Next Article in Journal
The Role of ATP-Binding Cassette Subfamily A in Colorectal Cancer Progression and Resistance
Next Article in Special Issue
Gestational Diabetes Mellitus and Small-for-Gestational-Age: An Insight into the Placental Molecular Biomarkers
Previous Article in Journal
The Machinery of Exosomes: Biogenesis, Release, and Uptake
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Ca2+-Activated K+ Channels and the Regulation of the Uteroplacental Circulation

Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(2), 1349; https://doi.org/10.3390/ijms24021349
Submission received: 14 December 2022 / Revised: 6 January 2023 / Accepted: 8 January 2023 / Published: 10 January 2023
(This article belongs to the Special Issue Pathogenesis of Pregnancy-Related Complication 2023)

Abstract

:
Adequate uteroplacental blood supply is essential for the development and growth of the placenta and fetus during pregnancy. Aberrant uteroplacental perfusion is associated with pregnancy complications such as preeclampsia, fetal growth restriction (FGR), and gestational diabetes. The regulation of uteroplacental blood flow is thus vital to the well-being of the mother and fetus. Ca2+-activated K+ (KCa) channels of small, intermediate, and large conductance participate in setting and regulating the resting membrane potential of vascular smooth muscle cells (VSMCs) and endothelial cells (ECs) and play a critical role in controlling vascular tone and blood pressure. KCa channels are important mediators of estrogen/pregnancy-induced adaptive changes in the uteroplacental circulation. Activation of the channels hyperpolarizes uteroplacental VSMCs/ECs, leading to attenuated vascular tone, blunted vasopressor responses, and increased uteroplacental blood flow. However, the regulation of uteroplacental vascular function by KCa channels is compromised in pregnancy complications. This review intends to provide a comprehensive overview of roles of KCa channels in the regulation of the uteroplacental circulation under physiological and pathophysiological conditions.

1. Introduction

Vascular tone in small arteries/arterioles governs vascular resistance and hence blood perfusion of a given tissue/organ. It is determined by the contractile state of vascular smooth muscle cells (VSMCs), which is regulated by dynamic changes in intracellular Ca2+ concentrations ([Ca2+]i) [1]. Vasoconstriction is initiated by an increase in [Ca2+]i primarily due to Ca2+ influx mediated by the L-type voltage-dependent Ca2+ (CaV1.2) channel in the plasma membrane and/or Ca2+ release mediated by ryanodine (RyR)/inositol trisphosphate (IP3R) receptors in the sarcoplasmic reticulum (SR) membrane. The activity of the CaV1.2 channel in VSMCs is regulated by the membrane potential. Potassium (K+) channels are dominant ion conductive pathways in the vasculature to set/regulate the membrane potential. Their activities in endothelial cells (ECs) and VSMCs participate in regulating Ca2+ homeostasis and vascular tone [2,3]. Membrane hyperpolarization induced by the opening of K+ channels closes the CaV1.2 channel in VSMCs, leading to a fall in [Ca2+]i and subsequent vasodilatation. In contrast, membrane depolarization caused by the closing of K+ channels opens the CaV1.2 channel, resulting in an increase in [Ca2+]i and vasoconstriction. Therefore, the dynamic interplay between CaV1.2 and K+ channels in the vasculature plays a pivotal role in regulating vascular tone. Among various types of K+ channels in ECs and VSMCs, Ca2+-activated K+ (KCa) channels are instrumental in the regulation of vascular tone [2,3,4,5,6,7].
Uteroplacental blood flow increases dramatically during pregnancy. Adequate uteroplacental blood perfusion is essential for the growth/development of the placenta and fetus, as well as the well-being of the mother. Uteroplacental blood flow is inversely proportional to uteroplacental vascular resistance. Increased uteroplacental blood flow in pregnancy is primarily achieved by lowering uteroplacental vascular resistance owing to the structural remodeling of spiral arteries, establishment of the placenta, and vasodilation [8,9]. Adaptive changes in the uteroplacental circulation are impaired in pregnancy complications such as preeclampsia, fetal growth restriction (FGR, also known as intrauterine growth restriction), and gestational diabetes, leading to insufficient perfusion of the placenta [9,10,11]. These disorders are interrelated. For example, early-onset preeclampsia is often associated with FGR, whereas gestational diabetes is a risk factor for preeclampsia [12,13]. Pregnancy complications are associated with maternal and perinatal morbidity and mortality [14,15,16] and predispose the mother and offspring to metabolic and cardiovascular diseases in later life [17,18,19,20]. The first experimental evidence that KCa channels participate in regulating uteroplacental blood flow was presented by Rosenfeld’s group in 2000 [21]. Since then, molecular and functional expression of KCa channels in uteroplacental vessels has received considerable attention. In this review, we highlight current knowledge on the roles of KCa channels in the regulation of uteroplacental circulation in physiological and pathophysiological conditions.

2. Overview of KCa Channels

KCa channels are a large family of K+ channels, which are activated by intracellular Ca2+ and selectively transport K+ ions. KCa channels contain six/seven-transmembrane domains, and are classified into two groups based on their biophysical properties [22]. One group includes the BKCa channel that has large single-channel conductance ranging from 100 to 300 pS [23,24] and is activated by micromolar [Ca2+]i and membrane depolarization [23,25]. The other group comprises small-conductance (SKCa) (KCa2.1–2.3) and intermediate-conductance (IKCa, KCa3.1) KCa channels that are voltage-insensitive and are activated by sub-micromolar [Ca2+]i. The SKCa channel has single-channel conductance of 5–20 pS [26,27], whereas the IKCa channel has unitary conductance of 20–40 pS [28,29].
A functional BKCa channel is composed of a tetramer of α-subunit that is encoded by the KCNMA1 gene. The BKCa channel achieves its functional diversity primarily through the association of α subunits with accessory subunits and other proteins, alternative splicing, and post-translational modifications such as phosphorylation, oxidation, and palmitoylation [30,31,32,33,34,35,36,37]. Each BKCa channel α subunit (125–140 kDa) contains seven transmembrane spanning segments (S0–S6) and a large cytoplasmic COOH-terminus (Figure 1). They form three main structural domains that serve distinct functions [38]. S1-S4 segments constitute the voltage-sensing domain that detects changes in the membrane potential. S5–S6 segments line the pore to control K+ permeation [39,40]. Two tandem RCK (regulator of conductance for K+) domains (RCK1 and RCK2) in the cytoplasmic COOH-terminus from each subunit form a Ca2+ gating ring and function as a Ca2+ sensor [41].
The BKCa channel is ubiquitously distributed among mammalian tissues [39] and usually associates with auxiliary β-subunits (~20 kDa). These accessory proteins are expressed in a cell-specific manner and display unique regulatory effects on the channel. Four distinct β-subunits, β1–4, are encoded by KCNMB1-4 [22]. The β1 subunit is primarily expressed in smooth muscle [42], whereas β2, β3, and β4 subunits are mostly expressed in neurons, chromaffin cells, kidney, heart, liver, and lung, among others [43,44,45]. The β-subunit consists of two transmembrane domains with intracellular N- and C-termini and a long extracellular loop (Figure 1). Up to four β-subunits could co-assemble with pore-forming α subunits [46,47]. Co-assembling with these auxiliary subunits alters the channel’s apparent sensitivity to Ca2+ and voltage as well as kinetic properties [35].
A group of leucine-rich repeat-containing (LRRC) proteins (~35 kDa) are identified as auxiliary γ subunits of the BKCa channel [48]. The expression of LRRC proteins is also tissue-dependent [49]. These LRRC proteins are structurally distinct from the β-subunit. They consist of a large, extracellular domain with six leucine-rich repeat units (LRR1–6), and a single transmembrane segment (Figure 1). In a manner similar to the β subunit, the association of γ subunits to α subunits also alters channel gating properties by increasing voltage sensitivity even in the absence of Ca2+ [35].
SKCa channels are encoded by KCNN1-3, whereas the IKCa channel is encoded by KCNN4. SKCa and IKCa channels share a similar topology to members of the KV channel superfamily and consist of six transmembrane segments (S1–S6) [50] (Figure 1). They are also tetrameric structures. The channel pore is formed by S5 and S6. However, the S4 segment of SKCa and IKCa channels contains fewer charged residues than its counterparts in the KV and BKCa channels, resulting in a lack of voltage dependence. These channels are expressed primarily in neurons and ECs. Although the activities of SKCa and IKCa channels are also controlled by intracellular Ca2+ levels, Ca2+ does not directly bind to channels. Instead, the Ca2+ sensitivity of these channels is achieved through the binding of Ca2+ to calmodulin (CaM) constitutively bound to the C-terminus of the channel [51,52].

3. KCa Channels and Vascular Function

3.1. KCa Channels in VSMCs

The BKCa channel α subunit is abundantly expressed in VSMCs of virtually all vascular beds. BKCa channel accessory β and γ subunits are also found in VSMCs [46,53,54,55]. The predominant β isoform in VSMCs is the β1 subunit [42]. Although β2 and β4 subunits are also present in VSMCs of some vessels, their expression is extremely low [56,57,58]. The association of accessory subunits with α subunits alters channel biophysical properties. Both β1 and γ subunits increase BKCa channel sensitivity to both Ca2+ and voltage in VSMCs [42,53].
SKCa channels are scantily expressed in VSMCs [59,60]. Although an apamin-sensitive K+ conductance has been demonstrated in VSMCs of some vascular beds [61,62,63], its identity has not been resolved. Similarly, evidence for the existence of IKCa channels in VSMCs is limited. The IKCa channel is either not or very poorly expressed in contractile VSMCs [64]. However, its expression in VSMCs is significantly upregulated during proliferation or under pathophysiological conditions such as myocardial infarction, vascular injury, and atherosclerosis [29,64,65,66,67]. Therefore, the IKCa channel likely plays a role in the angiogenesis and pathogenesis of atherosclerosis/restenosis. However, SKCa and IKCa channels are found to express in VSMCs of uterine and placental chorionic plate arteries [68,69].

3.2. KCa Channels in ECs

Endothelial expression of the BKCa channel appears to be erratic [70,71]. Molecular expression of the BKCa channel α subunit and channel activity have been reported in the intact endothelium and in isolated ECs from some blood vessels [72,73,74,75,76,77,78]. However, the BKCa channel is absent in ECs from other vascular beds [79,80,81,82,83,84]. In addition, the BKCa channel β1 subunit is absent in ECs [77,85]. Proliferation and chronic hypoxia trigger BKCa channel expression in ECs [86,87,88]. Of interest, the BKCa channel β4 subunit along with the α subunit is expressed in rat lung microvascular ECs, forming functional BKCa channels [77].
Both IKCa and SKCa channels are abundantly expressed in the endothelium [2,89]. The predominant SKCa and IKCa channels expressed in ECs are KCa2.3 (SK3) and KCa3.1 (IK1) channels, respectively [90,91]. It appears that SKCa and IKCa channels have distinct spatial localizations. Whereas KCa2.3 channels are widely distributed in the EC plasma membrane, KCa3.1 channels are primarily located in myoendothelial gap junctions (MEGJs) [92,93].

3.3. KCa Channels and the Regulation of Vascular Function

3.3.1. Activation of BKCa Channels in VSMCs

Given the large conductance and copious expression of the BKCa channel in VSMCs, small changes in the open probability of the channel have a significant impact on the membrane potential of VSMCs and vascular tone. BKCa channel activation in VSMCs is primarily linked to Ca2+ release events from the SR through RYRs and/or Ca2+ influx through CaV1.2 channels or nonselective cation ion channels (Figure 2) [94,95,96]. A fraction of RYRs in the SR membrane are in close proximity to BKCa channels in the plasma membrane of VSMCs and together they form Ca2+ signaling microdomains [97]. Concerted opening of several RyRs generates Ca2+ sparks and the local [Ca2+]i may reach ~10 μM within these microdomains [97,98,99]. Ca2+ sparks then activate BKCa channels to produce spontaneous transient outward currents (STOCs), which in turn promote membrane hyperpolarization and closure of the CaV1.2 channel. The BKCa channel β1 subunit plays a central role in linking Ca2+ sparks to the BKCa channel. Genetic deletion of the β1 subunit decreases the Ca2+ sensitivity of the BKCa channel, resulting in uncoupling BKCa channels from Ca2+ sparks [100]. In addition, reduced expression of the BKCa channel β1 subunit in type 2 diabetic murine VSMCs leads to abnormal coupling between Ca2+ sparks and the BKCa channel [101]. CaV1.2, BKCa, and transient receptor potential canonical 1 (TRPC1) channels can form complexes in the plasma membrane of VSMCs to provide an efficient mechanism for obtaining localized high Ca2+ concentrations to activate the BKCa channel [102,103,104,105]. Additionally, TRPV4, RyRs, and BKCa channels are also found to form Ca2+ signaling complexes to promote smooth muscle hyperpolarization [95]. Furthermore, the generation of Ca2+ sparks can be indirectly modulated by the CaV1.2 channel. CaV1.2 channel-mediated Ca2+ entry increases luminal SR Ca2+ and hence Ca2+ sparks [106]. Thus, the formation of Ca2+ microdomains/macromolecular complexes provides a rapid feedback and elicits an efficient regulation of Ca2+ signaling in VSMCs.

3.3.2. BKCa Channels and Vascular Tone

VSMCs of small arteries/arterioles possess intrinsic properties to constrict in response to an increase in intralumenal pressure and to dilate following a decrease in intralumenal pressure [107]. An increase in intralumenal pressure depolarizes the plasma membrane leading to the opening of the CaV1.2 channel and vasoconstriction/myogenic tone. However, myogenic vasoconstriction is regulated by a negative feedback mechanism conferred by the BKCa channel [5]. Membrane depolarization promotes Ca2+ sparks in VSMCs. In addition, Ca2+ entry through CaV1.2 and TRPV4 channels also enhances Ca2+ sparks that in turn activate the BKCa channel [96]. Activation of the BKCa channel in VSMCs triggers STOCs and subsequent membrane hyperpolarization, leading to CaV1.2 channel closure and vasodilation [94]. Therefore, the BKCa channel functions as a ‘brake’ to prevent excessive vasoconstriction. The importance of the BKCa channel in the regulation of vascular function has been well demonstrated by pharmacological and genetic manipulations. The blockade of the BKCa channel with iberiotoxin or tetraethylammonium (TEA) induces membrane depolarization, followed by an elevation of [Ca2+]i, vasoconstriction, and elevated blood pressure [42,108,109,110]. Genetic ablation of the BKCa channel α subunit leads to hypertension [111], suggesting an essential role of this channel in regulating blood pressure and controlling blood perfusion to organs. The BKCa channel β1 subunit is also vital in regulating vascular tone. The BKCa channel in VSMCs from β1 null mice has decreased Ca2+ sensitivity and reduced channel activity due to uncoupling the channel from Ca2+ sparks. These changes result in VSMC membrane depolarization and enhancement of vasoconstriction, which ultimately lead to the development of hypertension [42,100,112,113]. Not surprisingly, the expression of the BKCa channel β1 subunit in VSMCs is reduced in hypertension in patients [114] and in animal models [115,116,117]. In contrast, a gain-of-function mutation of the BKCa channel β1 subunit is associated with a low prevalence of hypertension in human studies [118,119,120]. In addition, the expression of the BKCa channel β1 subunit in VSMCs of rat mesenteric arteries is upregulated after hemorrhagic shock [121]. This upregulation enhances Ca2+ sensitivity of the BKCa channel, promotes VSMC membrane hyperpolarization, and reduces vasoconstriction to norepinephrine. Diabetes is also associated with suppressed expression of the BKCa channel β1 subunit in VSMCs [122,123].
The BKCa channel activity is fine-tuned by phosphorylation [37,124]. Many vasoactive agents alter vascular contractility via protein kinase-mediated phosphorylation of the BKCa channel. Endothelin, angiotensin II, 5-hydroxytryptamine, and 20-hydroxyeicosatetraenoic acid elicit vasoconstriction via serine/threonine kinase PKC- and/or tyrosine kinase c-Src-mediated inhibition of the BKCa channel in VSMCs [125,126,127,128,129]. Conversely, β-adrenergic agonists, adenosine, calcitonin gene-related peptide, and nitric oxide (NO) mainly produce vasorelaxation via PKA- or PKG-dependent activation of the BKCa channel in VSMCs [130,131,132,133,134,135,136].
NO can also regulate BKCa channel activity in VSMCs by altering the trafficking of the BKCa channel β1 subunit. NO is found to stimulate rapid surface trafficking of the BKCa channel β1 subunit via cGMP-PKG- and cAMP-PKA-dependent pathways, resulting in increased channel Ca2+ sensitivity/channel activity, and vasodilation [137]. Moreover, NO is able to directly activate the BKCa channel in VSMCs [138,139].

3.3.3. Activation of SKCa and IKCa Channels in ECs

The vascular endothelium plays a key role in regulating vascular tone. Activation of SKCa and IKCa channels is an essential process for endothelium-dependent vasorelaxation conferred by various vasoactive agents [60,81,140,141,142,143]. Endothelium-dependent vasodilators and physical stimuli such as fluid shear stress increase [Ca2+]i in ECs by triggering IP3R-mediated Ca2+ release from SR, store-operated Ca2+ entry, and TRPV4-mediated Ca2+ influx [144]. Ca2+ subsequently binds to calmodulin constitutively bound to SKCa and IKCa channels, resulting in channel conformational changes and channel activation [145].

3.3.4. SKCa and IKCa Channels and Vascular Tone

Opening endothelial SKCa and IKCa channels induces hyperpolarization, which could be transmitted to adjacent VSMCs via MEGJ, leading to hyperpolarization of VSMCs, closure of the CaV1.2 channel, and subsequent vasodilation (Figure 2) [2,146,147,148]. In addition, K+ ion accumulated in the extracellular space between ECs and VSMCs due to activation of endothelial SKCa and IKCa channels is proposed to cause hyperpolarization and relaxation of the VSMCs through activating the inwardly-rectifying K+ (Kir) channel and/or the Na+-K+-ATPase [149,150]. Furthermore, both SKCa and IKCa channels also participate in regulating NO synthesis and release from ECs [151,152,153]. The blockade of the SKCa channel with apamin and of the IKCa channel with charybdotoxin or triarylmethane-34 (TRAM-34) attenuates NO production in ECs [151,152]. Activation of endothelial SKCa and IKCa channels also promotes the release of endothelium-derived hyperpolarizing factor (EDHF) [154]. Depending on the size of the vessels, different mechanisms may be involved in the actions of SKCa and IKCa channels. Activating endothelial SKCa and IKCa channels causes vasorelaxation mainly via the release of NO in large arteries and EDHFs in small arteries, respectively [155,156]. NO and EDHFs released from ECs subsequently trigger BKCa channel activation in VSMCs, leading to vasorelaxation [139,157,158,159]. Pharmacologic blockade or genetic ablation of SKCa and/or IKCa channels depolarizes ECs and decreases vasoactive agent-evoked hyperpolarization of ECs and VSMCs, resulting in impaired vasorelaxation and reduced blood flow [59,151,152,160,161,162,163,164]. Conversely, SKCa and IKCa channel activation decreases vascular tone/blood pressure and increases blood flow [153,163,165,166,167]. The functional importance of SKCa and IKCa channels is furthermore supported by observations that deletion of either or both SKCa and IKCa genes is associated with the development of hypertension [59,164,168]. Consistent with these findings, the expression of SKCa2.3 and/or IKCa channels was reduced in mesenteric arteries from spontaneously or ANG II-induced hypertensive rats [169,170]. However, the IKCa channel is upregulated under certain pathophysiological conditions such as myocardial infarction, and atherosclerosis [64,171,172,173]. In addition, the expression of SKCa2.3 and IKCa channels is differently altered by chronic hypoxia in pulmonary arteries. Exposure to chronic hypoxia causes upregulation of the SKCa2.3 channel, but downregulation of the IKCa channel [174].

