium and increasing synthesis of EDHF work to stimu­ late calcium activated potassium channels through a few potential pathways. For example, EDHF itself may passively diffuse from endothelial cells to vascular smooth muscle cells and activate vascular smooth muscle BK channels to trigger hyperpolarization (Bellien et al., 2008). Alterna­ tively, EDHF itself may further elevate endothelial cell calcium levels, such as through stimulation of transient receptor potential (TRP) V4 channels that promote calcium influx from the extracellular environ­ ment (Ozkor and Quyyumi, 2011). This would elevate intracellular calcium levels beyond the action of the previously discussed endothelial agonists, resulting in enhanced calcium-induced opening of endothelial SK and IK channels (e.g., via the calcium-calmodulin binding mecha­ nism to SK/IK channel C termini). Release of potassium through endo­ thelial SK and IK channels produces the characteristic current observed in SK/IK channel stimulation experiments, and results in endothelial hyperpolarization. This hyperpolarization is then directly conducted to vascular smooth muscle via myoendothelial gap junctions to trigger smooth muscle hy­ perpolarization. Or, the efflux of potassium from endothelial cells into the subendothelial space can itself stimulate vascular smooth muscle inward rectifying potassium channels or sodium-potassium ATPase pumps that trigger smooth muscle hyperpolarization. Ultimately, as is self-evident, all these different EDHF pathways converge on endothelial SK channel activation and smooth muscle hyperpolarization that leads to closure of voltage-gated calcium channels on vascular smooth muscle cells (Ozkor and Quyyumi, 2011; Bellien et al., 2008; Garland et al., 2011). Closure of voltage gated calcium channels lowers smooth muscle cell intracellular calcium levels, inhibiting vasoconstriction and pro­ moting vasodilation. While it is well established that EDHF is a critical component of this framework for endothelial hyperpolarization, uncertainty regarding the specific place of EDHF in these pathways contributes a great deal to the controversy surrounding its identity. For example, if EDHF activates SK channels through increasing intracellular calcium, then molecules like EETs are strong candidates for EDHF because they can stimulate cell membrane TRPV4 channels that mediate calcium influx. Alternatively, because potassium released through SK channels can itself trigger endothelial-dependent vascular smooth muscle relaxation, it is possible that so-called “EDHF” is, in fact, simply, potassium. 2.5. Physiologic regulation of endothelial SK/IK channel activity Any discussion of SK channel function would be incomplete without mentioning the many regulatory mechanisms that modulate SK channel activity. First and foremost is calcium. Calcium is the most important ionic regulator of SK channels. Hence the name “calcium sensitive” potassium channels, and the instrumental role of cytosolic calcium in triggering activation of SK channels via mechanisms detailed earlier. Studies have shown co-localization of SK channels with L-type voltage gated cell membrane calcium channels, such as Cav1.3 and Cav1.2, lending credence to the notion of a close link between voltage gated calcium channel opening and SK channel activity (Lu et al., 2007). Experimental manipulation of intracellular calcium also affects SK channel trafficking, as shown by increased trafficking of SK channels to the plasma membrane of rabbit pulmonary veins in response to rising intracellular calcium (Ozgen et al., 2007). However, calcium is not the only ion that affects SK channel activity—other divalent cations do so as well. Indeed, this stems from the ability of other divalent cations, such as magnesium, to bind to charged inner pore residues of the SK channel S6 transmembrane domain, which are important regulators of SK channel sensitivity and open probability (Li and Aldrich, 2011). EDHF-SK/IK channel induced vasodilation may be inhibited by nitric oxide, itself a strong vasodilator. For example, experiments on pig cor­ onary arterioles show attenuated EDHF-mediated dilation after treat­ ment with nitric oxide donors, while endothelium-independent vasodilation was preserved (Bauersachs et al., 1996). Other experiments with nitric oxide synthase inhibitors in dog coronary arterioles produced comparable results (Nishikawa et al., 2000). Curiously, some evidence suggests that activation of SK3 and IK channels may promote endothelial nitric oxide synthase activation. Indeed, the IK channel activator NS309 has been shown to both produce endothelial and smooth muscle hy­ perpolarization AND increase nitric oxide formation in human endo­ thelial cells and porcine retinal arterioles (Balut et al., 2012; Dalsgaard et al., 2010; Schmidt et al., 2010). This may be a built-in mechanism of SK channel autoregulation to mitigate against the adverse effects of runaway activity. The specific means by which SK/IK channel activity bolster nitric oxide synthesis remain a matter of investigation; one hypothesis may be a paradoxical increase in the ion driving force for calcium across endothelial cell membrane calcium channels and coupling of calcium to eNOS stimulation. However, there is conflicting data on this matter (Dalsgaard et al., 2010). Overall, the interaction between nitric oxide and EDHF is an unresolved matter, and further work needs to be done to better characterize their relationship. Protein kinase C has also demonstrated some ability to modulate SK channel activity. Experiments aiming to isolate SK2/SK3 channel bind­ ing proteins via mass spectrometry, binding assays, and coimmunoprecipitation revealed catalytic and regulatory subunits of protein kinase CK2 and protein phosphatase 2 A constitutively bind to the cytoplasmic C and N termini of the SK channel protein (Bildl et al., 2004). Recall that calmodulin constitutively binds to the SK channel protein C terminus. Protein kinase CK2 phosphorylation of SK channel-bound calmodulin at the threonine 80 residue produces an almost 5-fold reduction in SK channel calcium sensitivity, leading to channel inhibition (Bildl et al., 2004; Allen et al., 2007). Note that in-vitro assays testing protein kinase CK2 interaction with SK2 have shown that protein kinase CK2 only appears to phosphorylate calmodulin when complexed with the SK channel calmodulin binding domain and does not phosphorylate the SK channel calmodulin binding domain alone without bound calmodulin (Allen et al., 2007). 4 S. Kant et al. European Journal of Cell Biology 101 (2022) 151208 Furthermore, with respect to SK2 channels, the N terminal domain lysine 121 residue has been shown to activate SK2-bound protein kinase CK2, which suggests that some intrinsic activation/feedback mecha­ nisms may be at play between the channel and its bound protein kinase CK2 (Allen et al., 2007). Meanwhile, activity of bound protein phos­ phatase 2A to dephosphorylate SK channel-bound calmodulin reverses the inhibitory effects of protein kinase CK2 on calcium sensitivity, shifting the channel’s state towards a heightened open probability. Note that most of the aforementioned experiments have been performed on SK2 and SK3 channels. Therefore, verifying these findings for SK1 and IK channels will assist with drawing firmer generalizations about the definitive place of protein kinase C in SK channel regulation. circulating levels of inflammatory markers such as CRP, TNF-alpha, and IL-6 (Piga et al., 2007; Festa et al., 2000). Hyperglycemia also blunts endothelial relaxation responses across different vascular beds in animal models and humans with type 1 and type 2 diabetes (Vriese et al., 2000). This may occur through several mechanisms, including elevated levels of oxygen-derived free radicals, reduced expression of endothelial nitric oxide synthase (eNOS), reduced phosphorylation of the eNOS active site Ser1177, or increased phos­ phorylation of the eNOS inhibitory site Thr495 (Shi and Vanhoutte, 2009; Matsumoto et al., 2014). Conversely, diabetes may promote increased responsiveness to endothelial constricting factors, such as endothelin-1, thereby increasing arterial stiffness (Tabit et al., 2010). Thus, diabetes disrupts the balance between endothelial relaxation and constriction in favor of constriction, leading to impaired vasomotor tone. Diabetes may also hamper vasorelaxation through direct dysregula­ tion of electrochemical signaling, and it is here that potentially pivotal roles for SK channels and EDHF become apparent (Burnham et al., 2006; Weston et al., 2008). Reduced EDFH-mediated vascular smooth muscle relaxation and reduced SK/IK channel currents have been noted in an assortment of rodent models of type 1 (Morikawa et al., 2005; Hosoya et al., 2010; Ding et al., 2005; Csanyi et al., 2007; Matsumoto and Wakabayashi et al., 2004; Shi, 2006; Zhu et al., 2010; Leo et al., 2011; Makino et al., 2000; Wigg et al., 2001; Absi et al., 2013; Ma et al., 2013; Sotnikova et al., 2011a, 2011b; De Vriese et al., 2000; Gokina et al., 2015) and type 2 (Burnham et al., 2006; Brøndum et al., 2009; Weston et al., 2008; Schach et al., 2014a, 2014b; Yin et al., 2017a, 2017b, 2017c; Cotter et al., 2010; Matsumoto et al., 2010; Oniki et al., 2013; Oniki et al., 2006; Matsumoto et al., 2006; Matsumoto et al., 2008; Matsumoto et al., 2009; Zhang et al., 2020; Xing et al., 2021; Song et al., 2020; Park et al., 2008) diabetes (Tables 1–4). Similar results have been found across a variety of different vascular beds in these rodent models, including the mesenteric circulation (Burnham et al., 2006; Brøndum et al., 2009; Weston et al., 2008; Schach et al., 2014a, 2014b; Morikawa et al., 2005; Hosoya et al., 2010; Ding et al., 2005; Leo et al., 2011; Makino et al., 2000; Wigg et al., 2001; Absi et al., 2013; Ma et al., 2013; Sotnikova et al., 2011a, 2011b; Matsumoto et al., 2010; Oniki et al., 2013; Oniki et al., 2006; Matsumoto et al., 2006; Matsumoto et al., 2008; Matsumoto et al., 2009; Shi, 2006), renal arcuate arteries (Yin et al., 2017a, 2017b, 2017c; Cotter et al., 2010; De Vriese et al., 2000), aortic endothelial cells ( Matsumoto and Wakabayashi et al., 2004), carotid arteries (Shi, 2006), femoral arteries (Shi, 2006), cavernous tissue endothelial cells (Zhu et al., 2010), and uterine vasculature (Gokina et al., 2015). In other vascular beds, the effect of diabetes on SK channel activity and EDHF responsiveness is less clear, at least based on observations in rodent models. For instance, Mishra et al. report preserved cremaster skeletal muscle arteriolar function in their type 2 diabetic GK rats (Mishra et al., 2021a, 2021b). Next, while several studies report that SK channel activity, along with endothelium-dependent vascular relaxa­ tion, decreases in coronary microvessels of genetically modified type 2 diabetic mice (Zhang et al., 2020; Xing et al., 2021; Song et al., 2020), others report no significant changes (Park et al., 2008). Likewise, Kaji­ kuri et al. report preserved EDHF responsiveness in LAD coronary ar­ teries of OLETF type 2 diabetic rats (Kajikuri et al., 2009). Importantly, the coronary microcirculation is one of the few vascular beds where SK channel investigations have also been done using human tissue, more specifically atrial microvessels (Table 4)—and those studies universally demonstrate decreased SK channel currents and decreased responsive­ ness to EDHF-mediated relaxation in diabetic vs nondiabetic coronary arterioles (Liu et al., 2015, 2018, 2020). Evidence for altered endothelial SK channel gene or protein expres­ sion in diabetes is extremely mixed. In some experiments, SK gene/ protein expression paradoxically increased even as overall EDHF responsiveness decreased in diabetic rodent mesenteric vessels (Burn­ ham et al., 2006; Weston et al., 2008; Schach et al., 2014a, 2014b). Some 3. SK channel pathology and endothelial dysfunction In the final section of this review, we move from SK channel physi­ ology to pathophysiology, with specific attention paid to endothelial dysfunction in disease states involving altered metabolism. Endothelial dysfunction, in the form of micro and macrovascular complications, is a hallmark of many disease processes. Earlier, we mentioned several ho­ meostatic functions of the vascular endothelium (thrombosis and coagulation, regulation of vascular tone, angiogenesis, immune modu­ lation). Pathological changes in these homeostatic functions occur with endothelial dysfunction, characterized by impaired vasodilation, vascular leakage, promotion of thrombosis, and promotion of inflam­ mation (Rajendran et al., 2013). Although not routinely done in the clinic, one can measure degree of endothelial dysfunction by assessing the ability of vessels to dilate or constrict in response to drug adminis­ tration (e.g., phenylephrine). Characterizing the molecular basis of endothelial dysfunction is an active focus of research. Given that endothelial dysfunction is itself a feature of many other systemic conditions rather than an isolated finding of its own, the initiating factors for endothelial dysfunction may differ under different circumstances. For example, endothelial dysfunction in bacterial sepsis is often driven by interactions between lipopolysac­ charide, TNF-alpha, and endothelial toll-like receptors that promote reactive oxygen species (ROS) production, oxidative stress, endothelialimmune activation, and apoptosis (Peters, 2003). Alternatively, in atherosclerosis, sudden rupture of plaques can alter blood vessel he­ modynamics, activating the endothelium into a more pro-thrombotic, pro-inflammatory phenotype (e.g., through heightened NF-kB expres­ sion, exposure of subendothelial collagen, and platelet activation) (Gimbrone and García-Cardeña, 2016; Davignon and Ganz, 2004). Although the precipitating events of endothelial dysfunction may be different in different situations, a growing body of literature suggests that there are shared phenotypic features that underpin endothelial dysfunction across several diseases, chiefly cardiovascular diseases that involve altered metabolic phenotypes. One such feature would be changes in the expression/function of endothelial cell membrane SK/IK channels. Hence the remainder of this review will elaborate on the roles of SK/IK channel dysfunction in three major pathological states that all involve aberrant or disrupted metabolic processes: diabetes, hyperten­ sion, and hypoxia-reperfusion. 