Next Article in Journal
Mechanical Disturbance of Osteoclasts Induces ATP Release That Leads to Protein Synthesis in Skeletal Muscle through an Akt-mTOR Signaling Pathway
Next Article in Special Issue
Serum Collected from Preeclamptic Pregnancies Drives Vasoconstriction of Human Omental Arteries—A Novel Ex Vivo Model of Preeclampsia for Therapeutic Development
Previous Article in Journal
An Ultra-Low Dose of ∆9-Tetrahydrocannabinol Alleviates Alzheimer’s Disease-Related Cognitive Impairments and Modulates TrkB Receptor Expression in a 5XFAD Mouse Model
Previous Article in Special Issue
Advanced Maternal Age Impairs Uterine Artery Adaptations to Pregnancy in Rats
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Potassium Channels in the Uterine Vasculature: Role in Healthy and Complicated Pregnancies

Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(16), 9446; https://doi.org/10.3390/ijms23169446
Submission received: 25 July 2022 / Revised: 16 August 2022 / Accepted: 19 August 2022 / Published: 21 August 2022
(This article belongs to the Special Issue Molecular Research of Vascular Aspects in Pregnancy)

Abstract

:
A progressive increase in maternal uterine and placental blood flow must occur during pregnancy to sustain the development of the fetus. Changes in maternal vasculature enable an increased uterine blood flow, placental nutrient and oxygen exchange, and subsequent fetal development. K+ channels are important modulators of vascular function, promoting vasodilation, inducing cell proliferation, and regulating cell signaling. Different types of K+ channels, such as Ca2+-activated, ATP-sensitive, and voltage-gated, have been implicated in the adaptation of maternal vasculature during pregnancy. Conversely, K+ channel dysfunction has been associated with vascular-related complications of pregnancy, including intrauterine growth restriction and pre-eclampsia. In this article, we provide an updated and comprehensive literature review that highlights the relevance of K+ channels as regulators of uterine vascular reactivity and their potential as therapeutic targets.

1. Introduction

1.1. Maternal Vascular Changes during Healthy Pregnancies and Pregnancy Complications

Pregnancy initiates major changes in the maternal uterine circulation that allow for the transfer of nutrients and oxygen to the developing fetus. During pregnancy, the uterine vasculature undergoes a process that includes marked vasodilation, increases in the lumen and length of the main uterine artery (UtA), as well as the downstream arcuate and myometrial arteries, and the remodeling of the spiral arteries along with the distal portion of the myometrial arteries [1]. The increased vasodilation and vascular remodeling serve to decrease the vascular tone and raise UtA blood flow, resulting in directing the major portion of the pregnancy rise in cardiac output to the uteroplacental circulation, which equates to approximately 1 L/min or 20% of cardiac output near term [2].
Many factors contribute to the increased vasodilation of uterine vasculature during pregnancy, including those affecting endothelial and vascular smooth muscle cell function. For instance, endothelial nitric oxide (NO) is a key mediator for the vasodilation prompted by shear stress, estrogen, or increased vascular endothelial growth factor production, and also for modulating circumferential remodeling [3,4,5,6,7]. Estrogen is a sex steroid hormone that is known to increase both locally and systemically during pregnancy and to cause the vasodilation of uterine vasculature and thus increase UtA blood flow [8] via acting to increase endothelial NO synthase expression and activity [9,10]. Estrogen also has potent effects in the UtA smooth muscle, where it can activate K+ channels to induce vasodilation [11]. Vascular remodeling during pregnancy results from tissue and cell hypertrophy, endothelial and vascular smooth muscle hyperplasia, and matrix remodeling (reviewed by [1]).
Abnormalities in these pregnancy-associated vascular dilation and remodeling processes are associated with the pregnancy complications of intrauterine growth restriction (IUGR) and pre-eclampsia, in which, the normal vasodilatory effects of acetylcholine (ACh), bradykinin, NO, endothelium-derived hyperpolarizing factor (EDHF), and thromboxane-mediated responses are impaired [12,13,14,15]. In addition, chronic hypoxia is one of the most common insults to fetal development, thought to be an underlying contributor to and raising the risk of IUGR and pre-eclampsia [16,17]. Chronic hypoxia has been shown to impair the reduction in vascular tone, uterine vasodilation, and growth occurring during normal pregnancy, which results in a lower rise in UtA blood flow compared to pregnancies occurring in a normoxic environment [17,18,19,20]. Furthermore, although not within the scope of this review, the placenta also plays important roles in the pathogenesis of vascular complications of pregnancy, as has been reviewed previously [21].