4. Adaptation/Maladaptation of the Uteroplacental Circulation in Normal Pregnancy and Pregnancy Complications

In a nonpregnant state, blood flow to the uterus is relatively low. For example, uterine blood flow is ~20–50 mL/min in nonpregnant humans and sheep, corresponding to 1–3% of the maternal cardiac output [175,176,177,178]. Uteroplacental blood flow increases dramatically during pregnancy, rising to 600 to 1000 mL/min at 36 to 38 weeks in human pregnancy [179,180] and >1000 mL/min in late sheep pregnancy [175,181,182,183]. Similarly, uteroplacental blood flow increases by 10- to 30-fold in near-term pregnant rats and guinea pigs [184,185,186]. Uteroplacental blood flow comprises ~20% of maternal cardiac output at term [179,186,187]. It is estimated that 80% to 90% of total uteroplacental blood flow perfuses the placenta at term and the remaining supplies the myometrium [175,188,189], providing sufficient nutrient and oxygen supply for the growth of the placenta and fetus. The hemodynamic changes in the uteroplacental circulation during pregnancy are primarily achieved by uterine vascular remodeling, reduced uteroplacental vascular resistance, and the formation of the placenta [8,9,190,191,192]. Notably, a variety of functional changes contribute to the adaptation. Myogenic tone is markedly attenuated in the uterine arteries of pregnant sheep [193]. Vasopressor response of uterine arteries to various vasoconstrictors such as α-adrenergic agonists, 5-hydroxytryptamine, endothelin 1, angiotensin II, and thromboxane is attenuated during pregnancy in humans and other species [194,195,196,197,198,199,200,201,202]. Moreover, the production of vasodilators including NO and EDHF in uterine arteries increases during pregnancy [203,204]. NO- and endothelium-dependent vasodilation in uterine arteries is also enhanced during pregnancy [203,204,205,206,207,208].
The adaptation of the uteroplacental circulation is compromised in preeclampsia, FGR, and gestational diabetes. Preeclampsia is associated with increased uteroplacental vascular resistance [209,210,211]. Uterine arteries from preeclamptic women and animal models of preeclampsia display enhanced vasoconstriction and blunted vasodilation to vasoactive agents [212,213,214,215,216,217,218]. In addition, shear stress-mediated NO release from uterine arterial endothelium is impaired in preeclampsia [219]. EDHF-mediated vasorelaxation of myometrial arteries is reduced in preeclampsia [214,220]. In a rat model of preeclampsia produced by reduced uterine perfusion pressure (RUPP) in pregnant animals, uterine arteries exhibit increased myogenic tone and decreased endothelium-dependent vasorelaxation [221]. Additionally, the refractoriness to angiotensin II in uterine arteries is lost in gestational hypertension [222,223]. Uteroplacental vascular resistance is increased in a mouse model of gestational diabetes [224]. Endothelium-dependent vasorelaxation is impaired in the myometrial arteries of women with diabetes [225]. As expected, uteroplacental blood flow is reduced in preeclampsia, FGR, and gestational diabetes [210,226,227,228,229].

5. KCa Channels and the Uteroplacental Circulation in Normal Pregnancy

5.1. KCa Channels in Uteroplacental Vasculature

Both real-time polymerase chain reaction (RT-PCR) and Western blot reveal the expression of BKCa channel α, β1, and β2 subunits in the uterine arteries of humans and sheep [21,57,230,231,232,233,234,235]. The β1 subunit is the predominant β isoform in uterine arteries, and the expression level of the β2 subunit is low. Immunohistochemistry further reveals that these BKCa channel subunits are located in VSMCs, but not in the endothelium, of uterine arteries [57,230,231,232]. The BKCa channel in VSMCs of uterine arteries is activated by an increase in [Ca2+]i, and has unitary conductance of 100–200 pS [21,236]. The BKCa channel γ subunit is also detected in both human and mouse uterine arteries [55,236]. SKCa and IKCa channels are also expressed in uterine arteries [68,237]. IKCa channel mRNA is detected in cultured human uterine microvascular ECs [238]. Both SKCa and IKCa channels have been visualized in the endothelium of human and sheep uterine arteries with immunohistochemistry [68,239]. Of interest, KCa2.2 and KCa2.3 channels are present in VSMCs of sheep uterine arteries [68]. BKCa, IKCa, and KCa2.3 channels are also detected in VSMCs and/or ECs of placental chorionic plate arteries of pregnant women [240,241].

5.2. KCa Channels in the Adaptation of the Uteroplacental Circulation in Normal Pregnancy

5.2.1. Estrogen as a Key Determinant of KCa Channel Upregulation

The expression of KCa channels in uteroplacental vessels is under the influence of estrogen during the ovarian cycle and pregnancy. Khan et al. demonstrate that the BKCa channel α subunit protein in ovine uterine arteries remains constant during both follicular and luteal phases of the ovarian cycle [232]. The protein level of the BKCa channel β1 subunit is higher in uterine arteries from follicular phase ewes than in vessels from luteal phase animals. Similarly, protein abundance of the BKCa channel α subunit in uterine arteries is negligibly affected by gestation, whereas the expression of the BKCa channel β1 subunit is upregulated in uterine arteries from pregnant sheep [57,233]. The upregulation of the BKCa channel β1 subunit expression in uterine arteries during the follicular phase of the ovarian cycle and during pregnancy is paralleling with elevated plasma estrogen levels [57,232]. Remarkably, prolonged treatment of nonpregnant sheep or isolated uterine arteries from nonpregnant animals with 17β-estradiol increases the BKCa channel β1 subunit expression in the uterine vasculature, resembling those changes that occurred during the ovarian cycle and gestation [230,233,235]. Similarly, estrogen treatment and pregnancy also increase BKCa channel β1 subunit expression in rat uterus [242]. These observations implicate estrogen as an initiator for the upregulation of BKCa channel expression in the uterus and its vascular beds in pregnancy. The expression of the BKCa channel β2 subunit in uterine arteries remains low and unchanged during pregnancy [57]. The increased expression of the BKCa channel β1 subunit alters channel stoichiometry and increases Ca2+ sensitivity. In addition, pregnancy and prolonged treatment of nonpregnant sheep with 17β-estradiol also upregulate the expression of NOS, PKG-1α, and cGMP in uterine arteries [57,230,232,243,244]. The upregulation of the NO-cGMP-cPKG pathway could stimulate the BKCa channel through phosphorylation [245]. The enhanced BKCa channel activity subsequently contributes to reduced uterine vascular resistance [233].
Pregnancy also upregulates SKCa channel expression in uterine arteries [68]. This upregulation is also simulated by ex vivo estrogen treatment of isolated uterine arteries from nonpregnant sheep. The expression of KCa2.3 and IKCa channels in the aorta is increased in pregnant mice [246]. Similarly, estrogen replacement in ovariectomized rats increases the KCa2.3 channel expression in the uterus and nonvascular smooth muscle [247,248]. In contrast, ovariectomy reduces KCa2.3 channel activity and endothelium-dependent vasorelaxation in mouse mesenteric arteries [249]. Likewise, incubating human uterine microvascular ECs with high concentrations of estrogen or serum from normal pregnant women promotes SKCa2.3 and IKCa channel expression [246]. Moreover, the treatment with serum from normal pregnant women increases plasma membrane abundance of SKCa2.3 and IKCa channels in human uterine microvascular ECs [250]. As expected, estrogen replacement in ovariectomized rats enhances EDHF-mediated vasodilation of uterine arteries [251]. However, estrogen replacement in ovariectomized mice reduces KCa2.3 channel expression in the uterus [252].

5.2.2. Mechanisms Underlying Estrogen-Mediated KCa Channel Upregulation

Estrogen usually regulates gene expression via interacting with its classical receptors, ERα and ERβ. The binding of estrogen results in conformational changes of estrogen receptors, allowing these receptors to interact with estrogen response elements (EREs) in the promoter region of target genes to regulate transcription [253]. However, examination of the cloned ovine KCNMB1 promoter sequences reveals that this promoter contains no EREs [235]. Instead, ERα interacts with Sp1 and binds to Sp1 binding sites to regulate KCNMB1 expression in ovine uterine arteries. Several putative transcription factor binding sites, containing CpG dinucleotides in or near their core binding sequences, have been identified in ovine KCNMB1 promoter, including Sp1 at −380 and AP1 at −652, −879, and −1202. Among these sites, the Sp1-380 binding element is essential for ovine KCNMB1 gene expression as deletion of this site significantly decreases the KCNMB1 promoter activity [235]. The importance of Sp1 in the regulation of expression of KCNMB1 is also demonstrated in nonvascular smooth muscle. Overexpression of Sp1 in smooth muscle cells of rabbit sphincter of Oddi enhances KCNMB1 promoter activity [254].
DNA methylation, the covalent addition of a methyl group (-CH3) to the base cytosine in the dinucleotide 5′-CpG-3′ catalyzed by DNA methyltransferases (DNMTs), is an important epigenetic mechanism controlling gene expression [255]. DNA methylation is usually associated with gene repression. CpG dinucleotides of the Sp1 binding site at the KCNMB1 gene promoter are highly methylated in the uterine arteries of nonpregnant sheep, resulting in low transcription factor binding and KCNMB1 promoter activity. Ten-eleven translocation methylcytosine dioxygenases (TETs) catalyze the conversion of 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC) in active DNA demethylation. Pregnancy via estrogen upregulates TET1 which in turn decreases CpG methylation at the Sp1 binding site and facilitates Sp1/ERα binding to the Sp1 binding site of KCNMB1, leading to the upregulation of the BKCa channel β1 subunit in uterine arteries [235,256] (Figure 3).
The increased SKCa channel expression in uterine arteries during pregnancy is also mediated by estrogen [68]. Estrogen regulates SKCa2.3 gene (KCNN3) expression through interactions between ERα and Sp1 in Cos7 and L6 cells [257]. Moreover, estrogen treatment stimulates the expression of the SKCa2.3 transcript in human myometrial cells overexpressing Sp1 [252]. These observations suggest an important role of Sp1 in the expression of the KCNN3 gene.
Vascular endothelial growth factor (VEGF) appears to play role in the pregnancy-induced upregulation of SKCa2.3 and IKCa channels. The upregulation of SKCa2.3 and IKCa channels induced by exposure to serum from normal pregnant women in cultured human uterine microvascular ECs is diminished by blocking VEGF receptors [246]. Serum from normal pregnant women and VEGF increases H2O2 generation and promote SKCa2.3 and IKCa channel expression via the H2O2/FYN/ERK pathway [246]. VEGF receptor activation also causes the downregulation of caveolin-1 and subsequently inhibits the internalization of SKCa2.3 and IKCa channels, leading to their high abundance in the plasma membrane in uterine vascular ECs in pregnancy [250]. It should be noted that placental VEGF expression is also subject to regulation by estrogen in pregnancy [258].

5.2.3. KCa Channels and the Adaptation of the Uteroplacental Circulation

Findings from in vivo and in vitro studies exploring the functional roles of KCa channels in the uterine circulation of nonpregnant sheep are quite intriguing. Despite the expression of the BKCa channel in uterine arteries of nonpregnant animals, stimulation of the BKCa channel with NS 1619 fails to promote vasorelaxation of these vessels [68,259]. In addition, the blockade of the BKCa channel with TEA also does not alter the myogenic tone of uterine arteries [233]. Moreover, basal uterine blood flow in nonpregnant sheep is negligibly altered by local infusion of TEA [21]. These findings suggest that the BKCa channel in the uterine arteries of nonpregnant sheep is quiescent and contributes minimally to the regulation of uterine vascular tone, vascular reactivity, and basal uterine blood flow. Interestingly, pregnancy ‘awakes’ the BKCa channel and the channel becomes active in ovine uterine arteries. Activation of the BKCa channel promotes vasorelaxation of uterine arteries from pregnant sheep [68,259], whereas inhibition of the BKCa channel increases the myogenic tone of uterine arteries [233]. Moreover, local infusion of TEA into uterine arteries decreases basal uterine blood flow by ~50% in pregnant sheep [182,260].
It is currently unknown why the BKCa channel is dormant in the uterine arteries of nonpregnant sheep. One possible explanation is the low abundance of the channel in uterine arteries. The other scenario is that the majority of the BKCa channel β1 subunit in uterine arteries of nonpregnant sheep are in the cytoplasm and do not form complexes with the α subunit at the surface membrane as observed in rat mesenteric and human cerebral arteries [137]. Leo et al. [137] demonstrate that NO stimulates rapid trafficking of the BKCa channel β1 subunit to the plasma membrane via a PKG-dependent pathway. Pregnancy is accompanied by parallel increases in NO, cGMP, protein kinase G-1α and the BKCa channel β1 subunit in uterine arteries [57,203,233]. We recently demonstrated that pregnancy increases the association of α and β1 subunits in uterine arteries [261]. The association of BKCa channel β1 and α subunits has been shown to increase channel activity by enhancing the channel’s Ca2+ sensitivity [42]. It is reasonable to speculate that the enhanced NO-PKG pathway in uterine arteries could stimulate the trafficking of the BKCa channel β1 subunit to the plasma membrane of VSMCs in addition to increased BKCa channel β1 subunit expression, thus facilitating the transition from the dormant BKCa channel in the nonpregnant state to the active channel in pregnancy.
BKCa channel activity is subject to modulation by protein kinases [37,124]. Activation of protein kinase C inhibits the BKCa channel in uterine arteries [233,262]. Thus, vasoconstriction induced by α-adrenergic ligands and thromboxane may involve PKC-mediated inhibition of the BKCa channel in this vessel [263,264]. Notably, PKC activity in uterine arteries is suppressed in pregnancy [193,264,265]. On the other hand, the production of vasodilators such as NO, calcitonin gene-related peptide, and adrenomedullin is increased in pregnancy and they produce vasorelaxation of uterine arteries apparently via cGMP-mediated activation of the BKCa channel [231,266,267]. Inhibition of the BKCa channel enhances uterine vasoconstriction induced by α-adrenergic ligands, thromboxane, and PKC activator in intact sheep and in isolated vessels [231,262,268,269]. Therefore, activation of the BKCa channel could offset vasoconstriction and prevents vasospasm of uterine arteries, which probably contributes to the refractoriness of uterine arteries to vasoconstrictors during normal pregnancy.
In VSMCs, the BKCa channel is primarily activated by Ca2+ sparks mediated by RyRs [270]. Activated BKCa channels then mediate K+ efflux in the form of STOCs, leading to membrane hyperpolarization, CaV1.2 channel closure, and vasorelaxation. We recently demonstrated that pregnancy-induced decreases in the myogenic tone of uterine arteries also involve the upregulation of RyR expression/function and enhanced Ca2+ sparks [271]. Moreover, pregnancy promotes the colocalization of RyR1/2 and the BKCa channel β1 subunit, leading to enhanced Ca2+ spark-STOC coupling [261]. The increased Ca2+ spark-STOC coupling then boosts STOCs, resulting in reduced uterine arterial myogenic tone in pregnancy [261,271].
NO and hydrogen sulfide (H2S) are recognized as important regulators of vascular function. Pregnancy increases NO and H2S production in both human and sheep uterine arteries, which contributes to estrogen-induced uterine vasodilation in pregnancy [244,272,273,274]. NO is a potent stimulator of the BKCa channel in VSMCs [139]. It is expected that NO also triggers BKCa activation in uterine arteries to promote vasodilation in pregnancy as there is a parallel increase in both the production of NO and cGMP and expression of the BKCa channel in uterine arteries during pregnancy [57,203,233]. A recent study reveals that H2S elicits vasodilation of uterine arteries via activating the BKCa channel [236].
EDHF plays an important role in regulating uterine vascular contractility during pregnancy [220,275]. Endothelial SKCa2.3 and IKCa channels mediate endothelial membrane hyperpolarization and participate in EDHF-mediated vasodilator response [148,276]. Pregnancy significantly potentiates EDHF-mediated vasodilation of uterine arteries [204,277]. For example, EDHF contributes to ~30% of endothelium-dependent vasorelaxation of uterine arteries in nonpregnant rats and this fraction increases to ~70% in pregnant animals [277]. A combination of apamin plus charybdotoxin or TRAM 34, but not of apamin plus the BKCa channel blocker iberiotoxin, abolished the EDHF-mediated dilation of human and rat uterine arteries, suggesting that SKCa and IKCa channels are major mediators of EDHF responses in uterine arteries [204,275,278]. MEGJs provide direct contact between the ECs and VSMCs. MEGJs are the primary pathway of EDHF-mediated relaxation of myometrial arteries in pregnancy [148]. The SKCa channel may also mediate NO-induced relaxation of uterine arteries [279]. In addition, the SKCa channel in uterine VSMCs participates in regulating the myogenic tone of uterine arteries [68].
The SKCa2.3 and IKCa channels also participate in uteroplacental angiogenesis and vascular remodeling during pregnancy. Inhibiting SKCa2.3 and IKCa channels in HUVECs with apamin and TRAM 34, respectively, inhibits the secretion of angiogenic factors, proliferation/migration, and tube formation [280]. On the other hand, overexpression of the SKCa2.3 channel increases the diameter of uterine arteries [281]. Similarly, SKCa2.3 channel overexpression also increases the ratio of VEGF to sFlt-1 and vessel size/numbers in the placenta [282].

6. KCa Channels and Uteroplacental Circulation in Pregnancy Complications

6.1. Aberrant Expression/Function of Uteroplacental Vascular KCa in Pregnancy Complications

The expression of the BKCa channel β1 subunit is repressed in human placental chorionic plate arteries in preeclampsia, which is associated with impaired NO-induced vasodilation [69]. In addition, preeclampsia also reduces the expression of the BKCa channel β1 subunit in umbilical vein ECs [283]. In a sheep model of preeclampsia, it is found that high-altitude acclimatization downregulates the BKCa channel β1 subunit in uterine arteries leading to increased uterine vascular tone [234,284]. The expression of the BKCa channel β1 subunit is also downregulated in the uterine arteries of a mouse model of preeclampsia induced by electrical stimulation, leading to increased uteroplacental vascular resistance [285].
Both SKCa and IKCa channels are downregulated in human placental chorionic plate arteries in preeclampsia [241]. The IKCa channel is also downregulated in ECs of the umbilical artery and vein from preeclamptic pregnancy [238,283]. The contribution of MEGJs to EDHF-induced relaxation of myometrial arteries is diminished in preeclampsia [214]. Treating cultured HUVECs with plasma from preeclamptic women mimics the impacts of preeclampsia on IKCa channel expression [238]. An increase in circulating testosterone level is an important risk factor for preeclampsia [286,287,288]. In a rat model of preeclampsia/FGR, elevated levels of plasma testosterone result in FGR [237]. Uterine arteries from pregnant rats chronically treated with testosterone display augmented vasoconstriction to thromboxane, phenylephrine, and angiotensin II. In addition, the prolonged testosterone treatment also downregulates the SKCa2.3 channel in uterine arteries, leading to diminished EDHF-mediated relaxation [237]. In pregnant guinea pigs, chronic hypoxia attenuates EDHF-mediated relaxation of uterine arteries [289], possibly due to impaired SKCa/IKCa channel expression/function.
Gestational diabetes is associated with the downregulation of both BKCa channel α and β1 subunits in human umbilical arterial smooth muscle cells [290]. Using a rat model in which gestational diabetes is induced by the injection of streptozotocin during pregnancy, Gokina’s group demonstrates that EDHF-induced uteroplacental vasodilation is impaired owing to reduced basal and agonist-stimulated [Ca2+]i in ECs [291]. Moreover, they also provide evidence that diabetes selectively causes dysfunction of the IKCa channel in uteroplacental arteries, which attributes to the impaired EDHF response [292,293]. Likewise, EDHF-induced vasorelaxation is reduced in uterine arteries of streptozotocin-treated pregnant mice [294].

6.2. Mechanisms Underlying the Dysregulation of KCa Channels in the Uteroplacental Circulation

6.2.1. Hypoxia and HIFs

Hypoxia during gestation is a major insult to maternal cardiovascular homeostasis and complicates adaptive changes in the uteroplacental circulation [295,296]. HIFs play a crucial role in cellular (mal)adaptation in response to hypoxia. Levels of HIF-1α increase in preeclamptic placentas, in placentas from human high-altitude pregnancy, in uterine arteries of high-altitude acclimatized pregnant sheep, and in placentas of a hypoxic rodent model of preeclampsia [297,298,299,300]. There are complex interplays among HIFs, ROS/endoplasmic reticulum (ER) stress, and epigenetic regulation [296]. For example, HIF-1α is stabilized by mitochondrial ROS [301], whereas HIF-1α through miR-210-induced downregulation of ISCU promotes mitochondrial ROS production [302]. Moreover, DNMT expression is upregulated by HIF-1α [303]. These factors can act alone and in concert to contribute to the pathogenesis of preeclampsia.
Gestational hypoxia attenuates the pregnancy-induced rise in uteroplacental blood flow, leading to increased incidence of preeclampsia and IUGR [299,304,305,306,307]. KCa channels in vascular beds are major targets of hypoxia [37,308]. Gestational hypoxia directly downregulates the BKCa channel β1 subunit and suppresses the upregulation of the BKCa channel β1 subunit and SKCa channels in ovine uterine arteries during pregnancy [68,234]. The attenuated expression of KCa channels culminates in decreased channel activities, leading to increased myogenic tone and diminished KCa channel-mediated vasorelaxation.