3.1. Diabetes and SK channel dysfunction The first major metabolic disease state highlighted here is hyper­ glycemia in the context of diabetes, obesity, and metabolic syndrome. Cardiovascular complications of uncontrolled/untreated hyperglycemia are well-known contributors to the increased morbidity and mortality rates of diabetic patients in comparison to the general population (Shi and Vanhoutte, 2017; Wilson et al., 2005). First, diabetes compromises endothelial barrier integrity by downregulating cell junction protein expression, leading to altered vascular permeability (Li et al., 2016; Chapouly et al., 2015). Second, diabetes induces a systemic inflamma­ tory state, for hyperglycemia can activate the endothelium and elevate 5 S. Kant et al. European Journal of Cell Biology 101 (2022) 151208 Table 1 SK channels in zucker obese/fatty rat model of type 2 diabetes. Animal Model Tissue and Cell Type SK Protein Expression in Diabetes SK Channel Gene Expression Vascular Function/ EDHF responsiveness SK/IK Channel Recording References Zucker Diabetic Fatty Rat, type 2 DM Zucker Diabetic Fatty rat, Type 2 DM Zucker Diabetic Fatty rat, Type 2 DM Zucker Diabetes fatty rat (ZDF) Zucker Diabetic Fatty Rats Mesenteric artery endothelial cells Mesenteric artery endothelial cells Mesenteric artery endothelial cells Mesenteric artery endothelial cells Renal arcuate artery endothelial cells N/A SK3: Increased IK: No change N/A Small mesenteric arteries: decreased Small mesenteric arteries: decreased Decreased IK: No change SK: Decreased N/A Burnham et al. (2006) Brondum et al., 2010 N/A Weston et al., 2008 decreased N/A Schach et al. (2014) Renal arcuate arteries: Decreased N/A Yin et al. (2017); Cotter et al. (2010) No significant differences IK: Increased IK: Increased Increased IK increased, SK no change N/A N/A Table 2 SK channels in variations of the streptozotocin-induced type 1 diabetic rat model. Animal Model Tissue and Cell Type SK Protein Expression in Diabetes SK Channel Gene Expression Vascular Function/ EDHF responsiveness SK/IK Channel Recording References Streptozotocininduced APOE deficient rats Streptozotocininduced APOE deficient rats Streptozotocininduced diabetic rats Mesenteric artery endothelial cells N/A N/A Small mesenteric arteries: decreased N/A Morikiawa et al., 2005; Hosoya et al. (2010) Mesenteric artery endothelial cells N/A Small mesenteric arteries: decreased N/A Ding et al. (2005) Aorta endothelial cells N/A SK2 and SK3: Decreased IK: No change N/A Thoracic aorta: Decreased N/A Streptozotocin induced diabetic rats Carotid, femoral, mesenteric artery endothelial cells Cavernous tissue endothelial cells Mesenteric artery endothelial cells N/A N/A N/A SK3: Decreased IK1: Decreased N/A SK3: Decreased IK1: Decreased N/A More significantly decreased EDHF in carotid and femoral vs mesenteric arteries Cavernous tissue: Decreased Csanyi et al. (2007); Matsumoto, Wakabayashi et al. (2004) Shi (2006) N/A Zhu et al. (2010) Small mesenteric arteries: decreased N/A Mesenteric artery endothelial cells Mesenteric artery endothelial cells and aorta Renal microcirculation endothelial cells Uterine vasculature SK3: Decreased N/A N/A N/A N/A N/A N/A Small mesenteric arteries: decreased Small mesenteric arteries and aorta: decreased Renal microvessels: decreased Leo et al. (2011); Makino et al. (2000); Wigg et al. (2001); Absi et al. (2013) Ma et al. (2013) N/A Sotnikova et al. (2011a, 2011b) De Vriese et al. (2000) N/A N/A Uterine microvessels: decreased Decreased Gokina et al. (2015) Streptozotocininduced diabetic rats Streptozotocininduced diabetic rats Streptozotocininduced diabetic rats Streptozotocininduced diabetic rats Streptozotocininduced diabetic rats Streptozotocininduced diabetic pregnant rats N/A Table 3 SK channels in GK and OLETF type 2 diabetic rats. Animal Model Tissue and Cell Type SK Protein Expression in Diabetes SK Channel Gene Expression Vascular Function/ EDHF responsiveness SK/IK Channel Recording References T2D GotoKakizaki rats cremaster skeletal muscle resistance arteries Mesenteric artery endothelial cells N/A N/A preserved N/A Mishra et al. (2021a, 2021b) N/A N/A Small mesenteric arteries: decreased Matsumoto et al. (2010); Oniki et al. (2013) Mesenteric artery endothelial cells N/A N/A Small mesenteric arteries: decreased Reduced SK and IK channel responses to stimulation N/A Mesenteric artery endothelial cells N/A N/A Small mesenteric arteries: decreased Reduced endothelial SK/ IK activity LAD coronary arteries N/A N/A LAD coronary arteries: preserved N/A Goto-Kakizaki type 2 diabetic rats Goto-Kakizaki type 2 diabetic rats OLETF type 2 diabetic rats OLETF type 2 diabetic rats experiments report decreased SK gene/protein expression in diabetic rodent mesenteric arteries (Ding et al., 2005; Ma et al., 2013) and cavernous tissue endothelial cells (Zhu et al., 2008). To complicate matters further, other studies report no changes in SK gene/protein Oniki et al. (2006) Matsumoto et al. (2006); Matsumoto et al. (2008); Matsumoto et al. (2009) Kajikuri et al. (2009) expression in diabetic rat mesenteric arteries (Brøndum et al., 2009), diabetic rat coronary arterioles (Zhang et al., 2020; Xing et al., 2021; Song et al., 2020), and human coronary arterioles (Liu et al., 2015, 2018, 2020). 6 S. Kant et al. European Journal of Cell Biology 101 (2022) 151208 Table 4 SK channels in other models of diabetes/metabolic syndrome. Animal Model or Human Tissue Tissue and Cell Type SK Protein Expression in Diabetes SK Channel Gene Expression Vascular Function/ EDHF responsiveness SK/IK Channel Recording References Mouse, genetically modified type 2 DM Heart endothelial cells No change N/A Coronary microvessel: Decreased Decreased Genetically modified type 2 diabetic mice Human, type 2 DM Coronary arterioles N/A N/A N/A Atrial No change No change Pig, coronary endothelial cells cultured in hyperglycemic media Coronary artery endothelial cells N/A N/A Coronary microvessels: Indeterminate Coronary microvessel: Decreased Coronary microvessels: decreased Zhang et al. (2020); Xing et al. (2021); Song et al., 2020 Park et al. (2008) Decreased Liu et al. (2015); Liu et al. (2018); Liu et al. (2020) Tsai et al. (2011) N/A underpin obesity-induced EDH dysfunction. Coronary arteriolar SK/IK channel dysfunction in diabetes is of particular interest for two reasons. First, the coronary endothelium ex­ presses a considerable number of SK channels, and SK channels are crucial mediators of coronary arteriolar relaxation (Grgic et al., 2009; Ledoux et al., 2006). Second, many people with diabetes experience more severe coronary artery disease than non-diabetics, and so targeting coronary SK channels in diabetic patients may provide some therapeutic benefit. Hence a considerable amount of work has been done attempting to understand the molecular basis of SK channel dysfunction in diabetic coronary arteries (Fig. 2); it is possible that similar mechanisms may be at play in other vascular beds as well, although more specific studies will Wide variation across different studies, particularly in animal models, may be a function of differences in the extent of diabetes-related vascular pathology in alternative animal models and the specific pro­ tocols used by individual groups concerning when to test vascular function. Indeed, different studies test for diabetes-induced changes in vascular function at different time points in the development of diabetes in their model system of choice. Thus, it may be that early on in diabetes, SK channel expression increases to compensate for disturbances in vascular function due to growing hyperglycemia; however, this attempted compensation is inadequate since vascular function almost always decreases anyway. As the disease process continues and vascular damage increases, gene/protein expression patterns change, and SK channel expression levels revert to baseline or even fall. Regarding the studies by Liu et al. on the human coronary microcirculation, the lack of changes in SK channel gene/protein expression alongside reduced SK channel currents suggests that the pathophysiology at play in this circumstance is most likely a post-translational or cytosol-to-membrane SK channel protein trafficking issue. Obesity often accompanies diabetes, especially type 2 DM; hence it bears mention in any discussion of diabetes/metabolic syndrome. In diet-induced models of obesity using Sprague-Dawley rats, EDHmediated relaxation of small mesenteric arteries is significantly decreased (Haddock et al., 2011; Gradel et al., 2018) (Table 5). The type of diet used to generate rat models may have an impact on the presence or absence of altered SK/IK channel expression. For example, feeding rats high fat chow alone to induce obesity resulted in upregulated IK channel protein expression despite overall diminished EDH activity (Haddock et al., 2011). In contrast, for rats fed a combination of high-fat chow and high-fructose drinking water to produce obesity, SK and IK channel mRNA expression were both downregulated (Gradel et al., 2018). Meanwhile, unlike with mesenteric arteries, vascular relaxation responses in the saphenous artery of rats fed a cafeteria-style diet for obesity induction were preserved, and IK channel protein expression increased (Chadha et al., 2010) Additional work is required to better characterize phenotypes of obesity and any molecular changes that Fig. 2. Proposed mechanisms for diabetic inactivation of endothelial SK/IK channels in human coronary endothelial cells. The dotted line reflects inhibi­ tion, and the solid black lines reflect enhancement. PKC = Protein Kinase C; NOX = NADPH oxidase. Table 5 SK channels in animal models of obesity. Animal Model Tissue and Cell Type SK Protein Expression in Diabetes SK Channel Gene Expression Vascular Function/ EDHF responsiveness SK/IK Channel Recording References Sprague Dawley rat model, diet-induced obesity Sprague-Dawley rats, high fat and high fructose model of obesity Cafeteria-style diet-induced obese rats Obese Zucker rats Mesenteric artery endothelial cells Mesenteric artery endothelial cells IK: Upregulated SK: No change N/A N/A Small mesenteric arteries: Decreased EDH Small mesenteric arteries: Decreased EDH IK: Increased SK: Decreased N/A Haddock et al. (2011) Gradel et al. (2018) Saphenous artery endothelial cells Coronary artery endothelial cells IK: Increased SK3: No change SK3 and IK: Increased Saphenous artery: preserved Coronary artery endothelial cells: preserved N/A Chadha et al. (2010) Climent et al, 2014 SK and IK channels: Downregulated in high fat and high fructose group N/A N/A 7 N/A S. Kant et al. European Journal of Cell Biology 101 (2022) 151208 be required. First, diabetes has been shown to increase the levels of NADH (nicotinamide adenine dinucleotide hydrogen) and the NADH/NAD+ (nicotinamide adenine dinucleotide) ratio in human coronary artery endothelial cells and the human myocardium (Liu et al., 2020). Increased intracellular NADH resulted in decreased endothelial SK channel currents while increased intracellular NAD+ increased endo­ thelial SK currents (Liu et al., 2020). The basic concept of pyridine nucleotide regulation of ion channels is not new-other studies have shown that high NADH/NAD+ ratios decrease cardiac sodium currents, and high intracellular NADH downregulates BK currents in rabbit pul­ monary artery smooth muscle cells (Liu et al., 2009; Lee et al., 1994). Therefore, given that a hallmark of diabetes is generalized impairment of mitochondrial metabolism and elevated intracellular NADH, it is possible that NADH facilitates diabetic inhibition of coronary SK chan­ nels to produce diminished electrical signaling. Meanwhile, reactive oxygen species (ROS) may also play a role in diabetic SK channel dysfunction. The mitochondria are major sources of enhanced ROS production in diabetes. Oxidative stress is a key mediator of cell injury and, in extreme cases, apoptosis (Kizhakekuttu et al., 2012; Cho et al., 2013). Note that increased ROS production will affect the redox status of key enzymes, such as eNOS and its cofactor tetrahy­ drobiopterin (Tabit et al., 2010). Experiments on diabetic mice have shown that administration of mito-Tempo, an inhibitor of mitochondrial ROS production, significantly improved coronary endothelium-dependent relaxation responses to ADP and NS309, and resulted in increased endothelial SK currents in comparison to diabetic mice that did not receive treatment (Xing et al., 2021, Fig. 3). It is possible that oxidation or nitration of SK channels due to the action of free radicals may impair their calcium sensitivity/overall function; however, further investigation will be necessary to identify specific mechanisms by which mitochondrial ROS may inhibit endothelial SK channels in diabetes. Finally, enhanced activity of protein kinase C may contribute to altered SK/IK channel activity in uncontrolled diabetes. Earlier, we described at length how protein kinase C beta inhibits SK channel activity by reducing channel calcium sensitivity. Hyperglycemia has been shown to increase activation of protein kinase C and diacylglycerol (DAG), with preferential activation of beta and delta isoforms in the diabetic vasculature (Koya and King, 1998). DAG levels rise due to overabundance of the glycolytic intermediate dihydroxyacetone phos­ phate, which is