1.2. K+ Channels as Regulators of Vascular Function

K+ channels are membrane proteins that allow K+ ions to move across cell membranes following an electrochemical gradient, thus hyperpolarizing these membranes and, in turn, regulating a wide variety of cellular processes, including the control of cell excitability, contraction, and proliferation [22,23]. Particularly important are the K+ channels’ contribution to vasodilatory responses in resistance arteries. Specifically, the membrane hyperpolarization induced by K+ channel activation in smooth muscle cells leads to closing voltage-gated Ca2+ channels and a reduction in Ca2+ entry, thereby preventing contraction [24]. In endothelial cells, K+ channels are involved in EDHF responses, independent of NO and prostaglandin (PG) production [25]. It has been proposed that the hyperpolarization of endothelial cells further increases the entry of Ca2+ by increasing its electrochemical gradient [26]. Additionally, K+-channel-dependent endothelial cell hyperpolarization can also be propagated to the smooth muscle via gap junctions [27]. Thus, K+ channels tightly regulate the balance between pro-contractile and pro-dilatory responses, which is key for the proper function of vascular vessels and their response to different physiological conditions. In the uterine vasculature, the vasodilatory roles of K+ channels seem to be critical for the rise in UtA blood flow during pregnancy. Multiple K+ channel types have been described to participate in this process, including Ca2+-activated, ATP-sensitive, and voltage-dependent K+ channels.
In the present article, we discussed the current literature available supporting the important role of these K+ channels in the uterine vascular adaptation to pregnancy under healthy conditions and how the impaired function of these channels is related to different pregnancy complications. The information provided in this article may be relevant for researchers aiming at elucidating the complex factors underlying the pregnancy-dependent changes in uterine vasculature and for the identification of candidates to develop therapeutic approaches to prevent or alleviate pregnancy complications associated with impaired uteroplacental perfusion. A summary of the pregnancy-dependent regulation of K+ channels in the uterine vasculature described below is shown in Figure 1.

2. Ca2+-Activated K+ Channels

Ca2+-activated K+ channels are ubiquitously distributed throughout the body, where they integrate intracellular Ca2+ signals with the regulation of the membrane potential. There are three main subfamilies of these channels: large conductance (BKCa, Slo1, KCa1.1), intermediate conductance (IK, KCa3.1), and small conductance (SK, KCa2.x) Ca2+-activated K+ channels [28].
The pore-forming BKCa channel α subunit is encoded by a single gene (KCNMA1). Each α subunit consists of seven transmembrane domains with an extracellular N-terminus and a large cytosolic C-terminus [29,30]. Tetramers of α subunits form functional channels, which can be associated in a 4:4 stoichiometry with two types of auxiliary proteins (β or γ), for which, four β (β1–β4) and four γ (γ1–γ4) have been described [31]. In the vasculature, BKCa channels are predominantly expressed in smooth muscle cells, where β1 and γ1 auxiliary subunits have been shown to increase BKCa activity [32,33,34].
The IK channel is encoded by the KCNN4 gene (KCa3.1, IK1, SK4), whereas the SK channels are encoded by KCNN1 (KCa2.1, SK1), KCNN2 (KCa2.2, SK2), and KCNN3 (KCa2.3, SK3) genes. The pore-forming α subunits of IK and SK channels have six transmembrane domains with both N- and C-terminus cytoplasmic. The IK and SK channels have often been described as occurring in endothelial cells [35].