6.2.2. Epigenetic Regulation

MicroRNAs (miRs) are non-coding RNAs and play important roles in regulating gene expression. miRs regulate gene expression by interacting with the 3′-untranslated region (3′-UTR) of target mRNAs to induce mRNA degradation and translational repression [309]. Circulating and uteroplacental levels of miR-210, a target of HIF-1α, are increased in preeclampsia, in high-altitude pregnancy, and in a high-altitude hypoxic sheep model of preeclampsia [284,310,311,312,313]. KCNMB1 and RYR2 each contain a miR-210 complementary binding site in their 3′-UTRs and both of them are targets of miR-210 [313]. Indeed, gestational hypoxia via miR-210-mediated downregulation of RyR2 and BKCa channel β1 subunit disrupts the Ca2+ spark-STOC coupling in uterine arteries and hence increases uterine arterial myogenic tone [313].
The dynamic of DNA methylation and demethylation is also an important epigenetic mechanism to fine-tune gene expression. DNA methylation catalyzed by a family of DNMTs transfers a methyl group from S-adenyl methionine to the cytosine residue in a CpG dinucleotide(s) to form 5-methylcytosine (5mC). In general, methylation in the promoter regions of genes is associated with the repression of transcription [314]. On the other hand, active DNA demethylation is initiated by TETs which mediate the oxidation of 5mC to 5-hydroxymethylcytosine (5hmC), thus reviving gene transcription [315]. Gestational hypoxia is found to upregulate DNMT3b in uterine arteries, hence enhancing DNA methylation [316] (Figure 3). TET1 is also a target of miR-210 and gestational hypoxia via miR-210 triggers the downregulation of TET1 in uterine arteries [284,317]. TET1 deficiency nullifies pregnancy-induced DNA demethylation [235,284,317]. Overall, these changes lead to hypermethylation of KCNMB1, downregulation of the BKCa channel β1 subunit in uterine arteries, and increased myogenic tone [284,316,317]. Gestational hypoxia also suppresses the expression of ERα in uterine arteries through hypermethylating the Erα-encoding gene ESR1, which could in turn impairs pregnancy- and estrogen-induced BKCa channel β1 subunit upregulation [318,319,320].

6.2.3. Oxidative/ER Stress

Pregnancy complications are in a state of exaggerated oxidative stress [321]. Reactive oxygen species (ROS) have been implicated in the pathogenesis of various cardiovascular disorders. Mitochondria and NADPH oxidases (NOX) are major sources of ROS in the vasculature [322]. Preeclampsia and gestational hypoxia are found to increase the expression/activity of NOX2 and ROS in the uterine arteries of pregnant sheep and HUVECs [283,300]. Mitochondrial ROS are increased in the placenta of a rat model of preeclampsia produced by reduced uterine perfusion pressure [323]. Likewise, gestational hypoxia also increases mitochondrial ROS via miR-210-mediated downregulation of ISCU and subsequent perturbation of mitochondrial respiration in uterine arteries [324]. ROS could exert its impacts on KCa channels directly or indirectly. Cys911 oxidation in the BKCa channel α subunit decreases Ca2+ sensitivity and impairs channel function [325]. Acute inhibition of ROS with apocynin (a NOX inhibitor) or N-acetylcysteine/EUK-134 (antioxidants) increases BKCa channel activity in uterine arterial VSMCs of pregnant sheep experiencing gestational hypoxia [259,300,326], suggesting that the BKCa channel in uterine arteries is tonically inhibited by ROS under hypoxia. Moreover, antioxidant treatment with N-acetylcysteine in ex vivo studies restores the capacity of estrogen to stimulate molecular and functional expression of the BKCa channel β1 subunit [259,326]. These findings suggest that gestational hypoxia-induced oxidative stress also impairs BKCa channel function by suppressing estrogen-induced KCNMB1 expression in uterine arteries. The Ca2+ spark-STOC coupling is disrupted by mitochondrial ROS, leading to increased myogenic tone. ROS derived from NOX2 also repress the expression of the BKCa channel β1 subunit in HUVECs from preeclamptic pregnancy [283]. Impaired uteroplacental perfusion in mice with gestational diabetes is associated with elevated oxidative stress in uterine arteries [224]. Although the impact of ROS on BKCa channel expression/function is not examined in uteroplacental VSMCs of gestational diabetes, NOX-derived ROS have been shown to mediate the downregulation of the BKCa channel β1 subunit in VSMCs of other vascular beds in diabetic mice [327].
The expression of the SKCa channel is downregulated by NOX2-derived ROS in umbilical vessels and HUVECs from preeclamptic pregnancy [238,283]. This downregulation is imitated by treating HUVECs with serum from women with preeclampsia, oxidized low-density lipoprotein, palmitic acid, and the superoxide donor xanthine/xanthine oxidase mixture [238,328]. Similarly, exogenous H2O2 suppresses the expression of IKCa and/or SKCa channels in cultured HUVECs [329]. In human uterine microvascular ECs, NOX4-derived superoxide mediates the downregulation of KCa2.3 and KCa3.1 channels induced by serum from preeclamptic women [246]. In addition, NOX4-derived ROS also promote the internalization of KCa2.3 and KCa3.1 channels by increasing the association of these channels with caveolin-1, clathrin, and Rab5c in human uterine microvascular ECs [250]. Testosterone suppresses mitochondrial respiration in uteroplacental and vascular cells [330,331]. Thus, the downregulation of the SKCa channel in uterine arteries of pregnant rats chronically treated with testosterone is probably mediated by mitochondrial ROS [237]. Chronic administration of Mito-Tempo in diabetic mice also normalizes the impaired SKCa activity in heart ECs [332].
Endoplasmic reticulum (ER) stress occurs when ER homeostasis is perturbed. Placentas from preeclamptic pregnancy, FGR, and diabetic pregnancy undergo ER stress [333,334,335,336]. Gestational hypoxia also triggers ER stress and activates unfolded protein response (UPR) in the human placenta and in ovine uterine arteries [337,338]. The ER stress inhibitor tauroursodeoxycholic acid and PERK inhibitor GSK2606414 relieve hypoxia-mediated suppression of Ca2+ sparks/STOCs and decrease myogenic tone in uterine arteries [337]. ER stress is found to cause downregulation of the BKCa channel β1 subunit and suppression of BKCa channel activity in VSMCs [339]. Similarly, SKCa2.3 and IKCa channel activities are also suppressed by ER stress in ECs [340]. Thus, ER stress also contributes to the maladaptation of the uteroplacental circulation by impairing KCa expression/function in pregnancy complications.

6.2.4. PKC

Preeclamptic serum increases PKC signaling in cultured HUVECs [341,342]. Gestational hypoxia upregulates PKC in the uterine arteries of pregnant sheep [343]. Activation of PKC inhibits BKCa channel activity and increases myogenic tone in the uterine arteries of pregnant sheep [233]. This mechanism also contributes to gestational hypoxia-induced suppression of SKCa channel activity [262]. Peroxisome proliferator-activated receptor-γ (PPARγ), a ligand-activated transcription factor, has been implicated in the pathogenesis of preeclampsia [344]. Mesenteric arteries from transgenic mice expressing dominant-negative mutant PPARγ displays increased myogenic tone, due to PKC-mediated inhibition of the BKCa channel in VSMCs [345]. Similarly, chronic inhibition of PPARγ during rat pregnancy attenuates uterine vasodilation and causes FGR [346]. Moreover, elevated expression of PKCβ in diabetic mouse aortas promotes the BKCa channel β1 subunit downregulation by impairing AKT signaling [327].

7. Conclusions

Uteroplacental blood flow increases markedly in pregnancy to meet the demand for placental and fetal growth. Uteroplacental vessels undergo extensive structural and functional changes to accommodate increased uteroplacental perfusion. However, these adaptative changes are impaired in pregnancy complications. Precise mechanisms underlying the adaptation/maladaptation of the uteroplacental circulation are not completely understood. Findings over the past twenty years have suggested important roles of KCa channels in the regulation of the uteroplacental circulation under physiological and pathophysiological conditions. Notably, estrogen plays a central role in upregulating KCa channel expression/function leading to reduced uterine vascular tone in normal pregnancy. Lines of evidence suggest that multiple mechanisms including HIFs/miR-210, oxidative stress/ER stress, and PKC contribute to KCa channel dysfunction in uteroplacental vessels, resulting in the maladaptation of the uteroplacental circulation in pregnancy complications. Thus, restoring KCa channel expression/function by targeting HIFs/miR-210, oxidative stress/ER stress, and PKC may offer avenues for the development of therapeutics for pregnancy complications.