rapidly reduced to glycerol-3-phosphate; this in turn increases de-novo DAG biosynthesis (Xia et al., 1994). Given that DAG is a powerful activator of PKC, increased production of DAG facilitates increased PKC activation. PKC overactivity leads to several harmful effects on endothelial cells. For example, PKC degrades endothelial tight junctions, increasing endothelial permeability to large macromolecules, such as albumin (Geraldes and King, 2010). In addition, PKC alters bioavailability of crucial endothelial-derived vasoactive factors, decreasing activity/pro­ duction of vasodilators like eNOS and prostacyclin while increasing production of vasoconstrictors like thromboxane A-2 and endothelin-1 (Geraldes and King, 2010). Furthermore, studies in bovine aortic endothelial cells suggest that hyperglycemia, through elevated PKC, interferes with gap junction mediated cell-cell communication. This effect is abolished with PKC inhibition (Inoguchi et al., 1995). This could have relevance for SK channel activity modulation if viewed through the lens of the myoendothelial coupling hypothesis of EDHF activity, wherein SK channel hyperpolarization of endothelial cells is transmitted to vascular smooth muscle cells via gap junctions. Reduced gap junction signaling would compromise endothelial SK-channel induced vascular smooth muscle relaxation, accounting for impaired responses observed in diabetes. It is also possible that increased PKC activity simply enhances the ability of PKC to reduce SK channel calcium sensitivity through heightened phosphorylation of SK-channel bound calmodulin. However, these are merely hypotheses; additional studies will be needed to determine any potential mechanisms. One last point with respect to diabetes and PKC-metformin has been shown to inhibit the PKC/extra­ cellular signal-regulated kinase (ERK) pathway in a rat model of type 2 diabetes, and this inhibition was linked to increased SK2 protein expression in atrial tissue (Liu et al., 2018a). Curiously, SK3 expression in metformin-treated diabetic rats decreased. Such findings suggest that if PKC is involved with SK channel synthesis/trafficking, specific effects may differ based on the specific SK channel subtype in question, an important complication that bears consideration for future studies. 3.2. Hypertension and SK channels Beyond diabetes, another major altered-metabolic disease state that may involve SK channel pathology is hypertension. Any discussion of hypertension must begin with a brief survey of the sheer size and scope of the problem at hand. Hypertension is a leading cause of cardiovas­ cular morbidity and mortality around the world, with an estimated global adult prevalence of over 30% (Mills et al., 2020). In the United States alone, in 2018 hypertension was implicated in over half a million deaths as either the primary or an important contributing factor (Centers for Disease Control and Prevention, March, 2020). Moreover, about 37 million adults in the United States have uncontrolled hypertension, defined as blood pressures greater than or equal to 140/90 (Centers for Disease Control and Prevention, Hypertension Prevalence, Treatment, and Control Estimates, 2019). Unsurprisingly, the costs of hypertension to the American healthcare system are staggering, in-excess of $131 billion year on year (Kirkland et al., 2018). Hypertension imposes a wide array of adverse effects on every organ system. These include increased risk of hemorrhagic or ischemic stroke, kidney disease, retinopathy, cardiac valvular disease, ischemic heart disease, and left ventricular dysfunction (Mensah et al., 2002). All these disparate pathologies stem from macrovascular and microvascular damage due to persistent uncontrolled hypertension. At the macro­ vascular level, hypertension can induce arteriolosclerosis in the small arteries and arterioles (Balakrishnan et al., 2015; Kono et al., 2016; Fig. 3. NADH, mROS, and SK Channels in Diabetes. This figure depicts NADHinhibited SK/IK channel activity via PKC and mROS activation in endothelial cells in the context of diabetes. 8 S. Kant et al. European Journal of Cell Biology 101 (2022) 151208 Gavornik and Galbavy, 2001). In benign essential hypertension, arte­ riolosclerosis is hyaline, characterized by simple thickening of vessel walls secondary to leakage of plasma proteins into the vessel media. In severe hypertension, hyperplastic arteriolosclerosis may be seen, char­ acterized by extensive myointimal cell hyperplasia and medial vascular smooth muscle proliferation that resembles an onion skin on histology. Thickening of vessel walls further reduces systemic arterial compliance, exacerbating already high systolic blood pressures. Hence the heart must work increasingly hard to maintain adequate organ perfusion, elevating the risk of left ventricular remodeling and left ventricular failure. At the microvascular level, hypertensive vessels exhibit extensive endothelial dysfunction. For example, hypertension accelerates the progression of atherosclerosis (Schiffrin, 2002; Ning et al., 2018; Alex­ ander, 1995). Mechanical injury to the endothelium due to increasingly turbulent blood flow/shear stress in hypertension may enhance intimal permeability to LDL and may also promote increased entry of monocytes into vessel intimae (Ning et al., 2018). In addition, increased levels of circulating angiotensin II may be found in hypertensive patients, which enhances atherosclerosis and the risk of aortic aneurysms in several animal models (Ning et al., 2018; Weiss et al., 2001). A growing body of evidence suggests that alterations in endothelial regulation of vasomotor tone have a significant role in microvascular changes induced by hypertension. Indeed, the normal endothelial response to increased shear stress involves increased release of vaso­ dilatory factors such as nitric oxide and prostacyclin, along with heightened endothelium-dependent hyperpolarization. Studies have shown that hypertension reduces the bioavailability of endothelial nitric oxide and increases production of endothelium-derived contracting factors, chiefly vasoconstrictive arachidonic acid metabolites such as thromboxane A2 (Goto et al., 2018; Vanhoutte et al., 2005). Dysregu­ lation of SK channels in peripheral vascular and microvascular endo­ thelial dysfunction during hypertension has been extensively studied. SK channels have an important role in regulation and control of blood pressure. For example, one of the most common forms of endo­ crine hypertension is primary hyperaldosteronism, and blockade of SK channels in human adrenocortical cells (e.g., with apamin) stimulates aldosterone secretion (Yang et al., 2016). With respect to endothelial cells, experiments with hypertensive connexin-40 deficient mice have found that stimulation of endothelial SK4/IK channels with SKA-31 reduced blood pressure by over 30 mmHg, providing additional evi­ dence of a role for SK channels in blood pressure modulation (Radtke et al., 2013). Moreover, spontaneously hypertensive mice show impaired responsiveness to the SK/IK channel activator NS309 in mesenteric arteries, along with reduced overall protein expression of SK and inward rectifier potassium channels (Weston et al., 2010). Others have also reported downregulation of SK channel expression in mesenteric arteries of rat models of spontaneously induced hyper­ tension and angiotensin induced hypertension (Seki et al., 2017; Hilgers and Webb, 2007). Alongside SK channel downregulation, Seki et al. report decreased expression of endothelial TRPV4 channels in their mouse model (Seki et al., 2017). This is of interest because calcium influx through TRPV4 channels may be involved in downstream acti­ vation of SK/IK channels (Seki et al., 2017). Hence reduced endothelial SK channel currents in hypertension may be the result of a combination of at least two factors: decreased overall SK channel expression and reduced activation of available SK channels due to TRPV4 downregulation. Approaching animal models of hypertension from a different angle, SK3 and IK1 knockout mice exhibit markedly impaired EDHF-mediated vasodilation and elevated arterial blood pressures (Bra¨hler et al., 2009; Yap et al., 2016a, 2016b; Taylor et al., 2003). Endothelial SK channel deficiency has also been linked with reduced nitric oxide mediated vasodilator responses in transgenic mouse models (Köhler and Ruth, 2010). Moreover, beyond displaying hypertensive phenotypes, IK channel deficient mice also exhibit mild left ventricular hypertrophy, mimicking a well-known myocardial consequence of long-standing uncontrolled hypertension in humans (Si et al., 2006). Curiously, some studies have found increased IK channel expression in mesenteric and renal arteries of spontaneously hypertensive rats alongside decreased SK channel expression, albeit with overall dimin­ ished endothelial cell responsiveness to NS309 (Giachini et al., 2009; Simonet et al., 2012). Meanwhile, a different rodent model of hyper­ tension, Dahl salt-sensitive hypertensive rats (DS-H rats), showed increased EDHF-like relaxation, after treatment with nitric oxide syn­ thase inhibitors (Goto et al., 2012). However, this particular EDHF-like response appears to be driven by BK channel upregulation rather than changes to SK/IK channels, given that iberiotoxin blockade of BK channels attenuated the relaxation response (Goto et al., 2012). Specific molecular mechanisms underpinning differences in SK and IK channel regulation in hypertension remain unelucidated. One possi­ bility may be differences in subcellular localization, with SK channels largely localized to endothelial cell membranes and IK channels largely localized to the myoendothelial projections that link endothelial cells with vascular smooth muscle cells (Goto et al., 2018). It is also important to note that nearly all mouse model studies examining SK channel function in hypertension thus far have been done on the peripheral circulation. Future research will need to examine SK channel changes in other vascular beds, such as the coronary, pulmonary, or cerebral mi­ crocirculations, before drawing conclusions about broad systemic im­ plications of SK channel dysfunction in hypertension. Furthermore, few studies have examined hypertensive human blood vessels to verify the changes in endothelial SK channel activity/expression seen in mouse models (Grgic et al., 2009). If these findings are confirmed, endothelial SK channel modulation, particularly SK3 and IK stimulation, may pre­ sent a novel therapeutic approach to improve endothelium-dependent vasodilation and lower blood pressure in individuals with hypertension. 3.3. Hypoxia-reperfusion and SK channels The final pathological condition featured in this review is hypoxia/ ischemia-reperfusion injury, another prototypical example of an altered metabolic disease state in which SK channel dysfunction may play a crucial role. All organ systems rely on aerobic metabolism as a main source of cellular energy production; hence maintenance of oxygen homeostasis is crucial for survival. While some organs, like skeletal muscle, are more resistant to fluctuations in oxygen levels, other organs, most notably the heart and the brain, are exquisitely vulnerable to ox­ ygen perturbations and cease to function normally within minutes of oxygen deprivation. Ischemia may also occur in medical settings such as cardiac surgery involving cardioplegia/cardiopulmonary bypass (CP/ CPB), where the heart is temporarily stopped, and a heart-lung machine takes over the role of maintaining the body’s circulation. Despite ad­ vances in cardioprotective strategies over the years, ischemiareperfusion injury still often occurs during/after CP/CPB, hallmarks of which include reduced coronary endothelial function and impaired relaxation that result in coronary vasospasm and myocardial malper­ fusion (Feng and Sellke, 2016; Robich et al., 2010). A vast array of deleterious consequences result from ischemia (Yang et al, 2016; Crossman, 2004; Chen et al., 2020). First, oxidative phos­ phorylation ceases, triggering a shift to anaerobic glycolysis that ele­ vates intracellular lactate levels. Increased lactic acid lowers intracellular pH, leading to suboptimal enzyme function and clumping of nuclear chromatin. Ultimately, anaerobic glycolysis cannot generate enough ATP to fuel metabolic needs. For example, an immediate consequence of low ATP is decreased activity of the cell membrane Na-K ATPase. Decreased Na-K ATPase activity leads to increased sodium influx, potassium efflux, and altered cellular resting membrane poten­ tials. Over time, disrupted membrane potential leads to calcium influx. Rising levels of intracellular calcium activate enzymes such as phos­ pholipases, proteases, and nucleases that degrade plasma membrane phospholipids, cytoskeletal proteins, and nuclear DNA. Calcium can also trigger destruction of mitochondrial membranes (via phospholipase A2) 9 S. Kant et al. European Journal of Cell Biology 101 (2022) 151208 and opening of mitochondrial permeability transition pores (MPTPs) that allow leakage of mitochondrial enzymes, like the pro-apoptotic cytochrome c, into the cytosol. In the case of hypoxia, where cellular oxygen levels are low but NOT zero, these effects occur more gradually over a longer period. The main treatments for tissue hypoxia/ischemia depend on the cause. If the problem is poor blood flow to a specific location, targeted reperfusion is the primary approach. If the problem is hypoxemia, then pulmonary interventions (e.g., supplemental oxygen, mechanical ventilation) may be necessary. Unfortunately, reperfusion is not without some adverse effects (Slegtenhorst et al., 2014; Cowled and Fitridge, 2011). First, rapid restoration of blood to oxygen-starved tissue can lead to oxygen overload that itself generates reactive oxygen species (ROS), while the antioxidant scavenging capacity