2.1. BKCa Channels in Healthy Pregnancies

In the uterine vasculature, as in other vascular beds, BKCa channels play important roles in promoting vasodilation and reducing myogenic tone. Specifically, pregnancy progressively raised BKCa channel activity in UtA smooth muscle cells isolated from pregnant sheep and mice compared to non-pregnant sheep and mice [36,37]. Moreover, in UtA from sheep, myogenic tone was increased by blocking BKCa channels with tetraethylammonium (TEA) in vessels from pregnant animals, but not from non-pregnant animals [36]. Similarly, the vasoconstrictor responses to the BKCa channel blocker iberiotoxin (IbTX) were augmented throughout pregnancy in mouse UtA [37]. Highlighting the important contribution to UtA blood flow, the in vivo infusion of the BKCa channel blocker TEA decreased UtA blood flow in pregnant sheep [38]. Knockout of the BKCa α subunit reduced the increase in UtA diameter during pregnancy and the number of pups per litter [37]. However, UtA resistance in vivo was not impacted [37], suggesting that other factors may compensate for the absence of BKCa channels in vivo.
The pregnancy-related changes in BKCa channel function are not likely due to an altered protein expression of the pore-forming α subunit. An increased expression of BKCa channel β1 and γ1 subunits has been described, although the α subunit expression did not change during pregnancy [36,37,39]. Furthermore, the protein expression of β1, γ1, and γ3 subunits has been described in whole UtA and cultured UtA smooth muscle cells isolated from pregnant women [40]. Studies have shown that the pregnancy-dependent increase in β1 subunit expression was dependent on estrogen activity [41] and epigenetic regulation of the KCNMB1 gene promoter site, Sp1, via a decrease in CpG methylation [42]. This pregnancy-induced epigenetic regulation of β1 subunit expression was associated with an increased expression of the demethylase ten-eleven translocation methylcytosine dioxygenase 1 [43]. In addition, mice lacking the γ1 subunit showed a reduced rise in UtA diameter at late pregnancy stages and diminished vasoconstrictor responses evoked by the BKCa blocker IbTX [37]. Overall, these studies suggest that alterations in the BKCa auxiliary subunits contribute to the characteristic rise in BKCa activity, increase in UtA diameter, and decrease in UtA myogenic tone occurring during pregnancy.
Besides their interaction with BKCa auxiliary subunits, BKCa channels are also regulated by intracellular signaling [44]. Protein kinase C (PKC), protein kinase G (PKG), and other cell signaling pathways have been shown to affect the activation of BKCa channels in the UtA in a pregnancy-dependent manner. PKC reduces BKCa channel activity in vascular smooth muscle cells [45], and PKC activity is decreased during pregnancy in ovine and porcine UtA [46,47]. Furthermore, the inhibition of BKCa channels enhanced the PKC-dependent vasoconstriction in pregnant sheep UtA [48], suggesting that the pregnancy-reduced PKC activity increases BKCa in UtA smooth muscle cells and leads to an enhanced vasodilation of the UtA throughout pregnancy. Conversely, other studies have described that pregnancy increased the levels of cGMP in UtA and enhanced PKG and therefore BKCa activation [39]. An increase in Ca2+ sparks, released from the sarcoplasmic reticulum via ryanodine receptor types 1 and 2, directly regulated BKCa channel activation in UtA smooth muscle cells, leading to spontaneous transient outward currents [49,50]. These authors also observed that a lack of ryanodine receptors 1 and 2 decreased BKCa β1 subunit expression and the interaction between BKCa α and β1, thus indirectly regulating BKCa channel activity [50].
Extracellular factors have also been described to modulate BKCa channels during pregnancy [44]. The calcitonin gene-related peptide (CGRP), a neuropeptide widely distributed throughout the central and peripheral nervous systems, is a potent vasodilator [51]. The CGRP concentration increases throughout pregnancy, is positively correlated with birth weight, and is reduced in pre-eclamptic women [52,53]. UtA vasodilatory responses to CGRP via activation of the CGRP receptor A [54] were reduced by blocking BKCa channels with TEA [55,56]. Another vasoactive peptide, adrenomedullin (ADM), whose levels are increased during pregnancy [57,58], was shown to vasodilate pregnant rat UtA in vitro via BKCa channel activation [59]. In pregnant mice, supplementation with melatonin, a peptide released by the pineal gland, and, during pregnancy, by the placenta [60], restored ACh-dependent UtA vasodilation that was reduced by blocking BKCa channels with IbTX [61]. Recently, another known vasodilator, hydrogen sulfide (H2S), has been described to vasodilate human UtA via the activation of BKCa channels [40]. H2S production is enhanced by pregnancy in sheep and human UtA [62,63]. Using an H2S donor, Li et al. observed an increase in K+ currents in UtA smooth muscle cells and relaxation of UtA rings from pregnant women [40]. These effects of H2S were reduced by IbTX preincubation and were independent of Ca2+ signaling [40]. H2S also augmented BKCa currents when applied to outside-out patches, suggesting that H2S could directly activate BKCa channel [40], although the probable mechanism remains unknown.