Author Contributions

Conceptualization, X.-Q.H. and L.Z.; writing—original draft preparation, X.-Q.H.; writing—review and editing, X.-Q.H. and L.Z. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by National Institutes of Health Grants HD083132 (L.Z.) and HL149608 (L.Z.).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are contained within the article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Ottolini, M.; Hong, K.; Sonkusare, S.K. Calcium signals that determine vascular resistance. Wiley Interdiscip. Rev. Syst. Biol. Med. 2019, 11, e1448. [Google Scholar] [CrossRef] [PubMed]
  2. Feletou, M. Endothelium-Dependent Hyperpolarization and Endothelial Dysfunction. J. Cardiovasc. Pharmacol. 2016, 67, 373–387. [Google Scholar] [CrossRef] [PubMed]
  3. Tykocki, N.R.; Boerman, E.M.; Jackson, W.F. Smooth Muscle Ion Channels and Regulation of Vascular Tone in Resistance Arteries and Arterioles. Compr. Physiol. 2017, 7, 485–581. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Nelson, M.T.; Quayle, J.M. Physiological roles and properties of potassium channels in arterial smooth muscle. Am. J. Physiol. 1995, 268, C799–C822. [Google Scholar] [CrossRef] [Green Version]
  5. Hill, M.A.; Yang, Y.; Ella, S.R.; Davis, M.J.; Braun, A.P. Large conductance, Ca2+-activated K+ channels (BKCa) and arteriolar myogenic signaling. FEBS Lett. 2010, 584, 2033–2042. [Google Scholar] [CrossRef] [Green Version]
  6. Jackson, W.F. Ion channels and the regulation of myogenic tone in peripheral arterioles. Curr. Top. Membr. 2020, 85, 19–58. [Google Scholar] [CrossRef]
  7. Jackson, W.F. Calcium-Dependent Ion Channels and the Regulation of Arteriolar Myogenic Tone. Front. Physiol. 2021, 12, 770450. [Google Scholar] [CrossRef] [PubMed]
  8. Brosens, I.; Puttemans, P.; Benagiano, G. Placental bed research: I. The placental bed: From spiral arteries remodeling to the great obstetrical syndromes. Am. J. Obstet. Gynecol. 2019, 221, 437–456. [Google Scholar] [CrossRef]
  9. Hu, X.; Zhang, L. Uteroplacental Circulation in Normal Pregnancy and Preeclampsia: Functional Adaptation and Maladaptation. Int. J. Mol. Sci. 2021, 22, 8622. [Google Scholar] [CrossRef]
  10. Staff, A.C.; Fjeldstad, H.E.; Fosheim, I.K.; Moe, K.; Turowski, G.; Johnsen, G.M.; Alnaes-Katjavivi, P.; Sugulle, M. Failure of physiological transformation and spiral artery atherosis: Their roles in preeclampsia. Am. J. Obstet. Gynecol. 2022, 226, S895–S906. [Google Scholar] [CrossRef]
  11. Ehlers, E.; Talton, O.O.; Schust, D.J.; Schulz, L.C. Placental structural abnormalities in gestational diabetes and when they develop: A scoping review. Placenta 2021, 116, 58–66. [Google Scholar] [CrossRef]
  12. Huppertz, B. The Critical Role of Abnormal Trophoblast Development in the Etiology of Preeclampsia. Curr. Pharm. Biotechnol. 2018, 19, 771–780. [Google Scholar] [CrossRef] [PubMed]
  13. Yang, Y.; Wu, N. Gestational Diabetes Mellitus and Preeclampsia: Correlation and Influencing Factors. Front. Cardiovasc. Med. 2022, 9, 831297. [Google Scholar] [CrossRef] [PubMed]
  14. Johns, E.C.; Denison, F.C.; Norman, J.E.; Reynolds, R.M. Gestational Diabetes Mellitus: Mechanisms, Treatment, and Complications. Trends Endocrinol. Metab. 2018, 29, 743–754. [Google Scholar] [CrossRef] [PubMed]
  15. Rana, S.; Lemoine, E.; Granger, J.P.; Karumanchi, S.A. Preeclampsia: Pathophysiology, Challenges, and Perspectives. Circ. Res. 2019, 124, 1094–1112. [Google Scholar] [CrossRef] [PubMed]
  16. Egan, A.M.; Dow, M.L.; Vella, A. A Review of the Pathophysiology and Management of Diabetes in Pregnancy. Mayo Clin. Proc. 2020, 95, 2734–2746. [Google Scholar] [CrossRef]
  17. Tooher, J.; Thornton, C.; Makris, A.; Ogle, R.; Korda, A.; Hennessy, A. All Hypertensive Disorders of Pregnancy Increase the Risk of Future Cardiovascular Disease. Hypertension 2017, 70, 798–803. [Google Scholar] [CrossRef] [PubMed]
  18. Andraweera, P.H.; Gatford, K.L.; Care, A.S.; Bianco-Miotto, T.; Lassi, Z.S.; Dekker, G.A.; Arstall, M.; Roberts, C.T. Mechanisms linking exposure to preeclampsia in utero and the risk for cardiovascular disease. J. Dev. Orig. Health Dis. 2020, 11, 235–242. [Google Scholar] [CrossRef] [PubMed]
  19. Turbeville, H.R.; Sasser, J.M. Preeclampsia beyond pregnancy: Long-term consequences for mother and child. Am. J. Physiol. Renal. Physiol. 2020, 318, F1315–F1326. [Google Scholar] [CrossRef]
  20. Melchiorre, K.; Thilaganathan, B.; Giorgione, V.; Ridder, A.; Memmo, A.; Khalil, A. Hypertensive Disorders of Pregnancy and Future Cardiovascular Health. Front. Cardiovasc. Med. 2020, 7, 59. [Google Scholar] [CrossRef]
  21. Rosenfeld, C.R.; White, R.E.; Roy, T.; Cox, B.E. Calcium-activated potassium channels and nitric oxide coregulate estrogen-induced vasodilation. Am. J. Physiol. Heart Circ. Physiol. 2000, 279, H319–H328. [Google Scholar] [CrossRef] [PubMed]
  22. Wei, A.D.; Gutman, G.A.; Aldrich, R.; Chandy, K.G.; Grissmer, S.; Wulff, H. International Union of Pharmacology. LII. Nomenclature and molecular relationships of calcium-activated potassium channels. Pharmacol. Rev. 2005, 57, 463–472. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Marty, A. Ca-dependent K channels with large unitary conductance in chromaffin cell membranes. Nature 1981, 291, 497–500. [Google Scholar] [CrossRef] [PubMed]
  24. Pallotta, B.S.; Magleby, K.L.; Barrett, J.N. Single channel recordings of Ca2+-activated K+ currents in rat muscle cell culture. Nature 1981, 293, 471–474. [Google Scholar] [CrossRef]
  25. McManus, O.B.; Magleby, K.L. Accounting for the Ca2+-dependent kinetics of single large-conductance Ca2+-activated K+ channels in rat skeletal muscle. J. Physiol. 1991, 443, 739–777. [Google Scholar] [CrossRef] [Green Version]
  26. Blatz, A.L.; Magleby, K.L. Single apamin-blocked Ca-activated K+ channels of small conductance in cultured rat skeletal muscle. Nature 1986, 323, 718–720. [Google Scholar] [CrossRef] [PubMed]
  27. Maylie, J.; Bond, C.T.; Herson, P.S.; Lee, W.S.; Adelman, J.P. Small conductance Ca2+-activated K+ channels and calmodulin. J. Physiol. 2004, 554, 255–261. [Google Scholar] [CrossRef] [PubMed]
  28. Ishii, T.M.; Silvia, C.; Hirschberg, B.; Bond, C.T.; Adelman, J.P.; Maylie, J. A human intermediate conductance calcium-activated potassium channel. Proc. Natl. Acad. Sci. USA 1997, 94, 11651–11656. [Google Scholar] [CrossRef] [Green Version]
  29. Neylon, C.B.; Lang, R.J.; Fu, Y.; Bobik, A.; Reinhart, P.H. Molecular cloning and characterization of the intermediate-conductance Ca2+-activated K+ channel in vascular smooth muscle: Relationship between K(Ca) channel diversity and smooth muscle cell function. Circ. Res. 1999, 85, e33–e43. [Google Scholar] [CrossRef] [Green Version]
  30. Kyle, B.D.; Braun, A.P. The regulation of BK channel activity by pre- and post-translational modifications. Front. Physiol. 2014, 5, 316. [Google Scholar] [CrossRef]
  31. Shipston, M.J.; Tian, L. Posttranscriptional and Posttranslational Regulation of BK Channels. Int. Rev. Neurobiol. 2016, 128, 91–126. [Google Scholar] [CrossRef] [PubMed]
  32. Kim, H.; Oh, K.H. Protein Network Interacting with BK Channels. Int. Rev. Neurobiol. 2016, 128, 127–161. [Google Scholar] [CrossRef] [PubMed]
  33. Hoshi, T.; Heinemann, S.H. Modulation of BK Channels by Small Endogenous Molecules and Pharmaceutical Channel Openers. Int. Rev. Neurobiol. 2016, 128, 193–237. [Google Scholar] [CrossRef] [PubMed]
  34. Latorre, R.; Castillo, K.; Carrasquel-Ursulaez, W.; Sepulveda, R.V.; Gonzalez-Nilo, F.; Gonzalez, C.; Alvarez, O. Molecular Determinants of BK Channel Functional Diversity and Functioning. Physiol. Rev. 2017, 97, 39–87. [Google Scholar] [CrossRef]
  35. Gonzalez-Perez, V.; Lingle, C.J. Regulation of BK Channels by Beta and Gamma Subunits. Annu. Rev. Physiol. 2019, 81, 113–137. [Google Scholar] [CrossRef]
  36. Sancho, M.; Kyle, B.D. The Large-Conductance, Calcium-Activated Potassium Channel: A Big Key Regulator of Cell Physiology. Front. Physiol. 2021, 12, 750615. [Google Scholar] [CrossRef]
  37. Hu, X.Q.; Zhang, L. Function and regulation of large conductance Ca2+-activated K+ channel in vascular smooth muscle cells. Drug Discov. Today 2012, 17, 974–987. [Google Scholar] [CrossRef] [Green Version]
  38. Meera, P.; Wallner, M.; Song, M.; Toro, L. Large conductance voltage- and calcium-dependent K+ channel, a distinct member of voltage-dependent ion channels with seven N-terminal transmembrane segments (S0-S6), an extracellular N terminus, and an intracellular (S9-S10) C terminus. Proc. Natl. Acad. Sci. USA 1997, 94, 14066–14071. [Google Scholar] [CrossRef] [Green Version]
  39. Salkoff, L.; Butler, A.; Ferreira, G.; Santi, C.; Wei, A. High-conductance potassium channels of the SLO family. Nat. Rev. Neurosci. 2006, 7, 921–931. [Google Scholar] [CrossRef]
  40. Lee, U.S.; Cui, J. BK channel activation: Structural and functional insights. Trends Neurosci. 2010, 33, 415–423. [Google Scholar] [CrossRef]
  41. Yuan, P.; Leonetti, M.D.; Hsiung, Y.; MacKinnon, R. Open structure of the Ca2+ gating ring in the high-conductance Ca2+-activated K+ channel. Nature 2012, 481, 94–97. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Brenner, R.; Perez, G.J.; Bonev, A.D.; Eckman, D.M.; Kosek, J.C.; Wiler, S.W.; Patterson, A.J.; Nelson, M.T.; Aldrich, R.W. Vasoregulation by the beta1 subunit of the calcium-activated potassium channel. Nature 2000, 407, 870–876. [Google Scholar] [CrossRef] [PubMed]
  43. Uebele, V.N.; Lagrutta, A.; Wade, T.; Figueroa, D.J.; Liu, Y.; McKenna, E.; Austin, C.P.; Bennett, P.B.; Swanson, R. Cloning and functional expression of two families of beta-subunits of the large conductance calcium-activated K+ channel. J. Biol. Chem. 2000, 275, 23211–23218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Behrens, R.; Nolting, A.; Reimann, F.; Schwarz, M.; Waldschutz, R.; Pongs, O. hKCNMB3 and hKCNMB4, cloning and characterization of two members of the large-conductance calcium-activated potassium channel beta subunit family. FEBS Lett. 2000, 474, 99–106. [Google Scholar] [CrossRef] [Green Version]
  45. Brenner, R.; Jegla, T.J.; Wickenden, A.; Liu, Y.; Aldrich, R.W. Cloning and functional characterization of novel large conductance calcium-activated potassium channel beta subunits, hKCNMB3 and hKCNMB4. J. Biol. Chem. 2000, 275, 6453–6461. [Google Scholar] [CrossRef] [Green Version]
  46. Tanaka, Y.; Meera, P.; Song, M.; Knaus, H.G.; Toro, L. Molecular constituents of maxi KCa channels in human coronary smooth muscle: Predominant alpha + beta subunit complexes. J. Physiol. 1997, 502 Pt. 3, 545–557. [Google Scholar] [CrossRef]
  47. Wang, Y.W.; Ding, J.P.; Xia, X.M.; Lingle, C.J. Consequences of the stoichiometry of Slo1 alpha and auxiliary beta subunits on functional properties of large-conductance Ca2+-activated K+ channels. J. Neurosci. 2002, 22, 1550–1561. [Google Scholar] [CrossRef] [Green Version]
  48. Gonzalez-Perez, V.; Zhou, Y.; Ciorba, M.A.; Lingle, C.J. The LRRC family of BK channel regulatory subunits: Potential roles in health and disease. J. Physiol. 2022, 600, 1357–1371. [Google Scholar] [CrossRef]
  49. Yan, J.; Aldrich, R.W. BK potassium channel modulation by leucine-rich repeat-containing proteins. Proc. Natl. Acad. Sci. USA 2012, 109, 7917–7922. [Google Scholar] [CrossRef] [Green Version]
  50. Kohler, M.; Hirschberg, B.; Bond, C.T.; Kinzie, J.M.; Marrion, N.V.; Maylie, J.; Adelman, J.P. Small-conductance, calcium-activated potassium channels from mammalian brain. Science 1996, 273, 1709–1714. [Google Scholar] [CrossRef]
  51. Sforna, L.; Megaro, A.; Pessia, M.; Franciolini, F.; Catacuzzeno, L. Structure, Gating and Basic Functions of the Ca2+-activated K Channel of Intermediate Conductance. Curr. Neuropharmacol. 2018, 16, 608–617. [Google Scholar] [CrossRef] [PubMed]
  52. Brown, B.M.; Shim, H.; Christophersen, P.; Wulff, H. Pharmacology of Small- and Intermediate-Conductance Calcium-Activated Potassium Channels. Annu. Rev. Pharmacol. Toxicol. 2020, 60, 219–240. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Evanson, K.W.; Bannister, J.P.; Leo, M.D.; Jaggar, J.H. LRRC26 is a functional BK channel auxiliary gamma subunit in arterial smooth muscle cells. Circ. Res. 2014, 115, 423–431. [Google Scholar] [CrossRef] [Green Version]
  54. Knaus, H.G.; Garcia-Calvo, M.; Kaczorowski, G.J.; Garcia, M.L. Subunit composition of the high conductance calcium-activated potassium channel from smooth muscle, a representative of the mSlo and slowpoke family of potassium channels. J. Biol. Chem. 1994, 269, 3921–3924. [Google Scholar] [CrossRef]
  55. Lorca, R.A.; Wakle-Prabagaran, M.; Freeman, W.E.; Pillai, M.K.; England, S.K. The large-conductance voltage- and Ca2+-activated K+ channel and its gamma1-subunit modulate mouse uterine artery function during pregnancy. J. Physiol. 2018, 596, 1019–1033. [Google Scholar] [CrossRef] [Green Version]
  56. Poulsen, A.N.; Wulf, H.; Hay-Schmidt, A.; Jansen-Olesen, I.; Olesen, J.; Klaerke, D.A. Differential expression of BK channel isoforms and beta-subunits in rat neuro-vascular tissues. Biochim. Biophys. Acta 2009, 1788, 380–389. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Rosenfeld, C.R.; Liu, X.T.; DeSpain, K. Pregnancy modifies the large conductance Ca2+-activated K+ channel and cGMP-dependent signaling pathway in uterine vascular smooth muscle. Am. J. Physiol. Heart Circ. Physiol. 2009, 296, H1878–H1887. [Google Scholar] [CrossRef] [Green Version]
  58. Wulf, H.; Hay-Schmidt, A.; Poulsen, A.N.; Klaerke, D.A.; Olesen, J.; Jansen-Olesen, I. Molecular investigations of BK(Ca) channels and the modulatory beta-subunits in porcine basilar and middle cerebral arteries. J. Mol. Histol. 2009, 40, 87–97. [Google Scholar] [CrossRef]
  59. Taylor, M.S.; Bonev, A.D.; Gross, T.P.; Eckman, D.M.; Brayden, J.E.; Bond, C.T.; Adelman, J.P.; Nelson, M.T. Altered expression of small-conductance Ca2+-activated K+ (SK3) channels modulates arterial tone and blood pressure. Circ. Res. 2003, 93, 124–131. [Google Scholar] [CrossRef] [Green Version]
  60. Burnham, M.P.; Bychkov, R.; Feletou, M.; Richards, G.R.; Vanhoutte, P.M.; Weston, A.H.; Edwards, G. Characterization of an apamin-sensitive small-conductance Ca2+-activated K+ channel in porcine coronary artery endothelium: Relevance to EDHF. Br. J. Pharmacol. 2002, 135, 1133–1143. [Google Scholar] [CrossRef]
  61. Gebremedhin, D.; Kaldunski, M.; Jacobs, E.R.; Harder, D.R.; Roman, R.J. Coexistence of two types of Ca2+-activated K+ channels in rat renal arterioles. Am. J. Physiol. 1996, 270, F69–F81. [Google Scholar] [PubMed]
  62. Quignard, J.F.; Feletou, M.; Edwards, G.; Duhault, J.; Weston, A.H.; Vanhoutte, P.M. Role of endothelial cell hyperpolarization in EDHF-mediated responses in the guinea-pig carotid artery. Br. J. Pharmacol. 2000, 129, 1103–1112. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Gauthier, K.M.; Spitzbarth, N.; Edwards, E.M.; Campbell, W.B. Apamin-sensitive K+ currents mediate arachidonic acid-induced relaxations of rabbit aorta. Hypertension 2004, 43, 413–419. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Kohler, R.; Wulff, H.; Eichler, I.; Kneifel, M.; Neumann, D.; Knorr, A.; Grgic, I.; Kampfe, D.; Si, H.; Wibawa, J.; et al. Blockade of the intermediate-conductance calcium-activated potassium channel as a new therapeutic strategy for restenosis. Circulation 2003, 108, 1119–1125. [Google Scholar] [CrossRef] [Green Version]
  65. Tharp, D.L.; Wamhoff, B.R.; Turk, J.R.; Bowles, D.K. Upregulation of intermediate-conductance Ca2+-activated K+ channel (IKCa1) mediates phenotypic modulation of coronary smooth muscle. Am. J. Physiol. Heart Circ. Physiol. 2006, 291, H2493–H2503. [Google Scholar] [CrossRef]
  66. Si, H.; Grgic, I.; Heyken, W.T.; Maier, T.; Hoyer, J.; Reusch, H.P.; Kohler, R. Mitogenic modulation of Ca2+-activated K+ channels in proliferating A7r5 vascular smooth muscle cells. Br. J. Pharmacol. 2006, 148, 909–917. [Google Scholar] [CrossRef] [Green Version]
  67. Toyama, K.; Wulff, H.; Chandy, K.G.; Azam, P.; Raman, G.; Saito, T.; Fujiwara, Y.; Mattson, D.L.; Das, S.; Melvin, J.E.; et al. The intermediate-conductance calcium-activated potassium channel KCa3.1 contributes to atherogenesis in mice and humans. J. Clin. Investig. 2008, 118, 3025–3037. [Google Scholar] [CrossRef] [Green Version]
  68. Zhu, R.; Hu, X.Q.; Xiao, D.; Yang, S.; Wilson, S.M.; Longo, L.D.; Zhang, L. Chronic hypoxia inhibits pregnancy-induced upregulation of SKCa channel expression and function in uterine arteries. Hypertension 2013, 62, 367–374. [Google Scholar] [CrossRef] [Green Version]
  69. He, M.; Li, F.; Yang, M.; Fan, Y.; Beejadhursing, R.; Xie, Y.; Zhou, Y.; Deng, D. Impairment of BKca channels in human placental chorionic plate arteries is potentially relevant to the development of preeclampsia. Hypertens. Res. 2018, 41, 126–134. [Google Scholar] [CrossRef]
  70. Nilius, B.; Droogmans, G. Ion channels and their functional role in vascular endothelium. Physiol. Rev. 2001, 81, 1415–1459. [Google Scholar] [CrossRef]
  71. Sandow, S.L.; Grayson, T.H. Limits of isolation and culture: Intact vascular endothelium and BKCa. Am. J. Physiol. Heart Circ. Physiol. 2009, 297, H1–H7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Rusko, J.; Tanzi, F.; van Breemen, C.; Adams, D.J. Calcium-activated potassium channels in native endothelial cells from rabbit aorta: Conductance, Ca2+ sensitivity and block. J. Physiol. 1992, 455, 601–621. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Ungvari, Z.; Csiszar, A.; Koller, A. Increases in endothelial Ca2+ activate K(Ca) channels and elicit EDHF-type arteriolar dilation via gap junctions. Am. J. Physiol. Heart Circ. Physiol. 2002, 282, H1760–H1767. [Google Scholar] [CrossRef] [Green Version]
  74. Brakemeier, S.; Eichler, I.; Knorr, A.; Fassheber, T.; Kohler, R.; Hoyer, J. Modulation of Ca2+-activated K+ channel in renal artery endothelium in situ by nitric oxide and reactive oxygen species. Kidney Int. 2003, 64, 199–207. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Wang, X.L.; Ye, D.; Peterson, T.E.; Cao, S.; Shah, V.H.; Katusic, Z.S.; Sieck, G.C.; Lee, H.C. Caveolae targeting and regulation of large conductance Ca2+-activated K+ channels in vascular endothelial cells. J. Biol. Chem. 2005, 280, 11656–11664. [Google Scholar] [CrossRef] [Green Version]
  76. Dong, D.L.; Zhang, Y.; Lin, D.H.; Chen, J.; Patschan, S.; Goligorsky, M.S.; Nasjletti, A.; Yang, B.F.; Wang, W.H. Carbon monoxide stimulates the Ca2+-activated big conductance k channels in cultured human endothelial cells. Hypertension 2007, 50, 643–651. [Google Scholar] [CrossRef] [Green Version]
  77. Vang, A.; Mazer, J.; Casserly, B.; Choudhary, G. Activation of endothelial BKCa channels causes pulmonary vasodilation. Vascul. Pharmacol. 2010, 53, 122–129. [Google Scholar] [CrossRef]
  78. Jackson-Weaver, O.; Osmond, J.M.; Riddle, M.A.; Naik, J.S.; Gonzalez Bosc, L.V.; Walker, B.R.; Kanagy, N.L. Hydrogen sulfide dilates rat mesenteric arteries by activating endothelial large-conductance Ca2+-activated K+ channels and smooth muscle Ca2+ sparks. Am. J. Physiol. Heart Circ. Physiol. 2013, 304, H1446–H1454. [Google Scholar] [CrossRef] [Green Version]