of ischemic cells is insuffi­ cient to compensate. Unchecked ROS can damage already injured mitochondria in reperfused tissue, increasing MPTP formation and release of mitochondrial contents into the cytosol, causing cell swelling and death (Slegtenhorst et al., 2014). Meanwhile, ROS can stimulate inflammation by promoting formation of inflammasomes and activation of pro-inflammatory cytokines such as IL-1 beta (Gross et al., 2011; Tschopp and Schroder, 2010). Finally, reperfusion may also lead to calcium overload that can facilitate cellular apoptosis. Endothelial dysfunction, most often manifesting as increased endo­ thelial activation and altered vasomotor tone, is an important aspect of pathology in hypoxia/ischemia-reperfusion due to disease or CP/CPB. For example, human umbilical vein endothelial cells exhibit increased ICAM-1 expression and enhanced monocyte adhesion following hypoxia (Liang et al., 2019). Hypoxia also contributes to depletion of BH4 and L-arginine, the former being an important cofactor of and the latter being a key substrate for endothelial nitric oxide synthase (eNOS) (Janaszak-Jasiecka et al., 2021). This results in two major consequences. First, eNOS stops generating normal levels of nitric oxide, leading to impaired vascular smooth muscle relaxation. Second, through depletion of BH4 and L-arginine, hypoxia elicits eNOS uncoupling as eNOS begins to produce superoxide anions instead of nitric oxide, which contributes to endothelial cell oxidative stress. In addition, ischemia-reperfusion has been shown to increase vascular sensitivity to the endothelial-derived vasoconstrictor endothelin-1 in dog coronary arteries and the coronary microvascula­ ture (Miura, 1996). Experiments in human coronary arterioles following cardioplegic ischemia-reperfusion during CP/CPB show reduced responsiveness to the endothelial-derived vasoconstrictor thromboxane A2 (Feng et al., 2011). Curiously, human coronary arterioles exhibit reduced contractile responses to endothelin-1 following CP/CPB, con­ trary to results seen in some animal models of ischemia-reperfusion (e. g., Miura, 1996) (Feng et al., 2010). Altered SK channel-EDHF activity gives rise to endothelial dysfunc­ tion following hypoxia/ischemia-reperfusion. At first, early animal models of ischemia-reperfusion suggested that EDHF responses were heightened after ischemia-reperfusion. For instance, rat middle cerebral arteries showed increased EDHF-mediated vasodilation following ischemia-reperfusion (Marrelli et al., 2001). Likewise, coronary arteri­ oles of dogs subjected to transient occlusion and reperfusion of the left circumflex coronary artery showed increased EDHF responses (Chan and Woodman, 1999). However, more recent studies paint a different picture. Human cor­ onary arterioles harvested from atrial tissue after CP/CPB display markedly reduced relaxation responses to the SK/IK channel activator NS309 compared with coronary arterioles harvested from atrial tissue before CP/CPB (Feng et al., 2008). Decline in SK channel activity is even more pronounced in coronary arterioles of diabetic patients following CP/CPB (Liu et al., 2018). In addition, mouse small coronary artery endothelial cells exposed to CP hypoxia-reoxygenation exhibit signifi­ cantly attenuated endothelial SK channel activity, along with intracel­ lular calcium overload that strongly correlated with decreased SK channel currents (Song et al., 2021). Pretreatment of mouse and human coronary artery endothelial cells with NS309 before CP hypoxia-oxygenation conferred significant protection of coronary endothelial function after hypoxia-reperfusion compared to control groups that received no pre-treatment (Zhang et al., 2020). In the specific context of CP, the type of cardioplegic solution used during hypoxia-reperfusion may impact the extent of EDHF/SK channel dysfunction. Although depolarizing (hyperkalemic) cardioplegia is currently the most common method for myocardial preservation during cardiac surgery, hyperpolarizing cardioplegia (e.g., through use of ATPsensitive potassium channel activators) has been shown to better pre­ serve EDHF-mediated vascular relaxation (He et al., 2004; He and Yang, 1997). Indeed, coronary microvascular responses to the EDHF stimu­ lator bradykinin are markedly attenuated during hyperkalemic car­ dioplegia, while hyperpolarizing cardioplegia results in less significant attenuation (He et al., 2004; He and Yang, 1997). Other models of hypoxia that do not involve CP/CPB provide similar results of blunted EDHF activity. For example, porcine coronary artery endothelial relaxation responses to SK/IK channel activators bradykinin or 1-ethyl-2-benzimidazolinone are decreased following in-vitro expo­ sure to hypoxia (Yang et al., 2013; Dong et al., 2005; Ziberna et al., 2009; Wang et al., 2017). Furthermore, rat models of chronic hypoxia-induced pulmonary hypertension show diminished endothelial-derived hyperpolarization in small pulmonary arteries (Kroigaard et al., 2013). Note also that experiments on the pulmonary vasculature in isolated rat lungs have shown that inhibition of SK channel activity with substances such as apamin strengthens pulmonary vasoconstriction in response to hypoxia (Dumas et al., 1999). Potential causes of altered SK/IK channel activity following hypoxia/ ischemia-reperfusion remain somewhat of a mystery, although several theories abound. One study by Wang et al. observed pretreatment of pig coronary artery endothelial cells with the TRPC3 channel activator 1oleoyl-2-acetyl-sn-glycerol (OAG) restores SK channel currents and EDHF vasodilation after hypoxia-reperfusion (Wang et al., 2017). Given that hypoxia inhibits TRPC3-mediated cellular calcium entry and TRPC3 trafficking to the endothelial cell surface, it is possible that dysregulated TRPC3 trafficking may be implicated in hypoxic SK/IK channel dysfunction (Huang et al., 2011). There is debate in the literature as to whether endothelial SK/IK channel levels decrease following hypoxia/ischemia-reperfusion, with some studies suggesting no (e.g., Feng et al., 2008; Zhang et al., 2020) and others suggesting yes (more specifically, decreased SK4/IK channel expression as reported by Yang et al., 2013; Kroigaard et al., 2013). Curiously, Kroigaard et al. observed upregulated SK3 channel protein in small pulmonary arteries in their chronically hypoxic rat models of pulmonary hypertension; nonetheless, EDH-type relaxation was overall reduced (Kroigaard et al., 2013). A decrease in overall SK/IK channel levels following hypoxia-reperfusion would directly explain the reduced currents. However, if overall SK/IK channel levels do not change, then the molecular mechanisms responsible for observed SK/IK channel dysfunction must be post-translational (Feng et al., 2008; Zhang et al., 2020). Potential possibilities in this scenario include (1) ischemia-reperfusion induced activation of protein kinases that inhibit SK/IK channel activation and open probability, (2) free radical (ROS) interference with SK channel activity, or (3) alterations in SK/IK channel trafficking from cytosol to cell membrane. Fig. 4 illustrates how several of these proposed mechanisms may come together to result in altered SK/IK channel activity in the context of cardioplegic hypoxia/reperfusion. Evidence exists that supports each of these hypotheses. First, increased protein kinase C (PKC) levels have been observed in myocardial tissue following CP/CPB (Sodha et al., 2008). As discussed earlier, PKC inhibits SK channel activity by phosphorylating SK channel-bound calmodulin, decreasing its responsiveness to calcium and reducing overall channel calcium sensitivity. However, the main isoform of PKC implicated in SK channel regulation is the beta isoform, and this study only noted upregulation of delta and epsilon isoforms (Sodha 10 S. Kant et al. European Journal of Cell Biology 101 (2022) 151208 calcium-mediated SK channel activity) as opposed to aggregate protein levels. Higher cytosolic vs cell membrane SK3 expression has been found following CP hypoxia-reperfusion in human coronary artery endothelial cells and mouse heart endothelial cells; SK4 channels, however, display no alterations in subcellular distribution (Zhang et al., 2020). Hence CP-hypoxia/reperfusion may induce SK3 channel internalization, which would explain diminished overall endothelial responses to SK activators like NS309. However, few studies exist replicating these findings, and details surrounding the processes by which SK3 internalization occurs remain unclear. Additional work is needed to elucidate the specific molecular processes at play. 3.4. Final thoughts: clinical and translational perspectives and questions Strong evidence of a role for SK channel dysfunction in the patho­ genesis of microvascular endothelial dysfunction in the alteredmetabolic disease states discussed in this review raises several impli­ cations for clinical and translational applications. Many such applica­ tions were mentioned in the preceding sections on diabetes, hypertension, and ischemia-reperfusion; here, the major highlights are summarized and emphasized. Beginning with diabetes, work done by Liu et al. demonstrating decreased SK currents in human diabetic cor­ onary microvessels, and Xing et al., which showed that the mROS in­ hibitor mito-Tempo improved coronary endothelium-dependent relaxation responses and endothelial SK currents in diabetic mice, was discussed at length Liu et al., 2018, 2020, 2015; Xing et al., 2021). This lends credence to the possibility that mROS inhibition may augment SK channel function in diabetic humans, who also exhibit high levels of cellular oxidative stress, thereby serving as a therapeutic tool for pro­ tecting coronary microvascular function. Animal models of hypertension and ischemia-reperfusion also display diminished SK channel currents, and treatment with SK channel activators such as NS309 or mROS inhibitors like mito-Tempo appears to improve/protect endothelial function in these circumstances (Song et al., 2021; Zhang et al., 2020; Seki et al., 2017; Goto et al., 2018). Hence there is potential for pharmacologic stimulation of SK channels to also be useful in treating disordered vasomotor tone in hypertensive vascular disease, and in protecting the microcirculation from ischemia-reperfusion damage during situations such as cardiopulmo­ nary bypass. However, although many experiments have been done using coronary microvascular tissue harvested from human atria, there remains a general dearth of research investigating the impact of phar­ macologic modulation of SK channels in human tissue more broadly (e. g. other non-coronary human microcirculations). Even with respect to animal models of diabetes, hypertension, and ischemia-reperfusion, nearly all major studies of SK channel modulation have been done on mesenteric and coronary microvessels. Future research will need to address the impact of SK channel dysfunction in other vascular beds, such as the pulmonary or renal mi­ crocirculations, in humans and animal models of diabetes/ hypertension/ischemia-reperfusion. Moreover, even though there is relative consensus concerning diminished SK channel activity in these disease states, it remains hotly contested whether SK channel expression levels themselves change and if so, how; this is another area ripe for future investigations. Finally, it will be important to eventually consider how SK channel modulators could be delivered to patients in a clinical setting. Indeed, most major experiments so far have utilized infusions of SK channel activators in living mouse models or application of SK channel activators in-vitro to isolated microvessels. Many questions remain regarding potential side effects of systemic SK channel activator infusion beyond the microcirculation (as SK channels are not only pre­ sent on endothelial cells) that may complicate clinical use. In addition, it may be worthwhile to investigate whether SK channel activators can be delivered via oral formulations (e.g. a pill), and compare the pharma­ codynamic and pharmacokinetic profile with IV and IM routes of Fig. 4. SK channel dysfunction during cardioplegic hypoxia/reoxygenation (CP-H/R). Increased NOX or NADH during CP-H/R activates PKC and induces mROS production, resulting in decreased SK channel activity, coronary endo­ thelial dysfunction, and reduced coronary arteriolar relaxation. et al., 2008). PKC beta inhibition has been demonstrated to improve SK channel currents in other disease processes (e.g., diabetes, discussed earlier). Additional studies are required to examine whether PKC beta expression or activity levels are altered in endothelial cells during hypoxia-reperfusion. Next, endogenous antioxidants such as glutathione peroxidase, su­ peroxide dismutase, and catalase have all been shown to attenuate myocardial injury caused by ischemia-reperfusion (Dhalla, 2000). Moreover, altered mitochondrial membrane potentials, in particular mitochondrial membrane hyperpolarization, have been shown to in­ crease mitochondrial ROS production in the setting of endothelial injury (Kluge et al., 2013). This is thought to be due in part to an increase in the NADH/NAD+ ratio and a decrease in electron flow rates through com­ plex I and complex III of the mitochondrial electron transport chain, which prolong the lifespan of reactive intermediates and promote su­ peroxide anion formation from oxygen reduction (Kluge et al., 2013). With respect to ischemia-reperfusion, inhibition of mitochondrial reactive oxygen species using the antioxidant Mito-Tempo enhances relaxation responses in mouse coronary artery endothelial cells following CP/CPB, compared to untreated control groups (Song et al., 2021). Mito-Tempo also reduced the magnitude of calcium overload, providing a potential mechanistic link between reduced mitochondrial ROS production and improved SK channel activity (Song et al., 2021). Here, it is important to recall that while calcium is required to activate SK channels, it only does so at certain levels—the theme, as often is the case in biological systems, is balance. Indeed, Song et al. demonstrate that free calcium concentrations of around 400 nM evoke strong in-vitro SK currents in their mouse heart endothelial cells. However, when free calcium concentrations rise to 2uM, SK channel currents drop to levels akin to those under 0 calcium conditions, suggesting that excess calcium is detrimental to SK channel activity. Overall, Mito-Tempo treatment resulted in stronger SK channel currents following CP hypoxia-reperfusion, albeit without any changes in SK channel protein expression pre/post CP, supporting the notion that mitochondrial ROS may affect SK channel activity (e.g., through tighter regulation of 11 European Journal of Cell Biology 101 (2022) 151208 S. Kant et al. administration. pharmacology and physiology 35 (4), 494–497. https://doi.org/10.1111/j.14401681.2008.04903.x. Bertuccio, C.A., Wang, T.T., Hamilton, K.L., Rodriguez-Gil, D.J., Condliffe, S.B., Devor, D. C., 2018. Plasma membrane insertion of KCA2.3 (SK3) is dependent upon the SNARE proteins, syntaxin-4 and snap23. PLoS One 13 (5). https://doi.org/10.1371/journal. pone.0196717. Bildl, W., Strassmaier, T., Thurm, H., Andersen, J., Eble, S., Oliver, D., Knipper, M., Mann, M., Schulte, U., Adelman, J.P., Fakler, B., 2004. Protein kinase ck2 is coassembled with SMALL CONDUCTANCE Ca2+-activated K+ channels and regulates CHANNEL GATING. Neuron 43 (6), 847–858. https://doi.org/10.1016/j. neuron.2004.08.033. Biorender. BioRender. (n.d.). Retrieved November 24, 2021, from 〈https://biorender. com/〉. Bra¨hler, Sebastian, Kaistha, A., Schmidt, V.J., Wo¨lfle Stephanie, E., Busch, C., Kaistha, B. P., Kacik, M., Hasenau, A.