  79. Marchenko, S.M.; Sage, S.O. Calcium-activated potassium channels in the endothelium of intact rat aorta. J. Physiol. 1996, 492 Pt 1, 53–60. [Google Scholar] [CrossRef] [Green Version]
  80. Kohler, R.; Degenhardt, C.; Kuhn, M.; Runkel, N.; Paul, M.; Hoyer, J. Expression and function of endothelial Ca2+-activated K+ channels in human mesenteric artery: A single-cell reverse transcriptase-polymerase chain reaction and electrophysiological study in situ. Circ. Res. 2000, 87, 496–503. [Google Scholar] [CrossRef]
  81. Bychkov, R.; Burnham, M.P.; Richards, G.R.; Edwards, G.; Weston, A.H.; Feletou, M.; Vanhoutte, P.M. Characterization of a charybdotoxin-sensitive intermediate conductance Ca2+-activated K+ channel in porcine coronary endothelium: Relevance to EDHF. Br. J. Pharmacol. 2002, 137, 1346–1354. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Gauthier, K.M.; Liu, C.; Popovic, A.; Albarwani, S.; Rusch, N.J. Freshly isolated bovine coronary endothelial cells do not express the BK Ca channel gene. J. Physiol. 2002, 545, 829–836. [Google Scholar] [CrossRef]
  83. Ledoux, J.; Bonev, A.D.; Nelson, M.T. Ca2+-activated K+ channels in murine endothelial cells: Block by intracellular calcium and magnesium. J. Gen. Physiol. 2008, 131, 125–135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Jow, F.; Sullivan, K.; Sokol, P.; Numann, R. Induction of Ca2+-activated K+ current and transient outward currents in human capillary endothelial cells. J. Membr. Biol. 1999, 167, 53–64. [Google Scholar] [CrossRef] [PubMed]
  85. Papassotiriou, J.; Kohler, R.; Prenen, J.; Krause, H.; Akbar, M.; Eggermont, J.; Paul, M.; Distler, A.; Nilius, B.; Hoyer, J. Endothelial K+ channel lacks the Ca2+ sensitivity-regulating beta subunit. FASEB J. 2000, 14, 885–894. [Google Scholar] [CrossRef] [PubMed]
  86. Kestler, H.A.; Janko, S.; Haussler, U.; Muche, R.; Hombach, V.; Hoher, M.; Wiecha, J. A remark on the high-conductance calcium-activated potassium channel in human endothelial cells. Res. Exp. Med. 1998, 198, 133–143. [Google Scholar] [CrossRef]
  87. Hughes, J.M.; Riddle, M.A.; Paffett, M.L.; Gonzalez Bosc, L.V.; Walker, B.R. Novel role of endothelial BKCa channels in altered vasoreactivity following hypoxia. Am. J. Physiol. Heart Circ. Physiol. 2010, 299, H1439–H1450. [Google Scholar] [CrossRef] [Green Version]
  88. Riddle, M.A.; Hughes, J.M.; Walker, B.R. Role of caveolin-1 in endothelial BKCa channel regulation of vasoreactivity. Am. J. Physiol. Cell Physiol. 2011, 301, C1404–C1414. [Google Scholar] [CrossRef] [Green Version]
  89. Kohler, R.; Olivan-Viguera, A.; Wulff, H. Endothelial Small- and Intermediate-Conductance K Channels and Endothelium-Dependent Hyperpolarization as Drug Targets in Cardiovascular Disease. Adv. Pharmacol. 2016, 77, 65–104. [Google Scholar] [CrossRef]
  90. Kohler, R.; Hoyer, J. The endothelium-derived hyperpolarizing factor: Insights from genetic animal models. Kidney Int. 2007, 72, 145–150. [Google Scholar] [CrossRef]
  91. Dalsgaard, T.; Kroigaard, C.; Misfeldt, M.; Bek, T.; Simonsen, U. Openers of small conductance calcium-activated potassium channels selectively enhance NO-mediated bradykinin vasodilatation in porcine retinal arterioles. Br. J. Pharmacol. 2010, 160, 1496–1508. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Sandow, S.L.; Neylon, C.B.; Chen, M.X.; Garland, C.J. Spatial separation of endothelial small- and intermediate-conductance calcium-activated potassium channels (K(Ca)) and connexins: Possible relationship to vasodilator function? J. Anat. 2006, 209, 689–698. [Google Scholar] [CrossRef]
  93. Dora, K.A.; Gallagher, N.T.; McNeish, A.; Garland, C.J. Modulation of endothelial cell KCa3.1 channels during endothelium-derived hyperpolarizing factor signaling in mesenteric resistance arteries. Circ. Res. 2008, 102, 1247–1255. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Nelson, M.T.; Cheng, H.; Rubart, M.; Santana, L.F.; Bonev, A.D.; Knot, H.J.; Lederer, W.J. Relaxation of arterial smooth muscle by calcium sparks. Science 1995, 270, 633–637. [Google Scholar] [CrossRef] [PubMed]
  95. Earley, S.; Heppner, T.J.; Nelson, M.T.; Brayden, J.E. TRPV4 forms a novel Ca2+ signaling complex with ryanodine receptors and BKCa channels. Circ. Res. 2005, 97, 1270–1279. [Google Scholar] [CrossRef] [Green Version]
  96. Jaggar, J.H.; Stevenson, A.S.; Nelson, M.T. Voltage dependence of Ca2+ sparks in intact cerebral arteries. Am. J. Physiol. 1998, 274, C1755–C1761. [Google Scholar] [CrossRef]
  97. Zhuge, R.; Fogarty, K.E.; Tuft, R.A.; Walsh, J.V., Jr. Spontaneous transient outward currents arise from microdomains where BK channels are exposed to a mean Ca2+ concentration on the order of 10 microM during a Ca2+ spark. J. Gen. Physiol. 2002, 120, 15–27. [Google Scholar] [CrossRef] [Green Version]
  98. Perez, G.J.; Bonev, A.D.; Nelson, M.T. Micromolar Ca2+ from sparks activates Ca2+-sensitive K+ channels in rat cerebral artery smooth muscle. Am. J. Physiol. Cell Physiol. 2001, 281, C1769–C1775. [Google Scholar] [CrossRef]
  99. Cheng, H.; Lederer, W.J. Calcium sparks. Physiol. Rev. 2008, 88, 1491–1545. [Google Scholar] [CrossRef] [Green Version]
  100. Pluger, S.; Faulhaber, J.; Furstenau, M.; Lohn, M.; Waldschutz, R.; Gollasch, M.; Haller, H.; Luft, F.C.; Ehmke, H.; Pongs, O. Mice with disrupted BK channel beta1 subunit gene feature abnormal Ca2+ spark/STOC coupling and elevated blood pressure. Circ. Res. 2000, 87, E53–E60. [Google Scholar] [CrossRef]
  101. Rueda, A.; Fernandez-Velasco, M.; Benitah, J.P.; Gomez, A.M. Abnormal Ca2+ spark/STOC coupling in cerebral artery smooth muscle cells of obese type 2 diabetic mice. PLoS ONE 2013, 8, e53321. [Google Scholar] [CrossRef] [PubMed]
  102. Liu, G.; Shi, J.; Yang, L.; Cao, L.; Park, S.M.; Cui, J.; Marx, S.O. Assembly of a Ca2+-dependent BK channel signaling complex by binding to beta2 adrenergic receptor. EMBO J. 2004, 23, 2196–2205. [Google Scholar] [CrossRef] [Green Version]
  103. Yamamura, H.; Ikeda, C.; Suzuki, Y.; Ohya, S.; Imaizumi, Y. Molecular assembly and dynamics of fluorescent protein-tagged single KCa1.1 channel in expression system and vascular smooth muscle cells. Am. J. Physiol. Cell Physiol. 2012, 302, C1257–C1268. [Google Scholar] [CrossRef] [PubMed]
  104. Suzuki, Y.; Yamamura, H.; Ohya, S.; Imaizumi, Y. Caveolin-1 facilitates the direct coupling between large conductance Ca2+-activated K+ (BKCa) and Cav1.2 Ca2+ channels and their clustering to regulate membrane excitability in vascular myocytes. J. Biol. Chem. 2013, 288, 36750–36761. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Kwan, H.Y.; Shen, B.; Ma, X.; Kwok, Y.C.; Huang, Y.; Man, Y.B.; Yu, S.; Yao, X. TRPC1 associates with BK(Ca) channel to form a signal complex in vascular smooth muscle cells. Circ. Res. 2009, 104, 670–678. [Google Scholar] [CrossRef] [Green Version]
  106. Essin, K.; Welling, A.; Hofmann, F.; Luft, F.C.; Gollasch, M.; Moosmang, S. Indirect coupling between Cav1.2 channels and ryanodine receptors to generate Ca2+ sparks in murine arterial smooth muscle cells. J. Physiol. 2007, 584, 205–219. [Google Scholar] [CrossRef] [PubMed]
  107. Davis, M.J.; Hill, M.A. Signaling mechanisms underlying the vascular myogenic response. Physiol. Rev. 1999, 79, 387–423. [Google Scholar] [CrossRef] [Green Version]
  108. Brayden, J.E.; Nelson, M.T. Regulation of arterial tone by activation of calcium-dependent potassium channels. Science 1992, 256, 532–535. [Google Scholar] [CrossRef] [PubMed]
  109. Lohn, M.; Lauterbach, B.; Haller, H.; Pongs, O.; Luft, F.C.; Gollasch, M. beta(1)-Subunit of BK channels regulates arterial wall [Ca2+] and diameter in mouse cerebral arteries. J. Appl. Physiol. 2001, 91, 1350–1354. [Google Scholar] [CrossRef]
  110. Xu, H.; Kandlikar, S.S.; Westcott, E.B.; Fink, G.D.; Galligan, J.J. Requirement for functional BK channels in maintaining oscillation in venomotor tone revealed by species differences in expression of the beta1 accessory subunits. J. Cardiovasc. Pharmacol. 2012, 59, 29–36. [Google Scholar] [CrossRef]
  111. Sausbier, M.; Arntz, C.; Bucurenciu, I.; Zhao, H.; Zhou, X.B.; Sausbier, U.; Feil, S.; Kamm, S.; Essin, K.; Sailer, C.A.; et al. Elevated blood pressure linked to primary hyperaldosteronism and impaired vasodilation in BK channel-deficient mice. Circulation 2005, 112, 60–68. [Google Scholar] [CrossRef] [PubMed]
  112. Oelze, M.; Warnholtz, A.; Faulhaber, J.; Wenzel, P.; Kleschyov, A.L.; Coldewey, M.; Hink, U.; Pongs, O.; Fleming, I.; Wassmann, S.; et al. NADPH oxidase accounts for enhanced superoxide production and impaired endothelium-dependent smooth muscle relaxation in BKbeta1-/- mice. Arter. Thromb. Vasc. Biol. 2006, 26, 1753–1759. [Google Scholar] [CrossRef] [PubMed]
  113. Zheng, Y.M.; Park, S.W.; Stokes, L.; Tang, Q.; Xiao, J.H.; Wang, Y.X. Distinct activity of BK channel beta1-subunit in cerebral and pulmonary artery smooth muscle cells. Am. J. Physiol. Cell Physiol. 2013, 304, C780–C789. [Google Scholar] [CrossRef] [Green Version]
  114. Yang, Y.; Li, P.Y.; Cheng, J.; Mao, L.; Wen, J.; Tan, X.Q.; Liu, Z.F.; Zeng, X.R. Function of BKCa channels is reduced in human vascular smooth muscle cells from Han Chinese patients with hypertension. Hypertension 2013, 61, 519–525. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Amberg, G.C.; Bonev, A.D.; Rossow, C.F.; Nelson, M.T.; Santana, L.F. Modulation of the molecular composition of large conductance, Ca2+ activated K+ channels in vascular smooth muscle during hypertension. J. Clin. Investig. 2003, 112, 717–724. [Google Scholar] [CrossRef] [Green Version]
  116. Amberg, G.C.; Santana, L.F. Downregulation of the BK channel beta1 subunit in genetic hypertension. Circ. Res. 2003, 93, 965–971. [Google Scholar] [CrossRef] [Green Version]
  117. Nieves-Cintron, M.; Amberg, G.C.; Nichols, C.B.; Molkentin, J.D.; Santana, L.F. Activation of NFATc3 down-regulates the beta1 subunit of large conductance, calcium-activated K+ channels in arterial smooth muscle and contributes to hypertension. J. Biol. Chem. 2007, 282, 3231–3240. [Google Scholar] [CrossRef] [Green Version]
  118. Fernandez-Fernandez, J.M.; Tomas, M.; Vazquez, E.; Orio, P.; Latorre, R.; Senti, M.; Marrugat, J.; Valverde, M.A. Gain-of-function mutation in the KCNMB1 potassium channel subunit is associated with low prevalence of diastolic hypertension. J. Clin. Investig. 2004, 113, 1032–1039. [Google Scholar] [CrossRef]
  119. Senti, M.; Fernandez-Fernandez, J.M.; Tomas, M.; Vazquez, E.; Elosua, R.; Marrugat, J.; Valverde, M.A. Protective effect of the KCNMB1 E65K genetic polymorphism against diastolic hypertension in aging women and its relevance to cardiovascular risk. Circ. Res. 2005, 97, 1360–1365. [Google Scholar] [CrossRef] [Green Version]
  120. Nielsen, T.; Burgdorf, K.S.; Grarup, N.; Borch-Johnsen, K.; Hansen, T.; Jorgensen, T.; Pedersen, O.; Andersen, G. The KCNMB1 Glu65Lys polymorphism associates with reduced systolic and diastolic blood pressure in the Inter99 study of 5729 Danes. J. Hypertens. 2008, 26, 2142–2146. [Google Scholar] [CrossRef]
  121. Zhao, G.; Zhao, Y.; Pan, B.; Liu, J.; Huang, X.; Zhang, X.; Cao, C.; Hou, N.; Wu, C.; Zhao, K.S.; et al. Hypersensitivity of BKCa to Ca2+ sparks underlies hyporeactivity of arterial smooth muscle in shock. Circ. Res. 2007, 101, 493–502. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. McGahon, M.K.; Dash, D.P.; Arora, A.; Wall, N.; Dawicki, J.; Simpson, D.A.; Scholfield, C.N.; McGeown, J.G.; Curtis, T.M. Diabetes downregulates large-conductance Ca2+-activated potassium beta 1 channel subunit in retinal arteriolar smooth muscle. Circ. Res. 2007, 100, 703–711. [Google Scholar] [CrossRef] [Green Version]
  123. Yi, F.; Wang, H.; Chai, Q.; Wang, X.; Shen, W.K.; Willis, M.S.; Lee, H.C.; Lu, T. Regulation of large conductance Ca2+-activated K+ (BK) channel beta1 subunit expression by muscle RING finger protein 1 in diabetic vessels. J. Biol. Chem. 2014, 289, 10853–10864. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Schubert, R.; Nelson, M.T. Protein kinases: Tuners of the BKCa channel in smooth muscle. Trends Pharmacol. Sci. 2001, 22, 505–512. [Google Scholar] [CrossRef]
  125. Hu, S.L.; Kim, H.S.; Jeng, A.Y. Dual action of endothelin-1 on the Ca2+-activated K+ channel in smooth muscle cells of porcine coronary artery. Eur. J. Pharmacol. 1991, 194, 31–36. [Google Scholar] [CrossRef] [PubMed]
  126. Lange, A.; Gebremedhin, D.; Narayanan, J.; Harder, D. 20-Hydroxyeicosatetraenoic acid-induced vasoconstriction and inhibition of potassium current in cerebral vascular smooth muscle is dependent on activation of protein kinase C. J. Biol. Chem. 1997, 272, 27345–27352. [Google Scholar] [CrossRef] [Green Version]
  127. Sun, C.W.; Falck, J.R.; Harder, D.R.; Roman, R.J. Role of tyrosine kinase and PKC in the vasoconstrictor response to 20-HETE in renal arterioles. Hypertension 1999, 33, 414–418. [Google Scholar] [CrossRef] [Green Version]
  128. Alioua, A.; Mahajan, A.; Nishimaru, K.; Zarei, M.M.; Stefani, E.; Toro, L. Coupling of c-Src to large conductance voltage- and Ca2+-activated K+ channels as a new mechanism of agonist-induced vasoconstriction. Proc. Natl. Acad. Sci. USA 2002, 99, 14560–14565. [Google Scholar] [CrossRef] [Green Version]
  129. Crozatier, B. Central role of PKCs in vascular smooth muscle cell ion channel regulation. J. Mol. Cell. Cardiol. 2006, 41, 952–955. [Google Scholar] [CrossRef]
  130. Robertson, B.E.; Schubert, R.; Hescheler, J.; Nelson, M.T. cGMP-dependent protein kinase activates Ca-activated K channels in cerebral artery smooth muscle cells. Am. J. Physiol. 1993, 265, C299–C303. [Google Scholar] [CrossRef]
  131. Cabell, F.; Weiss, D.S.; Price, J.M. Inhibition of adenosine-induced coronary vasodilation by block of large-conductance Ca2+-activated K+ channels. Am. J. Physiol. 1994, 267, H1455–H1460. [Google Scholar] [PubMed]
  132. Miyoshi, H.; Nakaya, Y. Calcitonin gene-related peptide activates the K+ channels of vascular smooth muscle cells via adenylate cyclase. Basic Res. Cardiol. 1995, 90, 332–336. [Google Scholar] [CrossRef]
  133. Song, Y.; Simard, J.M. beta-Adrenoceptor stimulation activates large-conductance Ca2+-activated K+ channels in smooth muscle cells from basilar artery of guinea pig. Pflug. Arch. 1995, 430, 984–993. [Google Scholar] [CrossRef] [PubMed]
  134. Paterno, R.; Faraci, F.M.; Heistad, D.D. Role of Ca2+-dependent K+ channels in cerebral vasodilatation induced by increases in cyclic GMP and cyclic AMP in the rat. Stroke 1996, 27, 1603–1607; discussion 1607–1608. [Google Scholar] [CrossRef] [PubMed]
  135. Zhao, Y.J.; Wang, J.; Rubin, L.J.; Yuan, X.J. Inhibition of K(V) and K(Ca) channels antagonizes NO-induced relaxation in pulmonary artery. Am. J. Physiol. 1997, 272, H904–H912. [Google Scholar] [CrossRef]
  136. Tanaka, Y.; Aida, M.; Tanaka, H.; Shigenobu, K.; Toro, L. Involvement of maxi-K(Ca) channel activation in atrial natriuretic peptide-induced vasorelaxation. Naunyn Schmiedebergs Arch. Pharmacol. 1998, 357, 705–708. [Google Scholar] [CrossRef]
  137. Leo, M.D.; Bannister, J.P.; Narayanan, D.; Nair, A.; Grubbs, J.E.; Gabrick, K.S.; Boop, F.A.; Jaggar, J.H. Dynamic regulation of beta1 subunit trafficking controls vascular contractility. Proc. Natl. Acad. Sci. USA 2014, 111, 2361–2366. [Google Scholar] [CrossRef] [Green Version]
  138. Bolotina, V.M.; Najibi, S.; Palacino, J.J.; Pagano, P.J.; Cohen, R.A. Nitric oxide directly activates calcium-dependent potassium channels in vascular smooth muscle. Nature 1994, 368, 850–853. [Google Scholar] [CrossRef]
  139. Mistry, D.K.; Garland, C.J. Nitric oxide (NO)-induced activation of large conductance Ca2+-dependent K+ channels (BK(Ca)) in smooth muscle cells isolated from the rat mesenteric artery. Br. J. Pharmacol. 1998, 124, 1131–1140. [Google Scholar] [CrossRef] [Green Version]
  140. Nakashima, M.; Mombouli, J.V.; Taylor, A.A.; Vanhoutte, P.M. Endothelium-dependent hyperpolarization caused by bradykinin in human coronary arteries. J. Clin. Investig. 1993, 92, 2867–2871. [Google Scholar] [CrossRef]
  141. Nakashima, Y.; Toki, Y.; Fukami, Y.; Hibino, M.; Okumura, K.; Ito, T. Role of K+ channels in EDHF-dependent relaxation induced by acetylcholine in canine coronary artery. Heart Vessels 1997, 12, 287–293. [Google Scholar] [CrossRef] [PubMed]
  142. Miura, H.; Liu, Y.; Gutterman, D.D. Human coronary arteriolar dilation to bradykinin depends on membrane hyperpolarization: Contribution of nitric oxide and Ca2+-activated K+ channels. Circulation 1999, 99, 3132–3138. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Crane, G.J.; Gallagher, N.; Dora, K.A.; Garland, C.J. Small- and intermediate-conductance calcium-activated K+ channels provide different facets of endothelium-dependent hyperpolarization in rat mesenteric artery. J. Physiol. 2003, 553, 183–189. [Google Scholar] [CrossRef] [PubMed]
  144. Tran, Q.K.; Ohashi, K.; Watanabe, H. Calcium signalling in endothelial cells. Cardiovasc. Res. 2000, 48, 13–22. [Google Scholar] [CrossRef] [Green Version]
  145. Schumacher, M.A.; Rivard, A.F.; Bachinger, H.P.; Adelman, J.P. Structure of the gating domain of a Ca2+-activated K+ channel complexed with Ca2+/calmodulin. Nature 2001, 410, 1120–1124. [Google Scholar] [CrossRef] [PubMed]
  146. Pogoda, K.; Kameritsch, P. Molecular regulation of myoendothelial gap junctions. Curr. Opin. Pharmacol. 2019, 45, 16–22. [Google Scholar] [CrossRef] [PubMed]
  147. Schmidt, K.; de Wit, C. Endothelium-Derived Hyperpolarizing Factor and Myoendothelial Coupling: The in vivo Perspective. Front. Physiol. 2020, 11, 602930. [Google Scholar] [CrossRef]
  148. Garland, C.J.; Dora, K.A. Endothelium-Dependent Hyperpolarization: The Evolution of Myoendothelial Microdomains. J. Cardiovasc. Pharmacol. 2021, 78, S3–S12. [Google Scholar] [CrossRef]
  149. Edwards, G.; Dora, K.A.; Gardener, M.J.; Garland, C.J.; Weston, A.H. K+ is an endothelium-derived hyperpolarizing factor in rat arteries. Nature 1998, 396, 269–272. [Google Scholar] [CrossRef]
  150. Dora, K.A.; Garland, C.J. Properties of smooth muscle hyperpolarization and relaxation to K+ in the rat isolated mesenteric artery. Am. J. Physiol. Heart Circ. Physiol. 2001, 280, H2424–H2429. [Google Scholar] [CrossRef]
  151. Stankevicius, E.; Lopez-Valverde, V.; Rivera, L.; Hughes, A.D.; Mulvany, M.J.; Simonsen, U. Combination of Ca2+-activated K+ channel blockers inhibits acetylcholine-evoked nitric oxide release in rat superior mesenteric artery. Br. J. Pharmacol. 2006, 149, 560–572. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Sheng, J.Z.; Braun, A.P. Small- and intermediate-conductance Ca2+-activated K+ channels directly control agonist-evoked nitric oxide synthesis in human vascular endothelial cells. Am. J. Physiol. Cell Physiol. 2007, 293, C458–C467. [Google Scholar] [CrossRef] [Green Version]
  153. Sheng, J.Z.; Ella, S.; Davis, M.J.; Hill, M.A.; Braun, A.P. Openers of SKCa and IKCa channels enhance agonist-evoked endothelial nitric oxide synthesis and arteriolar vasodilation. FASEB J. 2009, 23, 1138–1145. [Google Scholar] [CrossRef] [Green Version]
  154. Grgic, I.; Kaistha, B.P.; Hoyer, J.; Kohler, R. Endothelial Ca+-activated K+ channels in normal and impaired EDHF-dilator responses--relevance to cardiovascular pathologies and drug discovery. Br. J. Pharmacol. 2009, 157, 509–526. [Google Scholar] [CrossRef] [Green Version]
  155. Stankevicius, E.; Dalsgaard, T.; Kroigaard, C.; Beck, L.; Boedtkjer, E.; Misfeldt, M.W.; Nielsen, G.; Schjorring, O.; Hughes, A.; Simonsen, U. Opening of small and intermediate calcium-activated potassium channels induces relaxation mainly mediated by nitric-oxide release in large arteries and endothelium-derived hyperpolarizing factor in small arteries from rat. J. Pharmacol. Exp. Ther. 2011, 339, 842–850. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Feletou, M.; Vanhoutte, P.M. EDHF: An update. Clin. Sci. 2009, 117, 139–155. [Google Scholar] [CrossRef] [Green Version]
  157. Campbell, W.B.; Gebremedhin, D.; Pratt, P.F.; Harder, D.R. Identification of epoxyeicosatrienoic acids as endothelium-derived hyperpolarizing factors. Circ. Res. 1996, 78, 415–423. [Google Scholar] [CrossRef] [PubMed]
  158. Hayabuchi, Y.; Nakaya, Y.; Matsuoka, S.; Kuroda, Y. Endothelium-derived hyperpolarizing factor activates Ca2+-activated K+ channels in porcine coronary artery smooth muscle cells. J. Cardiovasc. Pharmacol. 1998, 32, 642–649. [Google Scholar] [CrossRef] [PubMed]
  159. Archer, S.L.; Gragasin, F.S.; Wu, X.; Wang, S.; McMurtry, S.; Kim, D.H.; Platonov, M.; Koshal, A.; Hashimoto, K.; Campbell, W.B.; et al. Endothelium-derived hyperpolarizing factor in human internal mammary artery is 11,12-epoxyeicosatrienoic acid and causes relaxation by activating smooth muscle BK(Ca) channels. Circulation 2003, 107, 769–776. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  160. Doughty, J.M.; Plane, F.; Langton, P.D. Charybdotoxin and apamin block EDHF in rat mesenteric artery if selectively applied to the endothelium. Am. J. Physiol. 1999, 276, H1107–H1112. [Google Scholar] [CrossRef]
  161. Si, H.; Heyken, W.T.; Wolfle, S.E.; Tysiac, M.; Schubert, R.; Grgic, I.; Vilianovich, L.; Giebing, G.; Maier, T.; Gross, V.; et al. Impaired endothelium-derived hyperpolarizing factor-mediated dilations and increased blood pressure in mice deficient of the intermediate-conductance Ca2+-activated K+ channel. Circ. Res. 2006, 99, 537–544. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Eichler, I.; Wibawa, J.; Grgic, I.; Knorr, A.; Brakemeier, S.; Pries, A.R.; Hoyer, J.; Kohler, R. Selective blockade of endothelial Ca2+-activated small- and intermediate-conductance K+-channels suppresses EDHF-mediated vasodilation. Br. J. Pharmacol. 2003, 138, 594–601. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Hannah, R.M.; Dunn, K.M.; Bonev, A.D.; Nelson, M.T. Endothelial SK(Ca) and IK(Ca) channels regulate brain parenchymal arteriolar diameter and cortical cerebral blood flow. J. Cereb. Blood Flow Metab. 2011, 31, 1175–1186. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Yap, F.C.; Weber, D.S.; Taylor, M.S.; Townsley, M.I.; Comer, B.S.; Maylie, J.; Adelman, J.P.; Lin, M.T. Endothelial SK3 channel-associated Ca2+ microdomains modulate blood pressure. Am. J. Physiol. Heart Circ. Physiol. 2016, 310, H1151–H1163. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Sankaranarayanan, A.; Raman, G.; Busch, C.; Schultz, T.; Zimin, P.I.; Hoyer, J.; Kohler, R.; Wulff, H. Naphtho[1,2-d]thiazol-2-ylamine (SKA-31), a new activator of KCa2 and KCa3.1 potassium channels, potentiates the endothelium-derived hyperpolarizing factor response and lowers blood pressure. Mol. Pharmacol. 2009, 75, 281–295. [Google Scholar] [CrossRef]
  166. Damkjaer, M.; Nielsen, G.; Bodendiek, S.; Staehr, M.; Gramsbergen, J.B.; de Wit, C.; Jensen, B.L.; Simonsen, U.; Bie, P.; Wulff, H.; et al. Pharmacological activation of KCa3.1/KCa2.3 channels produces endothelial hyperpolarization and lowers blood pressure in conscious dogs. Br. J. Pharmacol. 2012, 165, 223–234. [Google Scholar] [CrossRef] [Green Version]
  167. Mishra, R.C.; Belke, D.; Wulff, H.; Braun, A.P. SKA-31, a novel activator of SK(Ca) and IK(Ca) channels, increases coronary flow in male and female rat hearts. Cardiovasc. Res. 2013, 97, 339–348. [Google Scholar] [CrossRef] [Green Version]
  168. Brahler, S.; Kaistha, A.; Schmidt, V.J.; Wolfle, S.E.; Busch, C.; Kaistha, B.P.; Kacik, M.; Hasenau, A.L.; Grgic, I.; Si, H.; et al. Genetic deficit of SK3 and IK1 channels disrupts the endothelium-derived hyperpolarizing factor vasodilator pathway and causes hypertension. Circulation 2009, 119, 2323–2332. [Google Scholar] [CrossRef]
  169. Hilgers, R.H.; Webb, R.C. Reduced expression of SKCa and IKCa channel proteins in rat small mesenteric arteries during angiotensin II-induced hypertension. Am. J. Physiol. Heart Circ. Physiol. 2007, 292, H2275–H2284. [Google Scholar] [CrossRef] [Green Version]
  170. Weston, A.H.; Porter, E.L.; Harno, E.; Edwards, G. Impairment of endothelial SK(Ca) channels and of downstream hyperpolarizing pathways in mesenteric arteries from spontaneously hypertensive rats. Br. J. Pharmacol. 2010, 160, 836–843. [Google Scholar] [CrossRef]
  171. Saito, T.; Fujiwara, Y.; Fujiwara, R.; Hasegawa, H.; Kibira, S.; Miura, H.; Miura, M. Role of augmented expression of intermediate-conductance Ca2+-activated K+ channels in postischaemic heart. Clin. Exp. Pharmacol. Physiol. 2002, 29, 324–329. [Google Scholar] [CrossRef] [PubMed]
  172. Tharp, D.L.; Wamhoff, B.R.; Wulff, H.; Raman, G.; Cheong, A.; Bowles, D.K. Local delivery of the KCa3.1 blocker, TRAM-34, prevents acute angioplasty-induced coronary smooth muscle phenotypic modulation and limits stenosis. Arter. Thromb. Vasc. Biol. 2008, 28, 1084–1089. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Schach, C.; Resch, M.; Schmid, P.M.; Riegger, G.A.; Endemann, D.H. 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. 2014, 307, H1093–H1102. [Google Scholar] [CrossRef] [Green Version]
  174. Kroigaard, C.; Kudryavtseva, O.; Dalsgaard, T.; Wandall-Frostholm, C.; Olesen, S.P.; Simonsen, U. K(Ca)3.1 channel downregulation and impaired endothelium-derived hyperpolarization-type relaxation in pulmonary arteries from chronically hypoxic rats. Exp. Physiol. 2013, 98, 957–969. [Google Scholar] [CrossRef]
  175. Rosenfeld, C.R.; Morriss, F.H., Jr.; Makowski, E.L.; Meschia, G.; Battaglia, F.C. Circulatory changes in the reproductive tissues of ewes during pregnancy. Gynecol. Investig. 1974, 5, 252–268. [Google Scholar] [CrossRef] [PubMed]
  176. Silver, M.; Barnes, R.J.; Comline, R.S.; Burton, G.J. Placental blood flow: Some fetal and maternal cardiovascular adjustments during gestation. J. Reprod. Fertil. Suppl. 1982, 31, 139–160. [Google Scholar]
  177. Ziegler, W.F.; Bernstein, I.; Badger, G.; Leavitt, T.; Cerrero, M.L. Regional hemodynamic adaptation during the menstrual cycle. Obstet. Gynecol. 1999, 94, 695–699. [Google Scholar]
  178. Bernstein, I.M.; Ziegler, W.F.; Leavitt, T.; Badger, G.J. Uterine artery hemodynamic adaptations through the menstrual cycle into early pregnancy. Obstet. Gynecol. 2002, 99, 620–624. [Google Scholar]
  179. Palmer, S.K.; Zamudio, S.; Coffin, C.; Parker, S.; Stamm, E.; Moore, L.G. Quantitative estimation of human uterine artery blood flow and pelvic blood flow redistribution in pregnancy. Obstet. Gynecol. 1992, 80, 1000–1006. [Google Scholar] [PubMed]
  180. Konje, J.C.; Kaufmann, P.; Bell, S.C.; Taylor, D.J. A longitudinal study of quantitative uterine blood flow with the use of color power angiography in appropriate for gestational age pregnancies. Am. J. Obstet. Gynecol. 2001, 185, 608–613. [Google Scholar] [CrossRef]
  181. Magness, R.R.; Phernetton, T.M.; Gibson, T.C.; Chen, D.B. Uterine blood flow responses to ICI 182 780 in ovariectomized oestradiol-17beta-treated, intact follicular and pregnant sheep. J. Physiol. 2005, 565, 71–83. [Google Scholar] [CrossRef] [PubMed]
  182. Rosenfeld, C.R.; Roy, T.; DeSpain, K.; Cox, B.E. Large-conductance Ca2+-dependent K+ channels regulate basal uteroplacental blood flow in ovine pregnancy. J. Soc. Gynecol. Investig. 2005, 12, 402–408. [Google Scholar] [CrossRef] [PubMed]
  183. Rumball, C.W.; Bloomfield, F.H.; Harding, J.E. Cardiovascular adaptations to pregnancy in sheep and effects of periconceptional undernutrition. Placenta 2008, 29, 89–94. [Google Scholar] [CrossRef] [PubMed]
  184. Dowell, R.T.; Kauer, C.D. Uteroplacental blood flow at rest and during exercise in late-gestation conscious rats. J. Appl. Physiol. 1993, 74, 2079–2085. [Google Scholar] [CrossRef]
  185. Hart, M.V.; Hosenpud, J.D.; Hohimer, A.R.; Morton, M.J. Hemodynamics during pregnancy and sex steroid administration in guinea pigs. Am. J. Physiol. 1985, 249, R179–R185. [Google Scholar] [CrossRef]
  186. Peeters, L.L.; Grutters, G.; Martin, C.B., Jr. Distribution of cardiac output in the unstressed pregnant guinea pig. Am. J. Obstet. Gynecol. 1980, 138, 1177–1184. [Google Scholar] [CrossRef] [PubMed]
  187. Rosenfeld, C.R. Distribution of cardiac output in ovine pregnancy. Am. J. Physiol. 1977, 232, H231–H235. [Google Scholar] [CrossRef]
  188. Dowell, R.T.; Kauer, C.D. Maternal hemodynamics and uteroplacental blood flow throughout gestation in conscious rats. Methods Find. Exp. Clin. Pharmacol. 1997, 19, 613–625. [Google Scholar]
  189. Peeters, L.L.; Sparks, J.W.; Grutters, G.; Girard, J.; Battaglia, F.C. Uteroplacental blood flow during pregnancy in chronically catheterized guinea pigs. Pediatr. Res. 1982, 16, 716–720. [Google Scholar] [CrossRef] [Green Version]
  190. Osol, G.; Mandala, M. Maternal uterine vascular remodeling during pregnancy. Physiology 2009, 24, 58–71. [Google Scholar] [CrossRef] [Green Version]
  191. Degner, K.; Magness, R.R.; Shah, D.M. Establishment of the Human Uteroplacental Circulation: A Historical Perspective. Reprod. Sci. 2017, 24, 753–761. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  192. Albrecht, E.D.; Pepe, G.J. Regulation of Uterine Spiral Artery Remodeling: A Review. Reprod. Sci. 2020, 27, 1932–1942. [Google Scholar] [CrossRef] [PubMed]
  193. Xiao, D.; Buchholz, J.N.; Zhang, L. Pregnancy attenuates uterine artery pressure-dependent vascular tone: Role of PKC/ERK pathway. Am. J. Physiol. Heart Circ. Physiol. 2006, 290, H2337–H2343. [Google Scholar] [CrossRef] [PubMed]
  194. Chesley, L.C.; Talledo, E.; Bohler, C.S.; Zuspan, F.P. Vascular Reactivity to Angiotensin Ii and Norepinephrine in Pregnant Women. Am. J. Obstet. Gynecol. 1965, 91, 837–842. [Google Scholar] [CrossRef]
  195. Naden, R.P.; Rosenfeld, C.R. Effect of angiotensin II on uterine and systemic vasculature in pregnant sheep. J. Clin. Investig. 1981, 68, 468–474. [Google Scholar] [CrossRef]
  196. Magness, R.R.; Rosenfeld, C.R. Systemic and uterine responses to alpha-adrenergic stimulation in pregnant and nonpregnant ewes. Am. J. Obstet. Gynecol. 1986, 155, 897–904. [Google Scholar] [CrossRef]
  197. Weiner, C.P.; Thompson, L.P.; Liu, K.Z.; Herrig, J.E. Pregnancy reduces serotonin-induced contraction of guinea pig uterine and carotid arteries. Am. J. Physiol. 1992, 263, H1764–H1769. [Google Scholar] [CrossRef]
  198. Weiner, C.P.; Thompson, L.P.; Liu, K.Z.; Herrig, J.E. Endothelium-derived relaxing factor and indomethacin-sensitive contracting factor alter arterial contractile responses to thromboxane during pregnancy. Am. J. Obstet. Gynecol. 1992, 166, 1171–1178; discussion 1179–1181. [Google Scholar] [CrossRef]
  199. Annibale, D.J.; Rosenfeld, C.R.; Kamm, K.E. Alterations in vascular smooth muscle contractility during ovine pregnancy. Am. J. Physiol. 1989, 256, H1282–H1288. [Google Scholar] [CrossRef]
  200. Yang, D.; Clark, K.E. Effect of endothelin-1 on the uterine vasculature of the pregnant and estrogen-treated nonpregnant sheep. Am. J. Obstet. Gynecol. 1992, 167, 1642–1650. [Google Scholar] [CrossRef]
  201. Hermsteiner, M.; Zoltan, D.R.; Kunzel, W. The vasoconstrictor response of uterine and mesenteric resistance arteries is differentially altered in the course of pregnancy. Eur. J. Obstet. Gynecol. Reprod. Biol. 2001, 100, 29–35. [Google Scholar] [CrossRef] [PubMed]
  202. Mandala, M.; Gokina, N.; Osol, G. Contribution of nonendothelial nitric oxide to altered rat uterine resistance artery serotonin reactivity during pregnancy. Am. J. Obstet. Gynecol. 2002, 187, 463–468. [Google Scholar] [CrossRef] [PubMed]
  203. Xiao, D.; Pearce, W.J.; Zhang, L. Pregnancy enhances endothelium-dependent relaxation of ovine uterine artery: Role of NO and intracellular Ca2+. Am. J. Physiol. Heart Circ. Physiol. 2001, 281, H183–H190. [Google Scholar] [CrossRef] [Green Version]
  204. Gokina, N.I.; Kuzina, O.Y.; Vance, A.M. Augmented EDHF signaling in rat uteroplacental vasculature during late pregnancy. Am. J. Physiol. Heart Circ. Physiol. 2010, 299, H1642–H1652. [Google Scholar] [CrossRef] [PubMed]
  205. Ni, Y.; Meyer, M.; Osol, G. Gestation increases nitric oxide-mediated vasodilation in rat uterine arteries. Am. J. Obstet. Gynecol. 1997, 176, 856–864. [Google Scholar] [CrossRef]
  206. Veerareddy, S.; Cooke, C.L.; Baker, P.N.; Davidge, S.T. Vascular adaptations to pregnancy in mice: Effects on myogenic tone. Am. J. Physiol. Heart Circ. Physiol. 2002, 283, H2226–H2233. [Google Scholar] [CrossRef] [Green Version]
  207. Cooke, C.L.; Davidge, S.T. Pregnancy-induced alterations of vascular function in mouse mesenteric and uterine arteries. Biol. Reprod. 2003, 68, 1072–1077. [Google Scholar] [CrossRef] [Green Version]
  208. Nelson, S.H.; Steinsland, O.S.; Suresh, M.S.; Lee, N.M. Pregnancy augments nitric oxide-dependent dilator response to acetylcholine in the human uterine artery. Hum. Reprod. 1998, 13, 1361–1367. [Google Scholar] [CrossRef] [Green Version]
  209. Simmons, L.A.; Hennessy, A.; Gillin, A.G.; Jeremy, R.W. Uteroplacental blood flow and placental vascular endothelial growth factor in normotensive and pre-eclamptic pregnancy. BJOG 2000, 107, 678–685. [Google Scholar] [CrossRef] [Green Version]
  210. Takata, M.; Nakatsuka, M.; Kudo, T. Differential blood flow in uterine, ophthalmic, and brachial arteries of preeclamptic women. Obstet. Gynecol. 2002, 100, 931–939. [Google Scholar]
  211. Browne, V.A.; Toledo-Jaldin, L.; Davila, R.D.; Lopez, L.P.; Yamashiro, H.; Cioffi-Ragan, D.; Julian, C.G.; Wilson, M.J.; Bigham, A.W.; Shriver, M.D.; et al. High-end arteriolar resistance limits uterine artery blood flow and restricts fetal growth in preeclampsia and gestational hypertension at high altitude. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2011, 300, R1221–R1229. [Google Scholar] [CrossRef]
  212. Verlohren, S.; Niehoff, M.; Hering, L.; Geusens, N.; Herse, F.; Tintu, A.N.; Plagemann, A.; LeNoble, F.; Pijnenborg, R.; Muller, D.N.; et al. Uterine vascular function in a transgenic preeclampsia rat model. Hypertension 2008, 51, 547–553. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  213. Anderson, C.M.; Lopez, F.; Zhang, H.Y.; Pavlish, K.; Benoit, J.N. Reduced uteroplacental perfusion alters uterine arcuate artery function in the pregnant Sprague-Dawley rat. Biol. Reprod. 2005, 72, 762–766. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  214. Luksha, L.; Luksha, N.; Kublickas, M.; Nisell, H.; Kublickiene, K. Diverse mechanisms of endothelium-derived hyperpolarizing factor-mediated dilatation in small myometrial arteries in normal human pregnancy and preeclampsia. Biol. Reprod. 2010, 83, 728–735. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  215. Ashworth, J.R.; Baker, P.N.; Warren, A.Y.; Johnson, I.R. Mechanisms of endothelium-dependent relaxation in myometrial resistance vessels and their alteration in preeclampsia. Hypertens. Pregnancy 1999, 18, 57–71. [Google Scholar] [CrossRef]
  216. Ashworth, J.R.; Warren, A.Y.; Baker, P.N.; Johnson, I.R. Loss of endothelium-dependent relaxation in myometrial resistance arteries in pre-eclampsia. Br. J. Obstet. Gynaecol. 1997, 104, 1152–1158. [Google Scholar] [CrossRef]
  217. Wimalasundera, R.C.; Thom, S.A.; Regan, L.; Hughes, A.D. Effects of vasoactive agents on intracellular calcium and force in myometrial and subcutaneous resistance arteries isolated from preeclamptic, pregnant, and nonpregnant woman. Am. J. Obstet. Gynecol. 2005, 192, 625–632. [Google Scholar] [CrossRef]
  218. Kublickiene, K.R.; Grunewald, C.; Lindblom, B.; Nisell, H. Myogenic and endothelial properties of myometrial resistance arteries from women with preeclampsia. Hypertens. Pregnancy 1998, 17, 271–281. [Google Scholar] [CrossRef]
  219. Kublickiene, K.R.; Lindblom, B.; Kruger, K.; Nisell, H. Preeclampsia: Evidence for impaired shear stress-mediated nitric oxide release in uterine circulation. Am. J. Obstet. Gynecol. 2000, 183, 160–166. [Google Scholar] [CrossRef]
  220. Kenny, L.C.; Baker, P.N.; Kendall, D.A.; Randall, M.D.; Dunn, W.R. The role of gap junctions in mediating endothelium-dependent responses to bradykinin in myometrial small arteries isolated from pregnant women. Br. J. Pharmacol. 2002, 136, 1085–1088. [Google Scholar] [CrossRef] [Green Version]
  221. Reho, J.J.; Toot, J.D.; Peck, J.; Novak, J.; Yun, Y.H.; Ramirez, R.J. Increased Myogenic Reactivity of Uterine Arteries from Pregnant Rats with Reduced Uterine Perfusion Pressure. Pregnancy Hypertens. 2012, 2, 106–114. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  222. Erkkola, R.U.; Pirhonen, J.P. Uterine and umbilical flow velocity waveforms in normotensive and hypertensive subjects during the angiotensin II sensitivity test. Am. J. Obstet. Gynecol. 1992, 166, 910–916. [Google Scholar] [CrossRef] [PubMed]
  223. Gant, N.F.; Daley, G.L.; Chand, S.; Whalley, P.J.; MacDonald, P.C. A study of angiotensin II pressor response throughout primigravid pregnancy. J. Clin. Investig. 1973, 52, 2682–2689. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  224. Stanley, J.L.; Cheung, C.C.; Rueda-Clausen, C.F.; Sankaralingam, S.; Baker, P.N.; Davidge, S.T. Effect of gestational diabetes on maternal artery function. Reprod. Sci. 2011, 18, 342–352. [Google Scholar] [CrossRef]
  225. Chirayath, H.H.; Wareing, M.; Taggart, M.J.; Baker, P.N. Endothelial dysfunction in myometrial arteries of women with gestational diabetes. Diabetes Res. Clin. Pract. 2010, 89, 134–140. [Google Scholar] [CrossRef]
  226. Leiberman, J.R.; Meizner, I.; Mazor, M.; Insler, V. Blood supply to the uterus in preeclampsia. Eur. J. Obstet. Gynecol. Reprod. Biol. 1988, 28, 23–27. [Google Scholar] [CrossRef]
  227. Lunell, N.O.; Sarby, B.; Lewander, R.; Nylund, L. Comparison of uteroplacental blood flow in normal and in intrauterine growth-retarded pregnancy. Measurements with Indium-113m and a computer-linked gammacamera. Gynecol. Obstet. Investig. 1979, 10, 106–118. [Google Scholar] [CrossRef]
  228. Nylund, L.; Lunell, N.O.; Lewander, R.; Persson, B.; Sarby, B. Uteroplacental blood flow in diabetic pregnancy: Measurements with indium 113m and a computer-linked gamma camera. Am. J. Obstet. Gynecol. 1982, 144, 298–302. [Google Scholar] [CrossRef]
  229. Lunell, N.O.; Nylund, L.E.; Lewander, R.; Sarby, B. Uteroplacental blood flow in pre-eclampsia measurements with indium-113m and a computer-linked gamma camera. Clin. Exp. Hypertens. B 1982, 1, 105–117. [Google Scholar] [CrossRef]
  230. Nagar, D.; Liu, X.T.; Rosenfeld, C.R. Estrogen regulates {beta}1-subunit expression in Ca2+-activated K+ channels in arteries from reproductive tissues. Am. J. Physiol. Heart Circ. Physiol. 2005, 289, H1417–H1427. [Google Scholar] [CrossRef]