-L., Grgic, I., Si, H., Bond, C.T., Adelman, J.P., Wulff, H., de Wit, C., Hoyer, J., Ko¨hler, Ralf, 2009. Genetic deficit of SK3 and IK1 channels disrupts the endothelium-derived hyperpolarizing factor vasodilator pathway and causes hypertension. Circulation 119 (17), 2323–2332. https://doi.org/10.1161/ circulationaha.108.846634. Brøndum, E., Kold-Petersen, H., Simonsen, U., Aalkjaer, C., 2009. NS309 restores EDHFtype relaxation in mesenteric small arteries from type 2 diabetic ZDF rats. Br. J. Pharmacol. 159 (1), 154–165. https://doi.org/10.1111/j.1476-5381.2009.00525.x. Bryan, R.M., You, J., Golding, E.M., Marrelli, S.P., 2005. Endothelium-derived hyperpolarizing factor. Anesthesiology 102 (6), 1261–1277. https://doi.org/ 10.1097/00000542-200506000-00028. Burnham, M.P., Bychkov, R., Félétou, M., Richards, G.R., Vanhoutte, P.M., Weston, A.H., Edwards, G., 2002. Characterization of an apamin-sensitive small-conductance ca2 +-activated K+ channel in porcine coronary artery endothelium: relevance to Edhf. Br. J. Pharmacol. 135 (5), 1133–1143. https://doi.org/10.1038/sj.bjp.0704551. Burnham, M.P., Johnson, I.T., Weston, A.H., 2006. Impaired small-conductance ca2+activated K+ channel-dependent EDHF responses in type II diabetic ZDF rats. Br. J. Pharmacol. 148 (4), 434–441. https://doi.org/10.1038/sj.bjp.0706748. Busse, R., Edwards, G., Félétou, M., Fleming, I., Vanhoutte, P.M., Weston, A.H., 2002. Edhf: bringing the concepts together. Trends Pharmacol. Sci. 23 (8), 374–380. https://doi.org/10.1016/s0165-6147(02)02050-3. Bychkov, R., Burnham, M.P., Richards, G.R., Edwards, G., Weston, A.H., Félétou, M., Vanhoutte, P.M., 2002. Characterization of a charybdotoxin-sensitive intermediate conductance ca2+-activated K+ channel in porcine coronary endothelium: relevance to EDHF. Br. J. Pharmacol. 137 (8), 1346–1354. https://doi.org/10.1038/ sj.bjp.0705057. Centers for Disease Control and Prevention (CDC). Hypertension Cascade: Hypertension Prevalence, Treatment and Control Estimates Among US Adults Aged 18 Years and Older Applying the Criteria From the American College of Cardiology and American Heart Association’s 2017 Hypertension Guideline—NHANES 2013–2016external icon. Atlanta, GA: US Department of Health and Human Services; 2019. Centers for Disease Control and Prevention. Underlying Cause of Death, 1999–2018. CDC WONDER Online Database. Atlanta, GA: Centers for Disease Control and Prevention; 2018. http://wonder.cdc.gov/ucd-icd10.html. Accessed March 12, 2020. Chadha, P.S., Haddock, R.E., Howitt, L., Morris, M.J., Murphy, T.V., Grayson, T.H., Sandow, S.L., 2010. Obesity up-regulates intermediate conductance calciumactivated potassium channels and myoendothelial gap junctions to maintain endothelial vasodilator function. J. Pharm. Exp. Ther. 335, 284–293. Chan, E.C., Woodman, O.L., 1999. Enhanced role for the opening of potassium channels in relaxant responses to acetylcholine after myocardial ischaemia and reperfusion in dog coronary arteries. Br. J. Pharmacol. 126 (4), 925–932. https://doi.org/10.1038/ sj.bjp.0702376. Chapouly, C., Yao, Q., Vandierdonck, S., Larrieu-Lahargue, F., Mariani, J.N., Gadeau, A.P., Renault, M.-A., 2015. Impaired hedgehog signalling-induced endothelial dysfunction is sufficient to induce neuropathy: Implication in diabetes. Cardiovasc. Res. 109 (2), 217–227. https://doi.org/10.1093/cvr/cvv263. Chen, K., Pittman, R.N., Popel, A.S., 2008. Nitric oxide in the vasculature: where does it come from and where does it go? A quantitative perspective. Antioxid. Redox Signal. 10 (7), 1185–1198. https://doi.org/10.1089/ars.2007.1959. Chen, P.-S., Chiu, W.-T., Hsu, P.-L., Lin, S.-C., Peng, I.-C., Wang, C.-Y., Tsai, S.-J., 2020. Pathophysiological implications of hypoxia in human diseases. J. Biomed. Sci. 27 (1) https://doi.org/10.1186/s12929-020-00658-7. Cho, Y.-E., Basu, A., Dai, A., Heldak, M., Makino, A., 2013. Coronary endothelial dysfunction and mitochondrial reactive oxygen species in type 2 diabetic mice. Am. J. Physiol.-Cell Physiol. 305 (10) https://doi.org/10.1152/ajpcell.00234.2013. Climent, B., Moreno, L., Martínez, P., Contreras, C., Sánchez, A., Pérez-Vizcaíno, F., García-Sacristán, A., Rivera, L., Prieto, D., 2014. Upregulation of SK3 and IK1 channels contributes to the enhanced endothelial calcium signaling and the preserved coronary relaxation in obese Zucker Rats. PLoS One 9 (10). https://doi. org/10.1371/journal.pone.0109432. Cohen, R.A., Weisbrod, R.M., Gericke, M., Yaghoubi, M., Bierl, C., Bolotina, V.M., 1999. Mechanism of nitric oxide–induced vasodilatation. Circ. Res. 84 (2), 210–219. https://doi.org/10.1161/01.res.84.2.210. Comerma-Steffensen, S.G., Carvacho, I., Hedegaard, E.R., Simonsen, U., 2017. Small and intermediate calcium-activated potassium channel openers improve rat endothelial and erectile function. Front. Pharmacol. 8. https://doi.org/10.3389/ fphar.2017.00660. Cotter, M.A., Gibson, T.M., Nangle, M.R., Cameron, N.E., 2010. Effects of interleukin-6 treatment on neurovascular function, nerve perfusion and vascular endothelium in diabetic rats. Diabetes Obes. Metab. 12 (8), 689–699. https://doi.org/10.1111/ j.1463-1326.2010.01221.x. 4. Conclusion SK channel activity is instrumental in endothelial function and ho­ meostasis, particularly endothelium-dependent hyperpolarization. Activation of SK channels and subsequent potassium efflux is an inde­ pendent mediator of vascular smooth muscle relaxation in the absence of nitric oxide, prostacyclin, and endothelium-independent vasodilators. A variety of factors regulate SK channel activity under normal physio­ logic circumstances, including but not limited to calcium itself, cellular NADH/NAD+ ratios, and certain isoforms of protein kinase C. Conversely, SK channel dysfunction/dysregulation has been implicated as an important driver of vascular pathology in a variety of altered metabolic disease states, including diabetes mellitus/metabolic syn­ drome, hypertension, and hypoxia/ischemia-reperfusion. In each of these conditions, SK channel activity decreases, disrupting the balance between vasodilation and vasoconstriction and increasing the risk of generalized endothelial dysfunction. Increased protein kinase C activity, increased NADH/NAD+ ratios, increased ROS production, and altered SK channel localization all appear to promote SK channel pathology, suggesting that unifying themes of pathophysiology may exist across different disease states. Future studies will need to expand on the work done in animal models by exploring different vascular beds and, whenever possible, human tissue to verify the principles explored in this review. Ultimately, improved understanding of the role of SK channels in normal physiology and pathophysiology may lead to novel targeted treatments aimed at restoring endothelial function. Data availability No data was used for the research described in the article. Acknowledgments None. Funding This research project was mainly supported by the National Heart Lung and Blood Institute (NHLBI) of 1R01HL127072-01A1, 1R01 HL136347-01, and 3R01HL136347-04S1; and National Institute of General Medical Science (NIGMS) of 5P20-GM103652 (Pilot Project) to J.F. References Absi, M., Oso, H., Khattab, M., 2013. The effect of streptozotocin-induced diabetes on the EDHF-type relaxation and cardiac function in rats. J. Adv. Res. 4 (4), 375–383. https://doi.org/10.1016/j.jare.2012.07.005. Adelman, J.P., Maylie, J., Sah, P., 2012. Small-conductance ca2+-activated k+channels: form and function. Annu. Rev. Physiol. 74 (1), 245–269. https://doi.org/10.1146/ annurev-physiol-020911-153336. Alexander, R.W., 1995. Hypertension and the pathogenesis of Atherosclerosis. Hypertension 25 (2), 155–161. https://doi.org/10.1161/01.hyp.25.2.155. Allen, D., Fakler, B., Maylie, J., Adelman, J.P., 2007. Organization and regulation of SMALL CONDUCTANCE Ca2+-activated K+ Channel MULTIPROTEIN COMPLEXES. J. Neurosci. 27 (9), 2369–2376. https://doi.org/10.1523/jneurosci.3565-06.2007. Balakrishnan, M., Garcia-Tsao, G., Deng, Y., Ciarleglio, M., Jain, D., 2015. Hepatic arteriolosclerosis. Am. J. Surg. Pathol. 39 (7), 1000–1009. https://doi.org/10.1097/ pas.0000000000000419. Balut, C.M., Hamilton, K.L., Devor, D.C., 2012. Trafficking of intermediate (kca3.1) and small (kca2.x) conductance, ca2+-activated k+channels: a novel target for medicinal chemistry efforts? ChemMedChem 7 (10), 1741–1755. https://doi.org/10.1002/ cmdc.201200226. Bauersachs, Johann, Popp, Ru diger, Hecker, Markus, Sauer, Edith, Fleming, Ingrid, Busse, Rudi, 1996. Nitric Oxide Attenuates the Release of Endothelium-Derived Hyperpolarizing Factor. Circulation 94, 3341–3347. https://doi.org/10.1161/01. CIR.94.12.3341. Bellien, Jeremy, Thuillez, Christian, Joannides, Robinson, 2008. Role of endothelium derived hyperpolarizing factor in the upregulation of radial artery basal diameter and endothelium-dependent dilatation in vivo. Clinical and expeirmental 12 European Journal of Cell Biology 101 (2022) 151208 S. Kant et al. Cowled P., Fitridge R. Pathophysiology of Reperfusion Injury. In: Fitridge R., Thompson M., editors. Mechanisms of Vascular Disease: A Reference Book for Vascular Specialists [Internet]. Adelaide (AU): University of Adelaide Press; 2011. 18. Available from: https://www.ncbi.nlm.nih.gov/books/NBK534267/. Crossman, D.C., 2004. The pathophysiology of myocardial ischaemia. Heart 90 (5), 576–580. https://doi.org/10.1136/hrt.2003.029017. Csanyi, G., Lepran, I., Flesch, T., Telegdy, G., Szabo, G., Mezei, Z., 2007. Lack of endothelium-derived hyperpolarizing factor (EDHF) up-regulation in endothelial dysfunction in aorta in diabetic rats. Pharmacol. Rep. 59 (4), 447–455. Dalsgaard, T., Kroigaard, C., Bek, T., Simonsen, U., 2009. Role of calcium-activated potassium channels with small conductance in bradykinin-induced vasodilation of porcine retinal arterioles. Investig. Opthalmol. Vis. Sci. 50 (8), 3819. https://doi. org/10.1167/iovs.08-3168. Dalsgaard, T., Kroigaard, C., Misfeldt, M., Bek, T., Simonsen, U., 2010. Openers of small conductance calcium-activated potassium channels selectively enhance no-mediated bradykinin vasodilatation in porcine retinal arterioles. Br. J. Pharmacol. 160 (6), 1496–1508. https://doi.org/10.1111/j.1476-5381.2010.00803.x. Davignon, J., Ganz, P., 2004. Role of endothelial dysfunction in atherosclerosis. Circulation 109 (23_suppl_1). https://doi.org/10.1161/01.cir.0000131515.03336. f8. De Vriese, A.S., Van De Voorde, J., Blom, H.J., Vanhoutte, P.M., Verbeke, M., Lameire, N. H., 2000. The impaired renal vasodilator response attributed to endothelium-derived hyperpolarizing factor in streptozotocin - Induced diabetic rats is restored by 5methyltetrahydrofolate. Diabetologia 43, 1116–1125. Dhalla, N., 2000. Status of myocardial antioxidants in ischemia–reperfusion injury. Cardiovasc. Res. 47 (3), 446–456. https://doi.org/10.1016/s0008-6363(00)00078x. Ding, H., Hashem, M., Wiehler, W.B., Lau, W., Martin, J., Reid, J., Triggle, C., 2005. Endothelial dysfunction in the streptozotocin-induced diabetic apoE-deficient mouse. Br. J. Pharmacol. 146 (8), 1110–1118. https://doi.org/10.1038/sj. bjp.0706417. Dong, Y.-Y., Wu, M., Yim, A.P.C., He, G.-W., 2005. Hypoxia-reoxygenation, St. Thomas Cardioplegic solution, and Nicorandil on endothelium-derived hyperpolarizing factor in coronary microarteries. Ann. Thorac. Surg. 80 (5), 1803–1811. https://doi. org/10.1016/j.athoracsur.2005.04.047. Dopico, A.M., Bukiya, A.N., Jaggar, J.H., 2018. Calcium- and voltage-gated BK channels in vascular smooth muscle. Pflu¨g. Arch. - Eur. J. Physiol. 470 (9), 1271–1289. https://doi.org/10.1007/s00424-018-2151-y. Dora, K.A., Gallagher, N.T., McNeish, A., Garland, C.J., 2008. Modulation of Endothelial Cell K ca 3.1 channels during endothelium-derived hyperpolarizing factor signaling in mesenteric resistance arteries. Circ. Res. 102 (10), 1247–1255. https://doi.org/ 10.1161/circresaha.108.172379. Dumas, J.-P., Goirand, F., Bardou, M., Dumas, M., Rochette, L., Advenier, C., Giudicelli, J.-F., 1999. Role of potassium channels and nitric oxide in the relaxant effects elicited by β-adrenoceptor agonists on hypoxic vasoconstriction in the isolated perfused lung of the rat. Br. J. Pharmacol. 127 (2), 421–428. https://doi. org/10.1038/sj.bjp.0702575. Eichler, I., Wibawa, J., Grgic, I., Knorr, A., Brakemeier, S., Pries, A.R., Hoyer, J., Köhler, R., 2003. Selective blockade of endothelial ca2+-activated small- and intermediate-conductance K+-channels suppresses EDHF-mediated vasodilation. Br. J. Pharmacol. 138 (4), 594–601. https://doi.org/10.1038/sj.bjp.0705075. Félétou, Michel, Vanhoutte, P.M., 2006. Endothelium-derived hyperpolarizing factor. Arterioscler. Thromb. Vasc. Biol. 26 (6), 1215–1225. https://doi.org/10.1161/01. atv.0000217611.81085.c5. Félétou, M., 2009. Calcium-activated potassium channels and endothelial dysfunction: therapeutic options? Br. J. Pharmacol. 156 (4), 545–562. https://doi.org/10.1111/ j.1476-5381.2009.00052.x. Félétou, M., Vanhoutte, P.M., 2007. Endothelium-dependent hyperpolarizations: past beliefs and present facts. Ann. Med. 39 (7), 495–516. https://doi.org/10.1080/ 07853890701491000. Feng, J., Sellke, F., 2016. Microvascular dysfunction in patients with diabetes after cardioplegic arrest and cardiopulmonary bypass. Curr. Opin. Cardiol. 31 (6), 618–624. https://doi.org/10.1097/hco.0000000000000340. Feng, J., Liu, Y., Chu, L.M., Clements, R.T., Khabbaz, K.R., Robich, M.P., Bianchi, C., Sellke, F.W., 2011. Thromboxane-induced contractile response of human coronary arterioles is diminished after cardioplegic arrest. Ann. Thorac. Surg. 92 (3), 829–836. https://doi.org/10.1016/j.athoracsur.2011.04.049. Feng, J., Liu, Y., Clements, R.T., Sodha, N.R., Khabbaz, K.R., Senthilnathan, V., Nishimura, K.K., Alper, S.L., Sellke, F.W., 2008. Calcium-activated potassium channels contribute to human coronary microvascular dysfunction after cardioplegic arrest. Circulation 118 (14_suppl_1). https://doi.org/10.1161/ circulationaha.107.755827. Feng, J., Liu, Y., Khabbaz, K.R., Hagberg, R., Sodha, N.R., Osipov, R.M., Sellke, F.W., 2010. Endothelin-1-induced contractile responses of human coronary arterioles via endothelin-A receptors and PKC-α signaling pathways. Surgery 147 (6), 798–804. https://doi.org/10.1016/j.surg.2009.11.016. Festa, A., D’Agostino, R., Howard, G., Mykka¨nen, Leena, Tracy, R.P., Haffner, S.M., 2000. Chronic subclinical inflammation as part of the insulin resistance syndrome. Circulation 102 (1), 42–47. https://doi.org/10.1161/01.cir.102.1.42. Furchgott, R.F., Zawadzki, J.V., 1980. The obligatory role of endothelial cells in the relaxation of arterial smooth muscle by acetylcholine. Nature 288 (5789), 373–376. https://doi.org/10.1038/288373a0. Garland, C.J., Hiley, C.R., Dora, K.A., 2011. EDHF: Spreading the influence of the endothelium. Br. J. Pharmacol. 164 (3), 839–852. https://doi.org/10.1111/j.14765381.2010.01148.x. Gavornik, P., Galbavy, S., 2001. Clinical picture of arteriolosclerosis. Bratisl. Lek. Listy 102 (7), 326–331. Geraldes, P., King, G.L., 2010. Activation of protein kinase C isoforms and its impact on diabetic complications. Circ. Res. 106 (8), 1319–1331. https://doi.org/10.1161/ circresaha.110.217117. Gimbrone, M.A., García-Cardeña, G., 2016. Endothelial cell dysfunction and the pathobiology of atherosclerosis. Circ. Res. 118 (4), 620–636. https://doi.org/ 10.1161/circresaha.115.306301. Gokina, N.I., Bonev, A.D., Phillips, J., Gokin, A.P., Veilleux, K., Oppenheimer, K., Goloman, G., 2015. Impairment of IK Ca channels contributes to uteroplacental endothelial dysfunction in rat diabetic pregnancy. Am. J. Physiol. Circ. Physiol. 309, H592–H604. Goto, K., Kansui, Y., Oniki, H., Ohtsubo, T., Matsumura, K., Kitazono, T., 2012. Upregulation of endothelium-derived hyperpolarizing factor compensates for the loss of nitric oxide in mesenteric arteries of Dahl salt-sensitive hypertensive rats. Hypertens. Res. 35 (8), 849–854. https://doi.org/10.1038/hr.2012.36. Goto, K., Ohtsubo, T., Kitazono, T., 2018. Endothelium-dependent hyperpolarization (EDH) in hypertension: the role of endothelial ion channels. Int. J. Mol. Sci. 19 (1), 315. https://doi.org/10.3390/ijms19010315. Gradel, A.K.J., Salomonsson, M., Sørensen, C.M., Holstein-Rathlou, N.-H., Jensen, L.J., 2018. Long-term diet-induced hypertension in rats is associated with reduced expression and function of small artery skca, IKCA, and kir2.1 channels. Clin. Sci. 132 (4), 461–474. https://doi.org/10.1042/cs20171408. Greeno, E.W., Mantyh, P., Vercellotti, G.M., Moldow, C.F., 1993. Functional neurokinin 1 receptors for substance P are expressed by human vascular endothelium. J. Exp. Med. 177 (5), 1269–1276. https://doi.org/10.1084/jem.177.5.1269. Grgic, I., Eichler, I., Heinau, P., Si, H., Brakemeier, S., Hoyer, J., Ko¨hler, Ralf, 2005. Selective blockade of the intermediate-conductance ca 2+ -activated K + channel suppresses proliferation of microvascular and macrovascular endothelial cells and angiogenesis in vivo. Arterioscler. Thromb. Vasc. Biol. 25 (4), 704–709. https://doi. org/10.1161/01.atv.0000156399.12787.5c. Grgic, I., Kaistha, B.P., Hoyer, J., Köhler, R., 2009. Endothelial ca2+-activated k+ channels in normal and impaired edhf-dilator responses - relevance to cardiovascular pathologies and drug discovery. Br. J. Pharmacol. 157 (4), 509–526. https://doi. org/10.1111/j.1476-5381.2009.00132.x. Gross, O., Thomas, C.J., Guarda, G., Tschopp, J., 2011. The inflammasome: an integrated view. Immunol. Rev. 243 (1), 136–151. https://doi.org/10.1111/j.1600065x.2011.01046.x. Gryglewski, R.J., Uracz, W., Chłopicki, S., Marcinkiewicz, E., 2002. Bradykinin as a major endogenous regulator of endothelial function. Pediatr. Pathol. Mol. Med. 21 (3), 279–290. https://doi.org/10.1080/pdp.21.3.279.290. Haddock, R.E., Grayson, T.H., Morris, M.J., Howitt, L., Chadha, P.S., Sandow, S.L., 2011. Diet-induced obesity impairs endothelium-derived hyperpolarization via altered potassium channel signaling mechanisms. PLoS One 6 (1). https://doi.org/10.1371/ journal.pone.0016423. Halling, D.B., Kenrick, S.A., Riggs, A.F., Aldrich, R.W., 2014. Calcium-dependent stoichiometries of the KCA2.2 (SK) intracellular domain/calmodulin complex in solution. J. Gen. Physiol. 143 (2), 231–252. https://doi.org/10.1085/ jgp.201311007. He, G.-W., Yang, C.-Q., 1997. Superiority of hyperpolarizing to depolarizing cardioplegia in protection of coronary endothelial function. J. Thorac. Cardiovasc. Surg. 114 (4), 643–650. https://doi.org/10.1016/s0022-5223(97)70055-2. He, G.-W., Ge, Z.-D., Yim, A.P.C., Yang, Q., Zhang, R.-Z., 2004. Electrophysiologic and mechanical evidence of superiority of hyperpolarizing versus depolarizing cardioplegia in protection of endothelium-derived hyperpolarizing factor–mediated endothelial function: a study in coronary resistance arteries. J. Thorac. Cardiovasc. Surg. 127 (6), 1773–1780. https://doi.org/10.1016/j.jtcvs.2003.09.051. Hilgers, R.H., Webb, R.C., 2007. Reduced expression of skca and IKCA channel proteins in rat small mesenteric arteries during angiotensin II-induced hypertension. Am. J. Physiol.-Heart Circ. Physiol. 292 (5) https://doi.org/10.1152/ajpheart.00949.2006. Hornig, B., Drexler, H., 1997. Endothelial function and bradykinin in humans. Drugs 54 (Supplement 5), 42–47. https://doi.org/10.2165/00003495-199700545-00007. Hosoya, M., Ohashi, J., Sawada, A., Takaki, A., Shimokawa, H., 2010. Combination therapy with olmesartan and azelnidipine improves EDHF-mediated responses in diabetic apolipoprotein E-deficient mice. Circ. J. 74 (4), 798–806. https://doi.org/ 10.1253/circj.cj-09-0862. Huang, J.-H., He, G.-W., Xue, H.-M., Yao, X.-Q., Liu, X.-C., Underwood, M.J., Yang, Q., 2011. TRPC3 channel contributes to nitric oxide release: significance during normoxia and hypoxia–reoxygenation. Cardiovasc. Res. 91 (3), 472–482. https:// doi.org/10.1093/cvr/cvr102. Inoguchi, T., Ueda, F., Umeda, F., Yamashita, T., Nawata, H., 1995. Inhibition of intercellular communication via gap junction in cultured aortic endothelial cells by elevated glucose and Phorbol Ester. Biochem. Biophys. Res. Commun. 208 (2), 492–497. https://doi.org/10.1006/bbrc.1995.1365. Janaszak-Jasiecka, A., Siekierzycka, A., Płoska, A., Dobrucki, I.T., Kalinowski, L., 2021. Endothelial dysfunction driven by hypoxia—the influence of oxygen deficiency on no bioavailability. Biomolecules 11 (7), 982. https://doi.org/10.3390/ biom11070982. Kajikuri, J., Watanabe, Y., Ito, Y., Ito, R., Yamamoto, T., Itoh, T., 2009. Characteristic changes in coronary artery at the early hyperglycaemic stage in a rat type 2 diabetes model and the effects of pravastatin. Br. J. Pharm. 158, 621–632. Kirkland, E.B., Heincelman, M., Bishu, K.G., et al., 2018. Trends in healthcare expenditures among US adults with hypertension: national estimates, 2003-2014. J. Am. Heart Assoc. 7, e008731. Kizhakekuttu, T.J., Wang, J., Dharmashankar, K., Ying, R., Gutterman, D.D., Vita, J.A., Widlansky, M.E., 2012. Adverse alterations in mitochondrial function contribute to 13 S. Kant et al. European Journal of Cell Biology 101 (2022) 151208 type 2 diabetes mellitus–related endothelial dysfunction in humans. Arterioscler. Thromb. Vasc. Biol. 32 (10), 2531–2539. https://doi.org/10.1161/ atvbaha.112.256024. Kluge, M.A., Fetterman, J.L., Vita, J.A., 2013. Mitochondria and endothelial function. Circ. Res. 112 (8), 1171–1188. https://doi.org/10.1161/circresaha.111.300233. Köhler, R., Brakemeier, S., Kuhn, M., Behrens, C., Real, R., Degenhardt, C., et al., 2001. Impaired hyperpolarization in regenerated endothelium after balloon catheter injury. Circ. Res. 89, 174–179. Köhler, R., Degenhardt, C., Kuhn, M., Runkel, N., Paul, M., Hoyer, J., 2000. Expression and function of endothelial Ca2+ -activated K+ channels in human mesenteric artery: a single-cell reverse transcriptasepolymerase chain reaction and electrophysiological study in situ. Circ. Res. 87, 496–503. Kohler, M., Hirschberg, B., Bond, C.T., Kinzie, J.M., Marrion, N.V., Maylie, J., Adelman, J.P., 1996. Small-conductance, calcium-activated potassium channels from Mammalian Brain. Science 273 (5282), 1709–1714. https://doi.org/10.1126/ science.273.5282.1709. Köhler, R., Ruth, P., 2010. Endothelial dysfunction and blood pressure alterations in K+channel transgenic mice. Pflu¨g. Arch. - Eur. J. Physiol. 459 (6), 969–976. https:// doi.org/10.1007/s00424-010-0819-z. Kono, Keiji, Fujii, Hideki, Nakai, Kentaro, Goto, Shunsuke, Watanabe, Shuhei, Watanabe, Kentaro, Nishi, Shinichi, 2016. Relationship between Type of Hypertension and Renal Arteriolosclerosis in Chronic Glomerular Disease. Kidney and blood pressure research 41 (4), 374–383. https://doi.org/10.1159/000443440. Kowalczyk, A., Kleniewska, P., Kolodziejczyk, M., Skibska, B., Goraca, A., 2014. The role OF Endothelin-1 and Endothelin receptor antagonists in inflammatory response and sepsis. Arch. Immunol. Ther. Exp. 63 (1), 41–52. https://doi.org/10.1007/s00005014-0310-1. Koya, D., King, G.L., 1998. Protein kinase C activation and the development of diabetic complications. Diabetes 47 (6), 859–866. https://doi.org/10.2337/ diabetes.47.6.859. Kroigaard, C., Dalsgaard, T., Nielsen, G., Laursen, B.E., Pilegaard, H., Köhler, R., Simonsen, U., 2012. Activation of endothelial and epithelial KCA2.3 calciumactivated potassium channels by NS309 relaxes human small pulmonary arteries and bronchioles. Br. J. Pharmacol. 167 (1), 37–47. https://doi.org/10.1111/j.14765381.2012.01986.x. Kroigaard, C., Kudryavtseva, O., Dalsgaard, T., Wandall-Frostholm, C., Olesen, S.-P., Simonsen, U., 2013. KCA3.1 channel downregulation and impaired endotheliumderived hyperpolarization-type relaxation in pulmonary arteries from chronically hypoxic rats. Exp. Physiol. 98 (4), 957–969. https://doi.org/10.1113/ expphysiol.2012.066340. Krüger-Genge, A., Blocki, A., Franke, R.-P., Jung, F., 2019. Vascular endothelial cell biology: an update. Int. J. Mol. Sci. 20 (18), 4411. https://doi.org/10.3390/ ijms20184411. Kshatri, A.S., Gonzalez-Hernandez, A., Giraldez, T., 2018. Physiological roles and therapeutic potential of ca2+ activated potassium channels in the nervous system. Front. Mol. Neurosci. 11. https://doi.org/10.3389/fnmol.2018.00258. Kuroiwa, M., Aoki, H., Kobayashi, S., Nishimura, J., Kanaide, H., 1995. Mechanism of endothelium-dependent relaxation induced by substance P in the coronary artery of the pig. Br. J. Pharmacol. 116 (3), 2040–2047. https://doi.org/10.1111/j.14765381.1995.tb16409.x. Kvietys, P., Granger, D.N., 2014. Mediators and mechanisms of the increased blood FLOW, Vascular PERMEABILITY, and blood Vessel proliferation in Inflamed tissue. Pathobiol. Hum. Dis. 244–257. https://doi.org/10.1016/b978-0-12-3864567.01803-7. Ledoux, J., Werner, M.E., Brayden, J.E., Nelson, M.T., 2006. Calcium-activated potassium channels and the regulation of vascular tone. Physiology 21 (1), 69–78. https://doi.org/10.1152/physiol.00040.2005. Lee, C.-H., MacKinnon, R., 2018. Activation mechanism of a human SK-calmodulin channel complex elucidated by Cryo-EM structures. Science 360 (6388), 508–513. https://doi.org/10.1126/science.aas9466. Lee, S., Park, M., So, I., Earm, Y.E., 1994. NADH and NAD modulates CA2+-activated K+ channels in small pulmonary arterial smooth muscle cells of the rabbit. Pflu¨g. Arch. Eur. J. Physiol. 427 (3–4), 378–380. https://doi.org/10.1007/bf00374548. Lee, U.S., Cui, J., 2010. BK channel activation: structural and functional insights. Trends Neurosci. 33 (9), 415–423. https://doi.org/10.1016/j.tins.2010.06.004. Leo, C., Hart, J., Woodman, O., 2011. Impairment of both nitric oxide-mediated and EDHF-type relaxation in small mesenteric arteries from rats with streptozotocininduced diabetes. Br. J. Pharmacol. 162 (2), 365–377. https://doi.org/10.1111/ j.1476-5381.2010.01023.x. Li, B., Li, Y., Liu, K., Wang, X., Qi, J., Wang, B., Wang, Y., 2016. High glucose decreases claudins-5 and -11 in cardiac microvascular endothelial cells: antagonistic effects of tongxinluo. Endocr. Res. 42 (1), 15–21. https://doi.org/10.3109/ 07435800.2016.1163723. Li, W., Aldrich, R.W., 2011. Electrostatic influences of charged inner pore residues on the conductance and gating of small conductance ca2+ activated k+ channels. Proc. Natl. Acad. Sci. USA 108 (15), 5946–5953. https://doi.org/10.1073/ pnas.1103090108. Liang, X., Arullampalam, P., Yang, Z., Ming, X.