  231. Rosenfeld, C.R.; Word, R.A.; DeSpain, K.; Liu, X.T. Large conductance Ca2+-activated K+ channels contribute to vascular function in nonpregnant human uterine arteries. Reprod. Sci. 2008, 15, 651–660. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  232. Khan, L.H.; Rosenfeld, C.R.; Liu, X.T.; Magness, R.R. Regulation of the cGMP-cPKG pathway and large-conductance Ca2+-activated K+ channels in uterine arteries during the ovine ovarian cycle. Am. J. Physiol. Endocrinol. Metab. 2010, 298, E222–E228. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  233. Hu, X.Q.; Xiao, D.; Zhu, R.; Huang, X.; Yang, S.; Wilson, S.; Zhang, L. Pregnancy upregulates large-conductance Ca2+-activated K+ channel activity and attenuates myogenic tone in uterine arteries. Hypertension 2011, 58, 1132–1139. [Google Scholar] [CrossRef] [Green Version]
  234. Hu, X.Q.; Xiao, D.; Zhu, R.; Huang, X.; Yang, S.; Wilson, S.M.; Zhang, L. Chronic hypoxia suppresses pregnancy-induced upregulation of large-conductance Ca2+-activated K+ channel activity in uterine arteries. Hypertension 2012, 60, 214–222. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  235. Chen, M.; Dasgupta, C.; Xiong, F.; Zhang, L. Epigenetic upregulation of large-conductance Ca2+-activated K+ channel expression in uterine vascular adaptation to pregnancy. Hypertension 2014, 64, 610–618. [Google Scholar] [CrossRef]
  236. Li, Y.; Bai, J.; Yang, Y.H.; Hoshi, N.; Chen, D.B. Hydrogen Sulfide Relaxes Human Uterine Artery via Activating Smooth Muscle BKCa Channels. Antioxidants 2020, 9, 1127. [Google Scholar] [CrossRef] [PubMed]
  237. Chinnathambi, V.; Blesson, C.S.; Vincent, K.L.; Saade, G.R.; Hankins, G.D.; Yallampalli, C.; Sathishkumar, K. Elevated testosterone levels during rat pregnancy cause hypersensitivity to angiotensin II and attenuation of endothelium-dependent vasodilation in uterine arteries. Hypertension 2014, 64, 405–414. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  238. Choi, S.; Kim, J.A.; Na, H.Y.; Kim, J.E.; Park, S.; Han, K.H.; Kim, Y.J.; Suh, S.H. NADPH oxidase 2-derived superoxide downregulates endothelial KCa3.1 in preeclampsia. Free Radic. Biol. Med. 2013, 57, 10–21. [Google Scholar] [CrossRef]
  239. Rosenbaum, S.T.; Svalo, J.; Nielsen, K.; Larsen, T.; Jorgensen, J.C.; Bouchelouche, P. Immunolocalization and expression of small-conductance calcium-activated potassium channels in human myometrium. J. Cell. Mol. Med. 2012, 16, 3001–3008. [Google Scholar] [CrossRef]
  240. Brereton, M.F.; Wareing, M.; Jones, R.L.; Greenwood, S.L. Characterisation of K+ channels in human fetoplacental vascular smooth muscle cells. PLoS ONE 2013, 8, e57451. [Google Scholar] [CrossRef]
  241. Li, F.F.; He, M.Z.; Xie, Y.; Wu, Y.Y.; Yang, M.T.; Fan, Y.; Qiao, F.Y.; Deng, D.R. Involvement of dysregulated IKCa and SKCa channels in preeclampsia. Placenta 2017, 58, 9–16. [Google Scholar] [CrossRef] [PubMed]
  242. Benkusky, N.A.; Korovkina, V.P.; Brainard, A.M.; England, S.K. Myometrial maxi-K channel beta1 subunit modulation during pregnancy and after 17beta-estradiol stimulation. FEBS Lett. 2002, 524, 97–102. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  243. Salhab, W.A.; Shaul, P.W.; Cox, B.E.; Rosenfeld, C.R. Regulation of types I and III NOS in ovine uterine arteries by daily and acute estrogen exposure. Am. J. Physiol. Heart Circ. Physiol. 2000, 278, H2134–H2142. [Google Scholar] [CrossRef] [PubMed]
  244. Nelson, S.H.; Steinsland, O.S.; Wang, Y.; Yallampalli, C.; Dong, Y.L.; Sanchez, J.M. Increased nitric oxide synthase activity and expression in the human uterine artery during pregnancy. Circ. Res. 2000, 87, 406–411. [Google Scholar] [CrossRef] [Green Version]
  245. Hu, X.Q.; Song, R.; Zhang, L. Effect of Oxidative Stress on the Estrogen-NOS-NO-KCa Channel Pathway in Uteroplacental Dysfunction: Its Implication in Pregnancy Complications. Oxid. Med. Cell. Longev. 2019, 2019, 9194269. [Google Scholar] [CrossRef]
  246. Choi, S.; Kim, J.A.; Li, H.Y.; Lee, S.J.; Seok, Y.S.; Kim, T.H.; Han, K.H.; Park, M.H.; Cho, G.J.; Suh, S.H. Altered Redox State Modulates Endothelial KCa2.3 and KCa3.1 Levels in Normal Pregnancy and Preeclampsia. Antioxid. Redox Signal. 2019, 30, 505–519. [Google Scholar] [CrossRef]
  247. Rahbek, M.; Nazemi, S.; Odum, L.; Gupta, S.; Poulsen, S.S.; Hay-Schmidt, A.; Klaerke, D.A. Expression of the small conductance Ca2+-activated potassium channel subtype 3 (SK3) in rat uterus after stimulation with 17beta-estradiol. PLoS ONE 2014, 9, e87652. [Google Scholar] [CrossRef]
  248. Xiong, W.; Jiang, Y.; Yu, T.; Zheng, Y.; Jiang, L.; Shen, X.; Tang, Y.; Lin, L. Estrogen-regulated expression of SK3 channel in rat colonic smooth muscle contraction. Life Sci. 2020, 263, 118549. [Google Scholar] [CrossRef]
  249. Yap, F.C.; Taylor, M.S.; Lin, M.T. Ovariectomy-induced reductions in endothelial SK3 channel activity and endothelium-dependent vasorelaxation in murine mesenteric arteries. PLoS ONE 2014, 9, e104686. [Google Scholar] [CrossRef] [Green Version]
  250. Choi, S.; Kim, J.A.; Oh, S.; Park, M.H.; Cho, G.J.; Suh, S.H. Internalization and Transportation of Endothelial Cell Surface KCa2.3 and KCa3.1 in Normal Pregnancy and Preeclampsia. Oxid. Med. Cell. Longev. 2019, 2019, 5820839. [Google Scholar] [CrossRef] [Green Version]
  251. Burger, N.Z.; Kuzina, O.Y.; Osol, G.; Gokina, N.I. Estrogen replacement enhances EDHF-mediated vasodilation of mesenteric and uterine resistance arteries: Role of endothelial cell Ca2+. Am. J. Physiol. Endocrinol. Metab. 2009, 296, E503–E512. [Google Scholar] [CrossRef] [PubMed]
  252. Pierce, S.L.; England, S.K. SK3 channel expression during pregnancy is regulated through estrogen and Sp factor-mediated transcriptional control of the KCNN3 gene. Am. J. Physiol. Endocrinol. Metab. 2010, 299, E640–E646. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  253. Hewitt, S.C.; Korach, K.S. Estrogen Receptors: New Directions in the New Millennium. Endocr. Rev. 2018, 39, 664–675. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  254. Zhang, X.Y.; Wang, S.; Yan, Z.; Zhang, Y.Q.; Wan, Y.; Zhang, B.; Wang, L.F.; Chai, Y.B.; Wei, J.G. Promoter cloning and characterization of the rabbit BK channel beta1 subunit gene. Gene 2009, 438, 33–39. [Google Scholar] [CrossRef]
  255. Lyko, F. The DNA methyltransferase family: A versatile toolkit for epigenetic regulation. Nat. Rev. Genet. 2018, 19, 81–92. [Google Scholar] [CrossRef] [PubMed]
  256. Hu, X.Q.; Dasgupta, C.; Chen, M.; Xiao, D.; Huang, X.; Han, L.; Yang, S.; Xu, Z.; Zhang, L. Pregnancy Reprograms Large-Conductance Ca2+-Activated K+ Channel in Uterine Arteries: Roles of Ten-Eleven Translocation Methylcytosine Dioxygenase 1-Mediated Active Demethylation. Hypertension 2017, 69, 1181–1191. [Google Scholar] [CrossRef]
  257. Jacobson, D.; Pribnow, D.; Herson, P.S.; Maylie, J.; Adelman, J.P. Determinants contributing to estrogen-regulated expression of SK3. Biochem. Biophys. Res. Commun. 2003, 303, 660–668. [Google Scholar] [CrossRef]
  258. Robb, V.A.; Pepe, G.J.; Albrecht, E.D. Acute temporal regulation of placental vascular endothelial growth/permeability factor expression in baboons by estrogen. Biol. Reprod. 2004, 71, 1694–1698. [Google Scholar] [CrossRef]
  259. Zhu, R.; Huang, X.; Hu, X.Q.; Xiao, D.; Zhang, L. Gestational hypoxia increases reactive oxygen species and inhibits steroid hormone-mediated upregulation of Ca2+-activated K+ channel function in uterine arteries. Hypertension 2014, 64, 415–422. [Google Scholar] [CrossRef] [Green Version]
  260. Rosenfeld, C.R.; Cornfield, D.N.; Roy, T. Ca2+-activated K+ channels modulate basal and E(2)beta-induced rises in uterine blood flow in ovine pregnancy. Am. J. Physiol. Heart Circ. Physiol. 2001, 281, H422–H431. [Google Scholar] [CrossRef]
  261. Song, R.; Hu, X.Q.; Romero, M.; Holguin, M.A.; Kagabo, W.; Xiao, D.; Wilson, S.M.; Zhang, L. Ryanodine receptor subtypes regulate Ca2+ sparks/spontaneous transient outward currents and myogenic tone of uterine arteries in pregnancy. Cardiovasc. Res. 2021, 117, 792–804. [Google Scholar] [CrossRef] [PubMed]
  262. Xiao, D.; Zhu, R.; Zhang, L. Gestational hypoxia up-regulates protein kinase C and inhibits calcium-activated potassium channels in ovine uterine arteries. Int. J. Med. Sci. 2014, 11, 886–892. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  263. Zhang, H.; Zhang, L. Role of protein kinase C isozymes in the regulation of alpha1-adrenergic receptor-mediated contractions in ovine uterine arteries. Biol. Reprod. 2008, 78, 35–42. [Google Scholar] [CrossRef] [Green Version]
  264. Goulopoulou, S.; Hannan, J.L.; Matsumoto, T.; Webb, R.C. Pregnancy reduces RhoA/Rho kinase and protein kinase C signaling pathways downstream of thromboxane receptor activation in the rat uterine artery. Am. J. Physiol. Heart Circ. Physiol. 2012, 302, H2477–H2488. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  265. Magness, R.R.; Rosenfeld, C.R.; Carr, B.R. Protein kinase C in uterine and systemic arteries during ovarian cycle and pregnancy. Am. J. Physiol. 1991, 260, E464–E470. [Google Scholar] [CrossRef] [PubMed]
  266. Ross, G.R.; Yallampalli, U.; Gangula, P.R.; Reed, L.; Sathishkumar, K.; Gao, H.; Chauhan, M.; Yallampalli, C. Adrenomedullin relaxes rat uterine artery: Mechanisms and influence of pregnancy and estradiol. Endocrinology 2010, 151, 4485–4493. [Google Scholar] [CrossRef]
  267. Gangula, P.R.; Thota, C.; Wimalawansa, S.J.; Bukoski, R.D.; Yallampalli, C. Mechanisms involved in calcitonin gene-related Peptide-induced relaxation in pregnant rat uterine artery. Biol. Reprod. 2003, 69, 1635–1641. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  268. Rosenfeld, C.R.; Hynan, L.S.; Liu, X.T.; Roy, T. Large conductance Ca2+-activated K+ channels modulate uterine alpha1-adrenergic sensitivity in ovine pregnancy. Reprod. Sci. 2014, 21, 456–464. [Google Scholar] [CrossRef] [Green Version]
  269. Goulopoulou, S.; Hannan, J.; Matsumoto, T.; Webb, R.C. Pregnancy regulates thromboxane A(2)-induced contractions via endothelium-derived factors and large-conductance calcium-activated potassium channels in rat uterine artery. FASEB J. 2013, 27, 877.7. [Google Scholar] [CrossRef]
  270. Jaggar, J.H.; Porter, V.A.; Lederer, W.J.; Nelson, M.T. Calcium sparks in smooth muscle. Am. J. Physiol. Cell Physiol. 2000, 278, C235–C256. [Google Scholar] [CrossRef] [Green Version]
  271. Hu, X.Q.; Song, R.; Romero, M.; Dasgupta, C.; Huang, X.; Holguin, M.A.; Williams, V.; Xiao, D.; Wilson, S.M.; Zhang, L. Pregnancy Increases Ca2+ Sparks/Spontaneous Transient Outward Currents and Reduces Uterine Arterial Myogenic Tone. Hypertension 2019, 73, 691–702. [Google Scholar] [CrossRef] [PubMed]
  272. Xiao, D.; Liu, Y.; Pearce, W.J.; Zhang, L. Endothelial nitric oxide release in isolated perfused ovine uterine arteries: Effect of pregnancy. Eur. J. Pharmacol. 1999, 367, 223–230. [Google Scholar] [CrossRef] [PubMed]
  273. Sheibani, L.; Lechuga, T.J.; Zhang, H.; Hameed, A.; Wing, D.A.; Kumar, S.; Rosenfeld, C.R.; Chen, D.B. Augmented H2S production via cystathionine-beta-synthase upregulation plays a role in pregnancy-associated uterine vasodilation. Biol. Reprod. 2017, 96, 664–672. [Google Scholar] [CrossRef] [PubMed]
  274. Lechuga, T.J.; Qi, Q.R.; Magness, R.R.; Chen, D.B. Ovine uterine artery hydrogen sulfide biosynthesis in vivo: Effects of ovarian cycle and pregnancydagger. Biol. Reprod. 2019, 100, 1630–1636. [Google Scholar] [CrossRef] [PubMed]
  275. Gillham, J.C.; Myers, J.E.; Baker, P.N.; Taggart, M.J. Regulation of endothelial-dependent relaxation in human systemic arteries by SKCa and IKCa channels. Reprod. Sci. 2007, 14, 43–50. [Google Scholar] [CrossRef]
  276. Garland, C.J.; Dora, K.A. EDH: Endothelium-dependent hyperpolarization and microvascular signalling. Acta Physiol. 2017, 219, 152–161. [Google Scholar] [CrossRef] [PubMed]
  277. Senadheera, S.; Bertrand, P.P.; Grayson, T.H.; Leader, L.; Murphy, T.V.; Sandow, S.L. Pregnancy-induced remodelling and enhanced endothelium-derived hyperpolarization-type vasodilator activity in rat uterine radial artery: Transient receptor potential vanilloid type 4 channels, caveolae and myoendothelial gap junctions. J. Anat. 2013, 223, 677–686. [Google Scholar] [CrossRef]
  278. Meziani, F.; Van Overloop, B.; Schneider, F.; Gairard, A. Parathyroid hormone-related protein-induced relaxation of rat uterine arteries: Influence of the endothelium during gestation. J. Soc. Gynecol. Investig. 2005, 12, 14–19. [Google Scholar] [CrossRef]
  279. Kostrzewska, A.; Modzelewska, B.; Kleszczewski, T.; Batra, S. Effect of nitric oxide on responses of the human uterine arteries to vasopressin. Vascul. Pharmacol. 2008, 48, 9–13. [Google Scholar] [CrossRef]
  280. Li, F.; Xie, Y.; He, M.; Fan, Y.; Yang, M.; Wang, S.; Li, X.; Sun, Y.; Xu, H.; Liu, X.; et al. IkCa and SKCa might participate in preeclampsia through regulating placental angiogenesis. Pregnancy Hypertens. 2020, 21, 90–95. [Google Scholar] [CrossRef]
  281. Rada, C.C.; Pierce, S.L.; Nuno, D.W.; Zimmerman, K.; Lamping, K.G.; Bowdler, N.C.; Weiss, R.M.; England, S.K. Overexpression of the SK3 channel alters vascular remodeling during pregnancy, leading to fetal demise. Am. J. Physiol. Endocrinol. Metab. 2012, 303, E825–E831. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  282. Rada, C.C.; Murray, G.; England, S.K. The SK3 channel promotes placental vascularization by enhancing secretion of angiogenic factors. Am. J. Physiol. Endocrinol. Metab. 2014, 307, E935–E943. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  283. Chen, J.; Gao, Q.; Jiang, L.; Feng, X.; Zhu, X.; Fan, X.; Mao, C.; Xu, Z. The NOX2-derived reactive oxygen species damaged endothelial nitric oxide system via suppressed BKCa/SKCa in preeclampsia. Hypertens. Res. 2017, 40, 457–464. [Google Scholar] [CrossRef] [PubMed]
  284. Hu, X.Q.; Dasgupta, C.; Xiao, D.; Huang, X.; Yang, S.; Zhang, L. MicroRNA-210 Targets Ten-Eleven Translocation Methylcytosine Dioxygenase 1 and Suppresses Pregnancy-Mediated Adaptation of Large Conductance Ca2+-Activated K+ Channel Expression and Function in Ovine Uterine Arteries. Hypertension 2017, 70, 601–612. [Google Scholar] [CrossRef] [PubMed]
  285. Sun, Y.; Wang, C.; Zhang, N.; Liu, F. Melatonin ameliorates hypertension in hypertensive pregnant mice and suppresses the hypertension-induced decrease in Ca2+-activated K+ channels in uterine arteries. Hypertens. Res. 2021, 44, 1079–1086. [Google Scholar] [CrossRef]
  286. Laivuori, H.; Kaaja, R.; Rutanen, E.M.; Viinikka, L.; Ylikorkala, O. Evidence of high circulating testosterone in women with prior preeclampsia. J. Clin. Endocrinol. Metab. 1998, 83, 344–347. [Google Scholar] [CrossRef] [PubMed]
  287. Salamalekis, E.; Bakas, P.; Vitoratos, N.; Eleptheriadis, M.; Creatsas, G. Androgen levels in the third trimester of pregnancy in patients with preeclampsia. Eur. J. Obstet. Gynecol. Reprod. Biol. 2006, 126, 16–19. [Google Scholar] [CrossRef] [PubMed]
  288. Acromite, M.T.; Mantzoros, C.S.; Leach, R.E.; Hurwitz, J.; Dorey, L.G. Androgens in preeclampsia. Am. J. Obstet. Gynecol. 1999, 180, 60–63. [Google Scholar] [CrossRef]
  289. Mateev, S.; Sillau, A.H.; Mouser, R.; McCullough, R.E.; White, M.M.; Young, D.A.; Moore, L.G. Chronic hypoxia opposes pregnancy-induced increase in uterine artery vasodilator response to flow. Am. J. Physiol. Heart Circ. Physiol. 2003, 284, H820–H829. [Google Scholar] [CrossRef] [Green Version]
  290. Li, H.; An, J.R.; Seo, M.S.; Kang, M.; Heo, R.; Park, S.; Mun, S.Y.; Bae, Y.M.; Han, E.T.; Han, J.H.; et al. Downregulation of large-conductance Ca2+-activated K+ channels in human umbilical arterial smooth muscle cells in gestational diabetes mellitus. Life Sci. 2022, 288, 120169. [Google Scholar] [CrossRef]
  291. Gokina, N.I.; Bonev, A.D.; Gokin, A.P.; Goloman, G. Role of impaired endothelial cell Ca2+ signaling in uteroplacental vascular dysfunction during diabetic rat pregnancy. Am. J. Physiol. Heart Circ. Physiol. 2013, 304, H935–H945. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  292. Gokina, N.I.; Kuzina, O.Y.; Vance, A.M.; Phillips, J.K. Diabetic pregnancy impairs EDHF-dependent uteroplacental vasodilation mediated by intermediate-conductance Ca2+-activated K+ channels. FASEB J. 2010, 24, 976.13. [Google Scholar] [CrossRef]
  293. Gokina, N.I.; Bonev, A.D.; Phillips, J.; Gokin, A.P.; Veilleux, K.; Oppenheimer, K.; Goloman, G. Impairment of IKCa channels contributes to uteroplacental endothelial dysfunction in rat diabetic pregnancy. Am. J. Physiol. Heart Circ. Physiol. 2015, 309, H592–H604. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  294. Stanley, J.L.; Ashton, N.; Taggart, M.J.; Davidge, S.T.; Baker, P.N. Uterine artery function in a mouse model of pregnancy complicated by diabetes. Vascul. Pharmacol. 2009, 50, 8–13. [Google Scholar] [CrossRef] [PubMed]
  295. Ducsay, C.A.; Goyal, R.; Pearce, W.J.; Wilson, S.; Hu, X.Q.; Zhang, L. Gestational Hypoxia and Developmental Plasticity. Physiol. Rev. 2018, 98, 1241–1334. [Google Scholar] [CrossRef]
  296. Hu, X.Q.; Zhang, L. Hypoxia and the integrated stress response promote pulmonary hypertension and preeclampsia: Implications in drug development. Drug Discov. Today 2021, 26, 2754–2773. [Google Scholar] [CrossRef]
  297. Zamudio, S.; Wu, Y.; Ietta, F.; Rolfo, A.; Cross, A.; Wheeler, T.; Post, M.; Illsley, N.P.; Caniggia, I. Human placental hypoxia-inducible factor-1alpha expression correlates with clinical outcomes in chronic hypoxia in vivo. Am. J. Pathol. 2007, 170, 2171–2179. [Google Scholar] [CrossRef] [Green Version]
  298. Rolfo, A.; Many, A.; Racano, A.; Tal, R.; Tagliaferro, A.; Ietta, F.; Wang, J.; Post, M.; Caniggia, I. Abnormalities in oxygen sensing define early and late onset preeclampsia as distinct pathologies. PLoS ONE 2010, 5, e13288. [Google Scholar] [CrossRef] [Green Version]
  299. Zhou, J.; Xiao, D.; Hu, Y.; Wang, Z.; Paradis, A.; Mata-Greenwood, E.; Zhang, L. Gestational hypoxia induces preeclampsia-like symptoms via heightened endothelin-1 signaling in pregnant rats. Hypertension 2013, 62, 599–607. [Google Scholar] [CrossRef] [Green Version]
  300. Xiao, D.; Hu, X.Q.; Huang, X.; Zhou, J.; Wilson, S.M.; Yang, S.; Zhang, L. Chronic hypoxia during gestation enhances uterine arterial myogenic tone via heightened oxidative stress. PLoS ONE 2013, 8, e73731. [Google Scholar] [CrossRef] [Green Version]
  301. Chandel, N.S.; McClintock, D.S.; Feliciano, C.E.; Wood, T.M.; Melendez, J.A.; Rodriguez, A.M.; Schumacker, P.T. Reactive oxygen species generated at mitochondrial complex III stabilize hypoxia-inducible factor-1alpha during hypoxia: A mechanism of O2 sensing. J. Biol. Chem. 2000, 275, 25130–25138. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  302. Favaro, E.; Ramachandran, A.; McCormick, R.; Gee, H.; Blancher, C.; Crosby, M.; Devlin, C.; Blick, C.; Buffa, F.; Li, J.L.; et al. MicroRNA-210 regulates mitochondrial free radical response to hypoxia and krebs cycle in cancer cells by targeting iron sulfur cluster protein ISCU. PLoS ONE 2010, 5, e10345. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  303. Watson, C.J.; Collier, P.; Tea, I.; Neary, R.; Watson, J.A.; Robinson, C.; Phelan, D.; Ledwidge, M.T.; McDonald, K.M.; McCann, A.; et al. Hypoxia-induced epigenetic modifications are associated with cardiac tissue fibrosis and the development of a myofibroblast-like phenotype. Hum. Mol. Genet. 2014, 23, 2176–2188. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  304. Zamudio, S.; Palmer, S.K.; Dahms, T.E.; Berman, J.C.; Young, D.A.; Moore, L.G. Alterations in uteroplacental blood flow precede hypertension in preeclampsia at high altitude. J. Appl. Physiol. 1995, 79, 15–22. [Google Scholar] [CrossRef]