-F., 2019. Hypoxia enhances endothelial intercellular adhesion molecule 1 protein level through upregulation of arginase type II and mitochondrial oxidative stress. Front. Physiol. 10. https://doi.org/ 10.3389/fphys.2019.01003. Liu, C.-H., Hua, N., Fu, X., Pan, Y.-L., Li, B., Li, X.-D., 2018. Metformin regulates atrial SK2 and SK3 expression through inhibiting the PKC/ERK signaling pathway in type 2 diabetic rats. BMC Cardiovasc. Disord. 18 (1) https://doi.org/10.1186/s12872-0180950-x. Liu, M., Sanyal, S., Gao, G., Gurung, I.S., Zhu, X., Gaconnet, G., Kerchner, L.J., Shang, L. L., Huang, C.L.-H., Grace, A., London, B., Dudley, S.C., 2009. Cardiac na + current regulation by pyridine nucleotides. Circ. Res. 105 (8), 737–745. https://doi.org/ 10.1161/circresaha.109.197277. Liu, Y., Cole, V., Lawandy, I., Ehsan, A., Sellke, F.W., Feng, J., 2018. Decreased coronary arteriolar response to KCA channel opener after cardioplegic arrest in diabetic patients. Mol. Cell. Biochem. 445 (1–2), 187–194. https://doi.org/10.1007/s11010017-3264-x. Liu, Y., Kabakov, A.Y., Xie, A., Shi, G., Singh, A.K., Sodha, N.R., Ehsan, A., Usheva, A., Agbortoko, V., Koren, G., Dudley, S.C., Sellke, F.W., Feng, J., 2020. Metabolic regulation of endothelial SK channels and human coronary microvascular function. Int. J. Cardiol. 312, 1–9. https://doi.org/10.1016/j.ijcard.2020.03.028. Liu, Y., Sellke, E.W., Feng, J., Clements, R.T., Sodha, N.R., Khabbaz, K.R., Senthilnathan, V., Alper, S.L., Sellke, F.W., 2008. Calcium-activated potassium channels contribute to human skeletal muscle microvascular endothelial dysfunction related to cardiopulmonary bypass. Surgery 144 (2), 239–244. https://doi.org/ 10.1016/j.surg.2008.03.032. Liu, Y., Xie, A., Singh, A.K., Ehsan, A., Choudhary, G., Dudley, S., Sellke, F.W., Feng, J., 2015. Inactivation of endothelial small/intermediate conductance of calciumactivated potassium channels contributes to coronary arteriolar dysfunction in diabetic patients. J. Am. Heart Assoc. 4 (8) https://doi.org/10.1161/ jaha.115.002062. Lu, D., Kassab, G.S., 2011. Role of shear stress and stretch in vascular mechanobiology. J. R. Soc. Interface 8 (63), 1379–1385. https://doi.org/10.1098/rsif.2011.0177. Lu, L., Zhang, Q., Timofeyev, V., Zhang, Z., Young, J.N., Shin, H.-S., Knowlton, A.A., Chiamvimonvat, N., 2007. Molecular coupling of a ca 2+ -activated k + channel to ltype ca 2+ channels via α-actinin2. Circ. Res. 100 (1), 112–120. https://doi.org/ 10.1161/01.res.0000253095.44186.72. Luksha, L., Agewall, S., Kublickiene, K., 2009. Endothelium-derived hyperpolarizing factor in vascular physiology and cardiovascular disease. Atherosclerosis 202 (2), 330–344. https://doi.org/10.1016/j.atherosclerosis.2008.06.008. Luksha, L., Nisell, H., Kublickiene, K., 2004. The mechanism of EDHF-mediated responses in subcutaneous small arteries from healthy pregnant women. Am. J. Physiol.-Regul. Integr. Comp. Physiol. 286 (6) https://doi.org/10.1152/ ajpregu.00550.2003. Ma, X., Du, J., Zhang, P., Deng, J., Liu, J., Lam, F.F., Li, R.A., Huang, Y., Jin, J., Yao, X., 2013. Functional role of TRPV4-KCa2.3 signaling in vascular endothelial cells in normal and streptozotocin-induced diabetic rats. Hypertension 62 (1), 134–139. https://doi.org/10.1161/HYPERTENSIONAHA.113.01500. Majed, Batoule, Khalil, Raouf, 2012. Molecular Mechanisms Regulating the Vascular Prostacydlin Pathways and Their Adaptation during Pregnancy and in the Newborn. Pharmacological Reviews 64 (4), 540–582. https://doi.org/10.1124/pr.111.004770. Makino, A., Ohuchi, K., Kamata, K., 2000. Mechanisms underlying the attenuation of endothelium-dependent vasodilatation in the mesenteric arterial bed of the streptozotocin-induced diabetic rat. Br. J. Pharm. 130, 549–556. Marrelli SP, Childres WF, Goddard-Finegold J., Bryan RMJ. Potentiated EDHF-mediated dilations in the rat middle cerebral artery following ischemia/reperfusion. In: PM V, editor. EDHF 2000. London, England: Taylor & Francis; 2001. [Google Scholar]. Matsumoto, S., Shimabukuro, M., Fukuda, D., Soeki, T., Yamakawa, K., Masuzaki, H., Sata, M., 2014. Azilsartan, an angiotensin II type 1 receptor blocker, restores endothelial function by reducing vascular inflammation and by increasing the phosphorylation ratio SER1177/thr497 of endothelial nitric oxide synthase in diabetic mice. Cardiovasc. Diabetol. 13 (1) https://doi.org/10.1186/1475-2840-1330. Matsumoto, T., Ishida, K., Taguchi, K., Kobayashi, T., Kamata, K., 2010. Losartan normalizes endothelium-derived hyperpolarizing factor–mediated relaxation by activating Ca2+-activated K+ channels in mesenteric artery from type 2 diabetic GK rat. J. Pharmacol. Sci. 112 (3), 299–309. https://doi.org/10.1254/jphs.09308fp. Matsumoto, T., Kobayashi, T., Kamata, K., 2006. Mechanisms underlying the impaired EDHF-type relaxation response in mesenteric arteries from Otsuka Long-Evans Tokushima Fatty (OLETF) rats. Eur. J. Pharmacol. 538 (1–3), 132–140. https://doi. org/10.1016/j.ejphar.2006.04.006. Matsumoto, T., Nakayama, N., Ishida, K., Kobayashi, T., Kamata, K., 2009. Eicosapentaenoic acid improves imbalance between vasodilator and vasoconstrictor actions of endothelium-derived factors in mesenteric arteries from rats at chronic stage of type 2 diabetes. J. Pharmacol. Exp. Ther. 329 (1), 324–334. https://doi.org/ 10.1124/jpet.108.148718. Matsumoto, T., Noguchi, E., Ishida, K., Kobayashi, T., Yamada, N., Kamata, K., 2008. Metformin normalizes endothelial function by suppressing vasoconstrictor prostanoids in mesenteric arteries from OLETF rats, a model of type 2 diabetes. Am. J. Physiol. Heart Circ. Physiol. 295 (3), H1165–H1176. https://doi.org/10.1152/ ajpheart.00486.2008. Matsumoto, T., Wakabayashi, K., Kobayashi, T., Kamata, K., 2004. Alterations in vascular endothelial function in the aorta and mesenteric artery in type II diabetic rats. Can. J. Physiol. Pharmacol. 82 (3), 175–182. https://doi.org/10.1139/y04-002. Matsumoto, T., Wakabayashi, K., Kobayashi, T., Kamata, K., 2004. Diabetes-related changes in cAMP-dependent protein kinase activity and decrease in relaxation response in rat mesenteric artery. Am. J. Physiol. Heart Circ. Physiol. 287 (3), H1064–H1071. https://doi.org/10.1152/ajpheart.00069.2004. Maylie, J., Bond, C.T., Herson, P.S., Lee, W.-S., Adelman, J.P., 2004. Small conductance ca2+-activated K+channels and calmodulin. J. Physiol. 554 (2), 255–261. https:// doi.org/10.1113/jphysiol.2003.049072. Mensah, G.A., Croft, J.B., Giles, W.H., 2002. The heart, kidney, and brain as target organs in hypertension. Cardiol. Clin. 20 (2), 225–247. https://doi.org/10.1016/s07338651(02)00004-8. 14 S. Kant et al. European Journal of Cell Biology 101 (2022) 151208 Mills, K.T., Stefanescu, A., He, J., 2020. The global epidemiology of hypertension. Nat. Rev. Nephrol. 16 (4), 223–237. https://doi.org/10.1038/s41581-019-0244-2. Mishra, R.C., Kyle, B.D., Kendrick, D.J., Svystonyuk, D., Kieser, T.M., Fedak, P.W.M., Wulff, H., Braun, A.P., 2021a. KCA channel activation normalizes endothelial function in type 2 diabetic resistance arteries by improving intracellular ca2+ mobilization. Metabolism 114, 154390. https://doi.org/10.1016/j. metabol.2020.154390. Mishra, R.C., Kyle, B.D., Kendrick, D.J., Svystonyuk, D., Kieser, T.M., Fedak, P.W.M., Wulff, H., Braun, A.P., 2021b. KCA channel activation normalizes endothelial function in type 2 diabetic resistance arteries by improving intracellular ca2+ mobilization. Metabolism 114, 154390. https://doi.org/10.1016/j. metabol.2020.154390. Miura, M., 1996. Regulation and failure of coronary circulation. Jpn. Heart J. 37 (5), 585–602. https://doi.org/10.1536/ihj.37.585. Morikawa, K., Matoba, T., Kubota, H., Hatanaka, M., Fujiki, T., Takahashi, S., Takeshita, A., Shimokawa, H., 2005. Influence of diabetes mellitus, hypercholesterolemia, and their combination on EDHF-mediated responses in mice. J. Cardiovasc. Pharmacol. 45 (5), 485–490. https://doi.org/10.1097/01. fjc.0000159657.93922.cb. Nam, Y.-W., Orfali, R., Liu, T., Yu, K., Cui, M., Wulff, H., Zhang, M., 2017. Structural insights into the potency of SK channel positive modulators. Sci. Rep. 7 (1) https:// doi.org/10.1038/s41598-017-16607-8. Ning, B., Chen, Y., Waqar, A.B., Yan, H., Shiomi, M., Zhang, J., Chen, Y.E., Wang, Y., Itabe, H., Liang, J., Fan, J., 2018. Hypertension enhances advanced atherosclerosis and induces cardiac death in watanabe heritable hyperlipidemic rabbits. Am. J. Pathol. 188 (12), 2936–2947. https://doi.org/10.1016/j.ajpath.2018.08.007. Nishikawa, Y., Stepp, D.W., Chilian, W.M., 2000. Nitric oxide exerts feedback inhibition on edhf-induced coronary arteriolar dilation in vivo. Am. J. Physiol.-Heart Circ. Physiol. 279 (2) https://doi.org/10.1152/ajpheart.2000.279.2.h459. Oniki, H., Goto, K., Fujii, K., Kansui, Y., Murakami, N., Ohtsubo, T., Matsumura, K., Kitazono, T., 2013. Effects of the superoxide dismutase mimetic tempol on impaired endothelium-dependent and endothelium-independent relaxations in type II diabetic rats. Clin. Exp. Hypertens. 35 (2), 112–119. https://doi.org/10.3109/ 10641963.2012.702829. Oniki, H., Fujii, K., Kansui, Y., Goto, K., Iida, M., 2006. Effects of angiotensin II receptor antagonist on impaired endothelium-dependent and endothelium-independent relaxations in type II diabetic rats. J. Hypertens. 24, 331–338. Ozgen, N., Dun, W., Sosunov, E., Anyukhovsky, E., Hirose, M., Duffy, H., Boyden, P., Rosen, M., 2007. Early electrical remodeling in rabbit pulmonary vein results from trafficking of intracellular sk2 channels to membrane sites. Cardiovasc. Res. 75 (4), 758–769. https://doi.org/10.1016/j.cardiores.2007.05.008. Ozkor, M.A., Quyyumi, A.A., 2011. Endothelium-derived hyperpolarizing factor and vascular function. Cardiol. Res. Pract. 2011, 1–12. https://doi.org/10.4061/2011/ 156146. Papassotiriou, J., Kohler, R., Prenen, J., Krause, H., Akbar, M., Eggermont, J., et al., 2000. Endothelial K(+) channel lacks the Ca(2+) sensitivityregulating beta subunit. FASEB J. 14, 885–894. Park, Y., Capobianco, S., Gao, X., Falck, J.R., Dellsperger, K.C., Zhang, C., 2008. Role of EDHF in type 2 diabetes-induced endothelial dysfunction. Am. J. Physiol. Heart Circ. Physiol. 295 (5), H1982–H1988. https://doi.org/10.1152/ajpheart.01261.2007. Peters, K., 2003. Molecular basis of endothelial dysfunction in sepsis. Cardiovasc. Res. 60 (1), 49–57. https://doi.org/10.1016/s0008-6363(03)00397-3. Piga, R., Naito, Y., Kokura, S., Handa, O., Yoshikawa, T., 2007. Short-term high glucose exposure induces monocyte-endothelial cells adhesion and transmigration by increasing VCAM-1 and MCP-1 expression in human aortic endothelial cells. Atherosclerosis 193 (2), 328–334. https://doi.org/10.1016/j. atherosclerosis.2006.09.016. Radtke, Josephine, Schmidt, Kjestine, Wulff, Heike, Kholer, Ralf, Wit, Cor, 2013. Activation of KCa3.1 by SKA-31 induces arteriolar dilatation and lowers blood pressure in normo- and hypertensive connexin40-deficient mice. British Journal of Pharmacology 170 (2), 293–303. https://doi.org/10.1111/bph.1267. Rajendran, Peramaiyan, Rengarajan, Thamaraiselvan, Thangavel, Jayakumar, Nishigaki, Yutaka, Sakthisekaran, Dhanapal, Sethi, Gautam, Nishigaki, Ikuo, 2013. The Vascular Endothelium and Human Diseases. International Journal of Biological Sciences 9 (10), 1057–1069. https://doi.org/10.7150/ijbs.7502. Robich, M.P., Araujo, E.G., Feng, J., Osipov, R.M., Clements, R.T., Bianchi, C., Sellke, F. W., 2010. Altered coronary microvascular serotonin receptor expression after coronary artery bypass grafting with cardiopulmonary bypass. J. Thorac. Cardiovasc. Surg. 139 (4), 1033–1040. https://doi.org/10.1016/j.jtcvs.2009.05.032. Sandow, S.L., Goto, K., Rummery, N.M., Hill, C.E., 2004. Developmental changes in myoendothelial gap junction mediated vasodilator activity in the rat saphenous artery. J. Physiol. 556 (3), 875–886. https://doi.org/10.1113/ jphysiol.2003.058669. Sandow, Shaun, Neylon, Craig, Chen, Mao, Garland, Christopher, 2006. Spatial separation of endothelial small and intermediate conductance calcium-activated potassium channels (KCa) and connexins: possible relationship to vasodilator function? Journal of anatomy 209 (5), 689–698. https://doi.org/10.1111/j.14697580.2006.99647.x. Schach, C., Resch, M., Schmid, P.M., Riegger, G.A., Endemann, D.H., 2014a. Type 2 diabetes: increased expression and contribution of IKCa channels to vasodilation in small mesenteric arteries of ZDF rats. Am. J. Physiol. Heart Circ. Physiol. 307 (8), H1093–H1102. https://doi.org/10.1152/ajpheart.00240.2013. Schach, C., Resch, M., Schmid, P.M., Riegger, G.A., Endemann, D.H., 2014b. Type 2 diabetes: increased expression and contribution of IKCa channels to vasodilation in small mesenteric arteries of ZDF rats. Am. J. Physiol. Heart Circ. Physiol. 307 (8), H1093–H1102. https://doi.org/10.1152/ajpheart.00240.2013. Schiffrin, E.L., 2002. Beyond blood pressure: the endothelium and atherosclerosis progression. Am. J. Hypertens. 15 (S5) https://doi.org/10.1016/s0895-7061(02) 03006-6. Schmidt, K., Dubrovska, G., Nielsen, G., Fesüs, G., Uhrenholt, T.R., Hansen, P.B., Gudermann, T., Dietrich, A., Gollasch, M., de Wit, C., Köhler, R., 2010. Amplification of edhf-type vasodilatations in trpc1-deficient mice. Br. J. Pharmacol. 161 (8), 1722–1733. https://doi.org/10.1111/j.1476-5381.2010.00985.x. Seki, T., Goto, K., Kiyohara, K., Kansui, Y., Murakami, N., Haga, Y., Ohtsubo, T., Matsumura, K., Kitazono, T., 2017. Downregulation of endothelial transient receptor potential vanilloid type 4 channel and small-conductance of ca 2+ -activated K + channels underpins impaired endothelium-dependent hyperpolarization in hypertension. Hypertension 69 (1), 143–153. https://doi.org/10.1161/ hypertensionaha.116.07110. Sharma, N.R., Davis, M.J., 1995. Substance P-induced calcium entry in endothelial cells is secondary to depletion of intracellular stores. Am. J. Physiol.