  305. Zamudio, S.; Palmer, S.K.; Droma, T.; Stamm, E.; Coffin, C.; Moore, L.G. Effect of altitude on uterine artery blood flow during normal pregnancy. J. Appl. Physiol. 1995, 79, 7–14. [Google Scholar] [CrossRef]
  306. Keyes, L.E.; Armaza, J.F.; Niermeyer, S.; Vargas, E.; Young, D.A.; Moore, L.G. Intrauterine growth restriction, preeclampsia, and intrauterine mortality at high altitude in Bolivia. Pediatr. Res. 2003, 54, 20–25. [Google Scholar] [CrossRef]
  307. Julian, C.G.; Galan, H.L.; Wilson, M.J.; Desilva, W.; Cioffi-Ragan, D.; Schwartz, J.; Moore, L.G. Lower uterine artery blood flow and higher endothelin relative to nitric oxide metabolite levels are associated with reductions in birth weight at high altitude. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2008, 295, R906–R915. [Google Scholar] [CrossRef] [Green Version]
  308. Shimoda, L.A.; Polak, J. Hypoxia. 4. Hypoxia and ion channel function. Am. J. Physiol. Cell Physiol. 2011, 300, C951–C967. [Google Scholar] [CrossRef]
  309. Gebert, L.F.R.; MacRae, I.J. Regulation of microRNA function in animals. Nat. Rev. Mol. Cell. Biol. 2019, 20, 21–37. [Google Scholar] [CrossRef]
  310. Pineles, B.L.; Romero, R.; Montenegro, D.; Tarca, A.L.; Han, Y.M.; Kim, Y.M.; Draghici, S.; Espinoza, J.; Kusanovic, J.P.; Mittal, P.; et al. Distinct subsets of microRNAs are expressed differentially in the human placentas of patients with preeclampsia. Am. J. Obstet. Gynecol. 2007, 196, 261.e1–261.e6. [Google Scholar] [CrossRef]
  311. Colleoni, F.; Padmanabhan, N.; Yung, H.W.; Watson, E.D.; Cetin, I.; Tissot van Patot, M.C.; Burton, G.J.; Murray, A.J. Suppression of mitochondrial electron transport chain function in the hypoxic human placenta: A role for miRNA-210 and protein synthesis inhibition. PLoS ONE 2013, 8, e55194. [Google Scholar] [CrossRef] [PubMed]
  312. Anton, L.; Olarerin-George, A.O.; Schwartz, N.; Srinivas, S.; Bastek, J.; Hogenesch, J.B.; Elovitz, M.A. miR-210 inhibits trophoblast invasion and is a serum biomarker for preeclampsia. Am. J. Pathol. 2013, 183, 1437–1445. [Google Scholar] [CrossRef] [Green Version]
  313. Hu, X.Q.; Dasgupta, C.; Song, R.; Romero, M.; Wilson, S.M.; Zhang, L. MicroRNA-210 Mediates Hypoxia-Induced Repression of Spontaneous Transient Outward Currents in Sheep Uterine Arteries During Gestation. Hypertension 2021, 77, 1412–1427. [Google Scholar] [CrossRef]
  314. Greenberg, M.V.C.; Bourc’his, D. The diverse roles of DNA methylation in mammalian development and disease. Nat. Rev. Mol. Cell. Biol. 2019, 20, 590–607. [Google Scholar] [CrossRef] [PubMed]
  315. Wu, X.; Zhang, Y. TET-mediated active DNA demethylation: Mechanism, function and beyond. Nat. Rev. Genet. 2017, 18, 517–534. [Google Scholar] [CrossRef] [PubMed]
  316. Hu, X.Q.; Chen, M.; Dasgupta, C.; Xiao, D.; Huang, X.; Yang, S.; Zhang, L. Chronic hypoxia upregulates DNA methyltransferase and represses large conductance Ca2+-activated K+ channel function in ovine uterine arteries. Biol. Reprod. 2017, 96, 424–434. [Google Scholar] [CrossRef] [Green Version]
  317. Hu, X.Q.; Dasgupta, C.; Xiao, J.; Yang, S.; Zhang, L. Long-term high altitude hypoxia during gestation suppresses large conductance Ca2+-activated K+ channel function in uterine arteries: A causal role for microRNA-210. J. Physiol. 2018, 596, 5891–5906. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  318. Dasgupta, C.; Chen, M.; Zhang, H.; Yang, S.; Zhang, L. Chronic hypoxia during gestation causes epigenetic repression of the estrogen receptor-alpha gene in ovine uterine arteries via heightened promoter methylation. Hypertension 2012, 60, 697–704. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  319. Chang, K.; Xiao, D.; Huang, X.; Xue, Z.; Yang, S.; Longo, L.D.; Zhang, L. Chronic hypoxia inhibits sex steroid hormone-mediated attenuation of ovine uterine arterial myogenic tone in pregnancy. Hypertension 2010, 56, 750–757. [Google Scholar] [CrossRef] [Green Version]
  320. Chen, M.; Xiao, D.; Hu, X.Q.; Dasgupta, C.; Yang, S.; Zhang, L. Hypoxia Represses ER-alpha Expression and Inhibits Estrogen-Induced Regulation of Ca2+-Activated K+ Channel Activity and Myogenic Tone in Ovine Uterine Arteries: Causal Role of DNA Methylation. Hypertension 2015, 66, 44–51. [Google Scholar] [CrossRef] [Green Version]
  321. Phoswa, W.N.; Khaliq, O.P. The Role of Oxidative Stress in Hypertensive Disorders of Pregnancy (Preeclampsia, Gestational Hypertension) and Metabolic Disorder of Pregnancy (Gestational Diabetes Mellitus). Oxid. Med. Cell. Longev. 2021, 2021, 5581570. [Google Scholar] [CrossRef]
  322. Griendling, K.K.; Camargo, L.L.; Rios, F.J.; Alves-Lopes, R.; Montezano, A.C.; Touyz, R.M. Oxidative Stress and Hypertension. Circ. Res. 2021, 128, 993–1020. [Google Scholar] [CrossRef] [PubMed]
  323. Vaka, V.R.; McMaster, K.M.; Cunningham, M.W., Jr.; Ibrahim, T.; Hazlewood, R.; Usry, N.; Cornelius, D.C.; Amaral, L.M.; LaMarca, B. Role of Mitochondrial Dysfunction and Reactive Oxygen Species in Mediating Hypertension in the Reduced Uterine Perfusion Pressure Rat Model of Preeclampsia. Hypertension 2018, 72, 703–711. [Google Scholar] [CrossRef] [PubMed]
  324. Hu, X.Q.; Song, R.; Dasgupta, C.; Romero, M.; Juarez, R.; Hanson, J.; Blood, A.B.; Wilson, S.M.; Zhang, L. MicroRNA-210-mediated mtROS confer hypoxia-induced suppression of STOCs in ovine uterine arteries. Br. J. Pharmacol. 2022, 179, 4640–4654. [Google Scholar] [CrossRef] [PubMed]
  325. Tang, X.D.; Garcia, M.L.; Heinemann, S.H.; Hoshi, T. Reactive oxygen species impair Slo1 BK channel function by altering cysteine-mediated calcium sensing. Nat. Struct. Mol. Biol. 2004, 11, 171–178. [Google Scholar] [CrossRef]
  326. Hu, X.Q.; Huang, X.; Xiao, D.; Zhang, L. Direct effect of chronic hypoxia in suppressing large conductance Ca2+-activated K+ channel activity in ovine uterine arteries via increasing oxidative stress. J. Physiol. 2016, 594, 343–356. [Google Scholar] [CrossRef]
  327. Lu, T.; Chai, Q.; Yu, L.; d’Uscio, L.V.; Katusic, Z.S.; He, T.; Lee, H.C. Reactive oxygen species signaling facilitates FOXO-3a/FBXO-dependent vascular BK channel beta1 subunit degradation in diabetic mice. Diabetes 2012, 61, 1860–1868. [Google Scholar] [CrossRef] [Green Version]
  328. Wang, Y.; Wang, X.J.; Zhao, L.M.; Pang, Z.D.; She, G.; Song, Z.; Cheng, X.; Du, X.J.; Deng, X.L. Oxidative stress induced by palmitic acid modulates KCa2.3 channels in vascular endothelium. Exp. Cell Res. 2019, 383, 111552. [Google Scholar] [CrossRef]
  329. Zhao, L.; Wang, Y.; Ma, X.; Wang, Y.; Deng, X. Oxidative stress impairs IKCa- and SKCa-mediated vasodilatation in mesenteric arteries from diabetic rats. Nan Fang Yi Ke Da Xue Xue Bao 2013, 33, 939–944. [Google Scholar]
  330. Lopes, R.A.; Neves, K.B.; Pestana, C.R.; Queiroz, A.L.; Zanotto, C.Z.; Chignalia, A.Z.; Valim, Y.M.; Silveira, L.R.; Curti, C.; Tostes, R.C. Testosterone induces apoptosis in vascular smooth muscle cells via extrinsic apoptotic pathway with mitochondria-generated reactive oxygen species involvement. Am. J. Physiol. Heart Circ. Physiol. 2014, 306, H1485–H1494. [Google Scholar] [CrossRef] [Green Version]
  331. Mishra, J.S.; Blesson, C.S.; Kumar, S. Testosterone Decreases Placental Mitochondrial Content and Cellular Bioenergetics. Biology 2020, 9, 176. [Google Scholar] [CrossRef] [PubMed]
  332. Xing, H.; Zhang, Z.; Shi, G.; He, Y.; Song, Y.; Liu, Y.; Harrington, E.O.; Sellke, F.W.; Feng, J. Chronic Inhibition of mROS Protects Against Coronary Endothelial Dysfunction in Mice With Diabetes. Front. Cell Dev. Biol. 2021, 9, 643810. [Google Scholar] [CrossRef] [PubMed]
  333. Burton, G.J.; Yung, H.W.; Cindrova-Davies, T.; Charnock-Jones, D.S. Placental endoplasmic reticulum stress and oxidative stress in the pathophysiology of unexplained intrauterine growth restriction and early onset preeclampsia. Placenta 2009, 30 (Suppl. A), S43–S48. [Google Scholar] [CrossRef] [Green Version]
  334. Yung, H.W.; Calabrese, S.; Hynx, D.; Hemmings, B.A.; Cetin, I.; Charnock-Jones, D.S.; Burton, G.J. Evidence of placental translation inhibition and endoplasmic reticulum stress in the etiology of human intrauterine growth restriction. Am. J. Pathol. 2008, 173, 451–462. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  335. Yung, H.W.; Hemberger, M.; Watson, E.D.; Senner, C.E.; Jones, C.P.; Kaufman, R.J.; Charnock-Jones, D.S.; Burton, G.J. Endoplasmic reticulum stress disrupts placental morphogenesis: Implications for human intrauterine growth restriction. J. Pathol. 2012, 228, 554–564. [Google Scholar] [CrossRef] [Green Version]
  336. Yung, H.W.; Alnaes-Katjavivi, P.; Jones, C.J.; El-Bacha, T.; Golic, M.; Staff, A.C.; Burton, G.J. Placental endoplasmic reticulum stress in gestational diabetes: The potential for therapeutic intervention with chemical chaperones and antioxidants. Diabetologia 2016, 59, 2240–2250. [Google Scholar] [CrossRef]
  337. Hu, X.Q.; Song, R.; Romero, M.; Dasgupta, C.; Min, J.; Hatcher, D.; Xiao, D.; Blood, A.; Wilson, S.M.; Zhang, L. Gestational Hypoxia Inhibits Pregnancy-Induce.ed Upregulation of Ca2+ Sparks and Spontaneous Transient Outward Currents in Uterine Arteries Via Heightened Endoplasmic Reticulum/Oxidative Stress. Hypertension 2020, 76, 930–942. [Google Scholar] [CrossRef]
  338. Yung, H.W.; Cox, M.; Tissot van Patot, M.; Burton, G.J. Evidence of endoplasmic reticulum stress and protein synthesis inhibition in the placenta of non-native women at high altitude. FASEB J. 2012, 26, 1970–1981. [Google Scholar] [CrossRef]
  339. Sun, W.T.; Wang, X.C.; Mak, S.K.; He, G.W.; Liu, X.C.; Underwood, M.J.; Yang, Q. Activation of PERK branch of ER stress mediates homocysteine-induced BKCa channel dysfunction in coronary artery via FoxO3a-dependent regulation of atrogin-1. Oncotarget 2017, 8, 51462–51477. [Google Scholar] [CrossRef] [Green Version]
  340. Wang, X.C.; Sun, W.T.; Yu, C.M.; Pun, S.H.; Underwood, M.J.; He, G.W.; Yang, Q. ER stress mediates homocysteine-induced endothelial dysfunction: Modulation of IKCa and SKCa channels. Atherosclerosis 2015, 242, 191–198. [Google Scholar] [CrossRef] [Green Version]
  341. Haller, H.; Hempel, A.; Homuth, V.; Mandelkow, A.; Busjahn, A.; Maasch, C.; Drab, M.; Lindschau, C.; Jupner, A.; Vetter, K.; et al. Endothelial-cell permeability and protein kinase C in pre-eclampsia. Lancet 1998, 351, 945–949. [Google Scholar] [CrossRef] [PubMed]
  342. Jiang, R.; Teng, Y.; Huang, Y.; Gu, J.; Li, M. Protein kinase C-alpha activation induces NF-kB-dependent VCAM-1 expression in cultured human umbilical vein endothelial cells treated with sera from preeclamptic patients. Gynecol. Obstet. Investig. 2010, 69, 101–108. [Google Scholar] [CrossRef] [PubMed]
  343. Chang, K.; Xiao, D.; Huang, X.; Longo, L.D.; Zhang, L. Chronic hypoxia increases pressure-dependent myogenic tone of the uterine artery in pregnant sheep: Role of ERK/PKC pathway. Am. J. Physiol. Heart Circ. Physiol. 2009, 296, H1840–H1849. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  344. McCarthy, F.P.; Drewlo, S.; English, F.A.; Kingdom, J.; Johns, E.J.; Kenny, L.C.; Walsh, S.K. Evidence implicating peroxisome proliferator-activated receptor-gamma in the pathogenesis of preeclampsia. Hypertension 2011, 58, 882–887. [Google Scholar] [CrossRef] [Green Version]
  345. Ketsawatsomkron, P.; Lorca, R.A.; Keen, H.L.; Weatherford, E.T.; Liu, X.; Pelham, C.J.; Grobe, J.L.; Faraci, F.M.; England, S.K.; Sigmund, C.D. PPARgamma regulates resistance vessel tone through a mechanism involving RGS5-mediated control of protein kinase C and BKCa channel activity. Circ. Res. 2012, 111, 1446–1458. [Google Scholar] [CrossRef]
  346. Gokina, N.I.; Chan, S.L.; Chapman, A.C.; Oppenheimer, K.; Jetton, T.L.; Cipolla, M.J. Inhibition of PPARgamma during rat pregnancy causes intrauterine growth restriction and attenuation of uterine vasodilation. Front. Physiol. 2013, 4, 184. [Google Scholar] [CrossRef]
Figure 1. Ca2+-activated K+ (KCa) channel topology and structure. (A) Membrane topology of the BKCa channel α subunit. The α subunit contains the S0 segment, voltage sensing domain (S1–S4 segments), pore gate domain (S5 and S6 segments), and cytosolic domain containing RCK1 and RCK2. Ca2+ sensitivity is conferred by binding of Ca2+ to RCK1 and RCK2. (B) Membrane topology of BKCa channel β and γ subunits. The β subunit is comprised of two transmembrane domains, whereas the γ subunit possesses only one transmembrane domain with six leucine-rich repeat segments in its extracellular domain. (C) Membrane topology of SKCa and IKCa channels. Both SKCa and IKCa channels contain six transmembrane domains. Ca2+ sensitivity is conferred by constitutively bound calmodulin (CaM) to the intracellular C-terminus. (D) KCa channels are either heterotetrameric (BKCa) or homotetrameric (SKCa and IKCa) assemblies of subunits.
Figure 1. Ca2+-activated K+ (KCa) channel topology and structure. (A) Membrane topology of the BKCa channel α subunit. The α subunit contains the S0 segment, voltage sensing domain (S1–S4 segments), pore gate domain (S5 and S6 segments), and cytosolic domain containing RCK1 and RCK2. Ca2+ sensitivity is conferred by binding of Ca2+ to RCK1 and RCK2. (B) Membrane topology of BKCa channel β and γ subunits. The β subunit is comprised of two transmembrane domains, whereas the γ subunit possesses only one transmembrane domain with six leucine-rich repeat segments in its extracellular domain. (C) Membrane topology of SKCa and IKCa channels. Both SKCa and IKCa channels contain six transmembrane domains. Ca2+ sensitivity is conferred by constitutively bound calmodulin (CaM) to the intracellular C-terminus. (D) KCa channels are either heterotetrameric (BKCa) or homotetrameric (SKCa and IKCa) assemblies of subunits.
Ijms 24 01349 g001
Figure 2. Regulation of vascular function by cross-talks among ion channels. The BKCa channel is preferentially expressed in vascular smooth muscle cells (VSMCs), whereas SKCa and IKCa channels are primarily expressed in endothelial cells (ECs). Vasoconstriction is triggered by an increase in intracellular Ca2+ ([Ca2+]i) in VSMCs due to Ca2+ release from the sarcoplasmic reticulum (SR) and/or Ca2+ influx through the CaV 1.2 channel in the plasma membrane. Vasoconstriction is counteracted by activities of SKCa/IKCa channels in ECs and BKCa channels in VSMCs. The activation of SKCa and IKCa channels in ECs triggered by inositol triphosphate receptor (IP3R)-mediated Ca2+ release and/or transient receptor potential (TRP) channel-mediated Ca2+ influx promotes hyperpolarization and release of nitric oxide (NO)/endothelium-derived hyperpolarizing factor (EDRF). In addition, hyperpolarization in ECs could be transmitted to VSMCs via myoendothelial gag junctions (MEGJs). The BKCa channel is activated by RyR-mediated Ca2+ sparks in VSMCs. BKCa channel activity is also subject to regulation by EC-derived NO and EDHF. Moreover, the accumulation of K+ ions in the intercellular space hyperpolarizes VSMCs by activating the inwardly rectifying K+ (Kir) channel. Overall, these events promote hyperpolarization of VSMCs, which in turn leads to the closure of the CaV 1.2 channel and subsequent vasodilation. ACh, acetylcholine; BK, bradykinin; SP, substance P; GPCR, G protein-coupled receptor; PLC, phospholipase C; NOS, nitric oxide synthases; GC, guanylate cyclase; cGMP, cyclic guanosine monophosphate; PKG, protein kinase G.
Figure 2. Regulation of vascular function by cross-talks among ion channels. The BKCa channel is preferentially expressed in vascular smooth muscle cells (VSMCs), whereas SKCa and IKCa channels are primarily expressed in endothelial cells (ECs). Vasoconstriction is triggered by an increase in intracellular Ca2+ ([Ca2+]i) in VSMCs due to Ca2+ release from the sarcoplasmic reticulum (SR) and/or Ca2+ influx through the CaV 1.2 channel in the plasma membrane. Vasoconstriction is counteracted by activities of SKCa/IKCa channels in ECs and BKCa channels in VSMCs. The activation of SKCa and IKCa channels in ECs triggered by inositol triphosphate receptor (IP3R)-mediated Ca2+ release and/or transient receptor potential (TRP) channel-mediated Ca2+ influx promotes hyperpolarization and release of nitric oxide (NO)/endothelium-derived hyperpolarizing factor (EDRF). In addition, hyperpolarization in ECs could be transmitted to VSMCs via myoendothelial gag junctions (MEGJs). The BKCa channel is activated by RyR-mediated Ca2+ sparks in VSMCs. BKCa channel activity is also subject to regulation by EC-derived NO and EDHF. Moreover, the accumulation of K+ ions in the intercellular space hyperpolarizes VSMCs by activating the inwardly rectifying K+ (Kir) channel. Overall, these events promote hyperpolarization of VSMCs, which in turn leads to the closure of the CaV 1.2 channel and subsequent vasodilation. ACh, acetylcholine; BK, bradykinin; SP, substance P; GPCR, G protein-coupled receptor; PLC, phospholipase C; NOS, nitric oxide synthases; GC, guanylate cyclase; cGMP, cyclic guanosine monophosphate; PKG, protein kinase G.
Ijms 24 01349 g002
Figure 3. Epigenetic regulation of BKCa channel β1 subunit expression in uterine vasculature. The expression of the BKCa channel β1 subunit in uterine arteries of nonpregnant sheep is low due to hypermethylation of the KCNMB 1 promoter. Pregnancy increases the expression of the BKCa channel β1 subunit via promoting TET1-mediated demethylation of the promoter. Gestational hypoxia promotes methylation by upregulating DNMT3b and suppresses demethylation by downregulating TET1, leading to KCNMB 1 repression.
Figure 3. Epigenetic regulation of BKCa channel β1 subunit expression in uterine vasculature. The expression of the BKCa channel β1 subunit in uterine arteries of nonpregnant sheep is low due to hypermethylation of the KCNMB 1 promoter. Pregnancy increases the expression of the BKCa channel β1 subunit via promoting TET1-mediated demethylation of the promoter. Gestational hypoxia promotes methylation by upregulating DNMT3b and suppresses demethylation by downregulating TET1, leading to KCNMB 1 repression.
Ijms 24 01349 g003
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Hu, X.-Q.; Zhang, L. Ca2+-Activated K+ Channels and the Regulation of the Uteroplacental Circulation. Int. J. Mol. Sci. 2023, 24, 1349. https://doi.org/10.3390/ijms24021349

AMA Style

Hu X-Q, Zhang L. Ca2+-Activated K+ Channels and the Regulation of the Uteroplacental Circulation. International Journal of Molecular Sciences. 2023; 24(2):1349. https://doi.org/10.3390/ijms24021349

Chicago/Turabian Style

Hu, Xiang-Qun, and Lubo Zhang. 2023. "Ca2+-Activated K+ Channels and the Regulation of the Uteroplacental Circulation" International Journal of Molecular Sciences 24, no. 2: 1349. https://doi.org/10.3390/ijms24021349

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

Article Metrics

Back to TopTop