-Heart Circ. Physiol. 268 (3) https://doi.org/10.1152/ajpheart.1995.268.3.h962. Shi, Y., 2006. Augmented endothelium-derived hyperpolarizing factor-mediated relaxations attenuate endothelial dysfunction in femoral and mesenteric, but not in carotid arteries from type I diabetic rats. J. Pharm. Exp. Ther. 318, 276–281. Shi, Y, Vanhoutte, PM, 2009. Reactive oxygen-derived free radicals are key to the endothelial dysfunction of diabetes. Journal of Diabetes 1 (3), 151–162. https://doi. org/10.1111/j.1753-0407.2009.00030.x. Shi, Y., Vanhoutte, P.M., 2017. Macro- and microvascular endothelial dysfunction in diabetes. J. Diabetes 9 (5), 434–449. https://doi.org/10.1111/1753-0407.12521. Si, H., Heyken, W.-T., Wo¨lfle Stephanie, E., Tysiac, M., Schubert, R., Grgic, I., Vilianovich, L., Giebing, Gu¨nter, Maier, T., Gross, V., Bader, M., de Wit, C., Hoyer, J., Ko¨hler, Ralf, 2006. Impaired endothelium-derived hyperpolarizing factor-mediated dilations and increased blood pressure in mice deficient of the intermediateconductance ca 2+ -activated K + channel. Circ. Res. 99 (5), 537–544. https://doi. org/10.1161/01.res.0000238377.08219.0c. Simonet, S., Isabelle, M., Bousquenaud, M., Clavreul, N., Félétou, M., VayssettesCourchay, C., Verbeuren, T.J., 2012. KCA3.1 channels maintain endotheliumdependent vasodilatation in isolated perfused kidneys of spontaneously hypertensive rats after chronic inhibition of NOS. Br. J. Pharmacol. 167 (4), 854–867. https://doi. org/10.1111/j.1476-5381.2012.02062.x. Slegtenhorst, B.R., Dor, F.J., Rodriguez, H., Voskuil, F.J., Tullius, S.G., 2014. Ischemia/ reperfusion injury and its consequences on immunity and inflammation. Curr. Transplant. Rep. 1 (3), 147–154. https://doi.org/10.1007/s40472-014-0017-6. Sodha, N.R., Clements, R.T., Bianchi, C., Sellke, F.W., 2008. Cardiopulmonary bypass with cardioplegic arrest activates protein kinase C in the human myocardium. J. Am. Coll. Surg. 206 (1), 33–41. https://doi.org/10.1016/j.jamcollsurg.2007.06.308. Song, Y., Xing, H., He, Y., Zhang, Z., Shi, G., Wu, S., Liu, Y., Harrington, E.O., Sellke, F. W., Feng, J., 2021. Inhibition of mitochondrial reactive oxygen species improves coronary endothelial function after cardioplegic hypoxia/reoxygenation. J. Thorac. Cardiovasc. Surg. https://doi.org/10.1016/j.jtcvs.2021.06.029. Sotníková, R., Nedelčevová, J., Navarová, J., Nosáĺová, V., Drábiková, K., Szöcs, K., Křenek, P., Kyseĺová, Z., Bezek, S., Knezl, V., Dřímal, J., Brosková, Z., Kristová, V., Okruhlicová, L., Bernátová, I., Bauer, V., 2011a. Protection of the vascular endothelium in experimental situations. Interdiscip. Toxicol. 4 (1), 20–26. https:// doi.org/10.2478/v10102-011-0005-y. Sotníková, R., Nedelčevová, J., Navarová, J., Nosáĺová, V., Drábiková, K., Szöcs, K., Křenek, P., Kyseĺová, Z., Bezek, S., Knezl, V., Dřímal, J., Brosková, Z., Kristová, V., Okruhlicová, L., Bernátová, I., Bauer, V., 2011b. Protection of the vascular endothelium in experimental situations. Interdiscip. Toxicol. 4 (1), 20–26. https:// doi.org/10.2478/v10102-011-0005-y. Stocker, M., 2004. Ca2+-activated K+ channels: molecular determinants and function of the SK family. Nat. Rev. Neurosci. 5 (10), 758–770. https://doi.org/10.1038/ nrn1516. Tabit, Corey, Chung, William, Hamburg, Naomi, Vita, Joseph, 2010. Endothelial dysfunction in diabetes mellitus: Molecular mechanisms and clinical implications. Reviews in endocrine & metabolic disorders 11 (1), 61–74. https://doi.org/10.1007/ s11154-010-9134-4. Takai, J., Santu, A., Zheng, H., Koh, S.D., Ohta, M., Filimban, L.M., Lemaître, V., Teraoka, R., Jo, H., Miura, H., 2013. Laminar shear stress upregulates endothelial ca2+-activated K+ channels KCA2.3 and KCA3.1 via a Ca2+/calmodulin-dependent protein kinase kinase/AKT/P300 cascade. Am. J. Physiol.-Heart Circ. Physiol. 305 (4) https://doi.org/10.1152/ajpheart.00642.2012. Tangsucharit, P., Takatori, S., Zamami, Y., Goda, M., Pakdeechote, P., Kawasaki, H., Takayama, F., 2016. Muscarinic acetylcholine receptor M1 and M3 subtypes mediate acetylcholine-induced endothelium-independent vasodilatation in rat mesenteric arteries. J. Pharmacol. Sci. 130 (1), 24–32. https://doi.org/10.1016/j. jphs.2015.12.005. Taylor, M.S., Bonev, A.D., Gross, T.P., Eckman, D.M., Brayden, J.E., Bond, C.T., Adelman, J.P., Nelson, M.T., 2003. Altered expression of small-conductance ca 2+ -activated K + (SK3) channels modulates arterial tone and blood pressure. Circ. Res. 93 (2), 124–131. https://doi.org/10.1161/01.res.0000081980.63146.69. Tsai, S.H., Hein, T.W., Kuo, L., Yang, V.C., 2011. High glucose impairs EDHF-mediated dilation of coronary arterioles via reduced cytochrome P450 activity. Microvasc. Res. 82 (3), 356–363. https://doi.org/10.1016/j.mvr.2011.09.008. Tschopp, J., Schroder, K., 2010. NLRP3 inflammasome activation: the convergence of multiple signalling pathways on ROS production? Nat. Rev. Immunol. 10 (3), 210–215. https://doi.org/10.1038/nri2725. Vanhoutte, P.M., Feletou, M., Taddei, S., 2005. Endothelium-dependent contractions in hypertension. Br. J. Pharmacol. 144 (4), 449–458. https://doi.org/10.1038/sj. bjp.0706042. 15 S. Kant et al. European Journal of Cell Biology 101 (2022) 151208 Vriese, AS, Verbeuren, TJ, Van de Voorde, J, Lameire, NH, Vanhoutte, PM, 2000. Endothelial dysfunction in diabetes. British journal of pharmacology 130 (5), 963–974. https://doi.org/10.1038/sj.bjp.0703393. Walch, L., Brink, C., Norel, X., 2001. The muscarinic receptor subtypes in human blood vessels. Therapie 56 (3), 223–226. Wang, X.-C., Sun, W.-T., Fu, J., Huang, J.-H., Yu, C.-M., Underwood, M.J., He, G.-W., Yang, Q., 2017. Impairment of coronary endothelial function by hypoxiareoxygenation involves TRPC3 inhibition-mediated KCA channel dysfunction: implication in ischemia-reperfusion injury. Sci. Rep. 7 (1) https://doi.org/10.1038/ s41598-017-06247-3. Wei, A.D., Gutman, G.A., Aldrich, R., Chandy, K.G., Grissmer, S., Wulff, H., 2005. International Union of Pharmacology. LII. nomenclature and molecular relationships of calcium-activated potassium channels. Pharmacol. Rev. 57 (4), 463–472. https:// doi.org/10.1124/pr.57.4.9. Weiss, D., Kools, J.J., Taylor, W.R., 2001. Angiotensin II–induced hypertension accelerates the development of atherosclerosis in Apoe-deficient mice. Circulation 103 (3), 448–454. https://doi.org/10.1161/01.cir.103.3.448. Weston, A.H., Porter, E.L., Harno, E., Edwards, G., 2010. Impairment of endothelial SKCA channels and of downstream hyperpolarizing pathways in mesenteric arteries from spontaneously hypertensive rats. Br. J. Pharmacol. 160 (4), 836–843. https:// doi.org/10.1111/j.1476-5381.2010.00657.x. Wigg, S.J., Tare, M., Tonta, M.A., O’Brien, R.C., Meredith, I.T., Parkington, H.C., 2001. Comparison of effects of diabetes mellitus on an EDHF-dependent and an EDHFindependent artery. Am. J. Physiol. Heart Circ. Physiol. 281 (1), H232–H240. https://doi.org/10.1152/ajpheart.2001.281.1.H232. Wilson, P.W.F., D’Agostino, R.B., Parise, H., Sullivan, L., Meigs, J.B., 2005. Metabolic syndrome as a precursor of cardiovascular disease and type 2 diabetes mellitus. Circulation 112 (20), 3066–3072. https://doi.org/10.1161/ circulationaha.105.539528. Wu, R.S., Marx, S.O., 2010. The BK potassium channel in the vascular smooth muscle and kidney: α- and β-subunits. Kidney Int. 78 (10), 963–974. https://doi.org/10.1038/ ki.2010.325. Wulff, H., Köhler, R., 2013. Endothelial small-conductance and intermediateconductance KCA channels. J. Cardiovasc. Pharmacol. 61 (2), 102–112. https://doi. org/10.1097/fjc.0b013e318279ba20. Xia, P., Inoguchi, T., Kern, T.S., Engerman, R.L., Oates, P.J., King, G.L., 1994. Characterization of the mechanism for the chronic activation of diacylglycerolprotein kinase C pathway in diabetes and hypergalactosemia. Diabetes 43 (9), 1122–1129. https://doi.org/10.2337/diab.43.9.1122. Xing, H., Zhang, Z., Shi, G., He, Y., Song, Y., Liu, Y., Harrington, E.O., Sellke, F.W., Feng, J., 2021. Chronic inhibition of mROS protects against coronary endothelial dysfunction in mice with diabetes. Front. Cell Dev. Biol. 9, 643810 https://doi.org/ 10.3389/fcell.2021.643810. Yamamoto, Y., Imaeda, K., Suzuki, H., 1999. Endothelium-dependent hyperpolarization and intercellular electrical coupling in guinea-pig mesenteric arterioles. J. Physiol. 514 (2), 505–513. https://doi.org/10.1111/j.1469-7793.1999.505ae.x. Yang, H., Zhang, G., Cui, J., 2015. BK channels: Multiple sensors, one activation gate. Front. Physiol. 6 https://doi.org/10.3389/fphys.2015.00029. Yang, Q., He, G.W., Underwood, M.J., Yu, C.M., 2016. Cellular and molecular mechanisms of endothelial ischemia/reperfusion injury: perspectives and implications for postischemic myocardial protection. Am. J. Transl. Res. 8 (2), 765–777. Yang, Q., Huang, J.-H., Man, Y.-B., Yao, X.-Q., He, G.-W., 2011. Use of intermediate/ small conductance calcium-activated potassium-channel activator for Endothelial Protection. J. Thorac. Cardiovasc. Surg. 141 (2) https://doi.org/10.1016/j. jtcvs.2010.04.005. Yang, T., Zhang, H.-L., Liang, Q., Shi, Y., Mei, Y.-A., Barrett, P.Q., Hu, C., 2016. Smallconductance ca 2+ -activated potassium channels negatively regulate aldosterone secretion in human adrenocortical cells. Hypertension 68 (3), 785–795. https://doi. org/10.1161/hypertensionaha.116.07094. Yang, Y., Li, P.-Y., Cheng, J., Mao, L., Wen, J., Tan, X.-Q., Liu, Z.-F., Zeng, X.-R., 2013. Function of BK ca channels is reduced in human vascular smooth muscle cells from Han Chinese patients with hypertension. Hypertension 61 (2), 519–525. https://doi. org/10.1161/hypertensionaha.111.00211. Yap, F.C., Weber, D.S., Taylor, M.S., Townsley, M.I., Comer, B.S., Maylie, J., Adelman, J. P., Lin, M.T., 2016a. Endothelial SK3 channel-associated CA2+ microdomains modulate blood pressure. Am. J. Physiol.-Heart Circ. Physiol. 310 (9) https://doi. org/10.1152/ajpheart.00787.2015. Yap, F.C., Weber, D.S., Taylor, M.S., Townsley, M.I., Comer, B.S., Maylie, J., Adelman, J. P., Lin, M.T., 2016b. Endothelial SK3 channel-associated CA2+ microdomains modulate blood pressure. Am. J. Physiol.-Heart Circ. Physiol. 310 (9) https://doi. org/10.1152/ajpheart.00787.2015. Yin, D.D., Wang, Q.C., Zhou, X., Li, Y., 2017a. Endothelial dysfunction in renal arcuate arteries of obese Zucker rats: the roles of nitric oxide, endothelium-derived hyperpolarizing factors, and calcium-activated K+ channels. PLoS One 12 (8), e0183124. https://doi.org/10.1371/journal.pone.0183124. Yin, D., Wang, Q., Zhou, X., Li, Y., 2017b. Endothelial dysfunction in renal arcuate arteries of obese Zucker rats: the roles of nitric oxide, endothelium-derived hyperpolarizing factors, and calcium-activated K+ channels. PLoS One 12 (8). https://doi.org/10.1371/journal.pone.0183124. Yin, D., Wang, Q., Zhou, X., Li, Y., 2017c. Endothelial dysfunction in renal arcuate arteries of obese Zucker rats: the roles of nitric oxide, endothelium-derived hyperpolarizing factors, and calcium-activated K+ channels. PLoS One 12 (8). https://doi.org/10.1371/journal.pone.0183124. Zhang, Z., Shi, G., Liu, Y., Xing, H., Kabakov, A.Y., Zhao, A.S., Agbortoko, V., Kim, J., Singh, A.K., Koren, G., Harrington, E.O., Sellke, F.W., Feng, J., 2020. Coronary endothelial dysfunction prevented by small-conductance calcium-activated potassium channel activator in mice and patients with diabetes. J. Thorac. Cardiovasc. Surg. 160 (6) https://doi.org/10.1016/j.jtcvs.2020.01.078. Zhu, J.-H., Jia, R.-P., Xu, L.-W., Wu, J.-P., Wang, Z.-Z., Wang, S.-K., Bo, C.-J., 2010. Reduced expression of SK3 and IK1 channel proteins in the cavernous tissue of diabetic rats. Asian J. Androl. 12, 599–604. Ziberna, L., Lunder, M., Kužner, J., Drevenšek, G., 2009. NORMOTHERMIC and hypothermic models for studying the deleterious effects of hypoxia-reoxygenation on EDHF-mediated relaxation in isolated porcine coronary arteries. J. Pharmacol. Toxicol. Methods 59 (1), 1–6. https://doi.org/10.1016/j.vascn.2008.09.003. Zyrianova, T., Lopez, B., Liao, A., Gu, C., Wong, L., Ottolia, M., Olcese, R., Schwingshackl, A., 2021. BK channels regulate LPS-induced CCL-2 release from human pulmonary endothelial cells. Am. J. Respir. Cell Mol. Biol. 64 (2), 224–234. https://doi.org/10.1165/rcmb.2020-0228oc. Further reading Carneiro, Fernando, Lima, Victor, Carneiro, Zidonia, Dorrance, Anne, Webb, Clinton, Tostes, Rita, 2009. Up-regulation of intermediate calcium-activated potassium channels (IKCa), counterbalances the impaired endothelium-dependent vasodilation in SHRSP. Translational research: the journal of laboratory and clinical medicine 154 (4), 183–193. https://doi.org/10.1016/j.trsl.2009.07.003. Carvalho-de-Souza, J.L., Varanda, W.A., Tostes, R.C., Chignalia, A.Z., 2013. BK channels in cardiovascular diseases and aging. Aging Dis. 4 (1), 38–49. Félétou, M., Köhler, R., Vanhoutte, P.M., 2011. Nitric oxide: orchestrator of endotheliumdependent responses. Ann. Med. 44 (7), 694–716. https://doi.org/10.3109/ 07853890.2011.585658. McNeish, A.J., Sandow, S.L., Neylon, C.B., Chen, M.X., Dora, K.A., Garland, C.J., 2006. Evidence for involvement of both IK CA and SK ca channels in hyperpolarizing responses of the rat middle cerebral artery. Stroke 37 (5), 1277–1282. https://doi. org/10.1161/01.str.0000217307.71231.43. Tajbakhsh, N., Sokoya, E.M., 2014. Compromised endothelium-dependent hyperpolarization-mediated dilations can be rescued by NS309 in obese Zucker rats. Microcirculation 21 (8), 747–753. https://doi.org/10.1111/micc.12157. Ziegler, O., Anderson, K., Liu, Y., Ehsan, A., Fingleton, J., Sodha, N., Feng, J., Sellke, F. W., 2020. Skeletal muscle microvasculature response to β-adrenergic stimuli is diminished with cardiac surgery. Surgery 167 (2), 493–498. https://doi.org/ 10.1016/j.surg.2019.07.018. 16