2.2. BKCa Channel Impaired Function in Pregnancy Complications

Chronic hypoxia, one of the most potent factors acting to impair vascular function and pregnancy outcome, affects BKCa channel function. Exposure to chronic hypoxia augmented UtA vascular tone in pregnant animals and increased PKC activity [48,64], with a concomitant reduction in BKCa activity [48]. Reactive oxygen species have also been described to reduce the BKCa-induced vasodilation of pregnant UtA from hypoxic-exposed sheep through a mechanism that involves the inhibition of steroid hormones function and downregulation of BKCa β1 subunits [65,66]. As described above, the epigenetic pregnancy-dependent demethylation of the β1 gene promoter leads to an increased expression of this auxiliary subunit [42]; it has been proposed that chronic hypoxia also affects this process by enhancing DNA methyltransferase activity, increasing the methylation of the β1 subunit promoter and decreasing its expression, resulting in reduced BKCa-dependent currents and vasodilation in the UtA [67]. Moreover, the increased DNA methylation of the estrogen receptor α gene promoter by hypoxia downregulates its expression, leading to an increased UtA myogenic tone via reduced BKCa activity [68].
In addition, hypoxia increased the expression of microRNA 210 (miR-210), which represses ten-eleven translocation methylcytosine dioxygenase 1, leading to the hypermethylation of KCNMB1 and reduced BKCa channel activity in the UtA [69,70]. Notably, miR-210 has been described as a master hypoxamiR (hypoxia-induced microRNA) and its expression is directly increased by hypoxia-induced factors [71]. Furthermore, circulating and placental miR-210 levels were higher in pre-eclamptic women compared to healthy women [72,73,74], and miR-210 has been shown to regulate the expression of targets that contribute to impaired trophoblast function and reduced angiogenesis [75,76], both key features of pre-eclampsia. In placental tissue from pre-eclamptic women, the increased expression of miR-210 has also been associated with a reduction in the K+ channel modulatory factor 1, KCMF1 [77], which is related to cell proliferation, further suggesting miR-210 as a link between hypoxia and pregnancy complications. MicroRNAs can be released to the circulation in exosomes or bound to proteins, and it has been postulated that the increase in miR-210 in the plasma of pre-eclamptic women could be the result of an increased exosomal secretion [78]; thus, it is possible that miR-210 secreted through placental-derived exosomes to the maternal circulation reduces BKCa-dependent vasodilation in the uterine vasculature. Further studies may elucidate the role of microRNA-containing exosomes in the regulation of ion channel function associated with pregnancy complications.
Altogether, the increase in BKCa activity appears to be highly regulated in the UtA during pregnancy and altered under hypoxic conditions. However, whether BKCa activity is similarly impaired under conditions of human pregnancy complications, such as pre-eclampsia and IUGR, remains to be determined.
Suggesting an important role for BKCa channel activity in such pregnancy complications are observations in stress-induced hypertensive mice during pregnancy. These hypertensive mice showed reduced levels of the BKCa β1 subunit in the UtA, and this reduction was partially reversed by supplementation with melatonin [61]. Functionally, the pharmacological blockage of BKCa channels with IbTX further reduced the already diminished ACh-dependent vasodilation in vitro in UtA from hypertensive pregnant mice compared to normotensive pregnant mice, and this effect of BKCa blockade was also prevented by melatonin supplementation [61]. These results highlight the role of impaired BKCa channel function in the pathophysiology of gestational hypertension.

2.3. SK and IK Channels in Healthy Pregnancies

SK and IK channels have been studied as important contributors to EDHF vascular responses and uteroplacental function during pregnancy. The pharmacological blockade of SK (apamin) and IK (charybdotoxin or TRAM-34) channels in human myometrial arteries reduced the EDHF-mediated vasodilation evoked by bradykinin [79]. A recent study showed that concurrently blocking SK and IK (with apamin and charybdotoxin, respectively) reduced the vasodilation induced by CGRP, ADM, and adrenomedullin 2 in pregnant human UtA [80]. Moreover, in UtA from both non-pregnant and late pregnant rats, blocking SK and IK channels abolished ACh-dependent vasodilation and prevented the decrease in intracellular Ca2+ levels in smooth muscle cells [81]. The SK-dependent component of ACh-induced UtA vasodilation was decreased in pregnant mice lacking relaxin, a hormone produced by the placenta with an important vasodilatory role in renal arteries, whereas the IK-channel-dependent component of ACh-evoked UtA vasodilation was augmented [82]. Furthermore, both SK and IK channels in the endothelium contributed to the ACh-dependent relaxation of UtA in non-pregnant buffaloes [83]. The IK channel blocker TRAM-34 prevented the dilation of non-pregnant and pregnant rat radial UtA (downstream of main UtA) induced by the activation of the transient receptor potential vanilloid type 3 ion channel [84]. In addition, activating IK channels with the channel opener SKA-31 elicited larger vasodilation in intact vessels from pregnant rats compared to non-pregnant rats or pregnant rats but with their endothelium removed [84]. Altogether, these findings suggest that SK and IK channels are important mechanisms for enhancing EDHF-mediated vasodilation in UtA. In ovine UtA, pregnancy also increased the SK-dependent vasodilation; however, this effect seems to be mediated by SK2 and SK3 channels predominantly expressed in the smooth muscle, since the removal of the endothelium did not change the vasodilation induced by an SK channel opener [85]. Using a different approach, another study showed that treating human uterine microvascular endothelial cells with serum from pregnant women compared to non-pregnant women increased the plasma membrane localization of SK3 and IK channels via a decreased caveolin-dependent internalization of SK3 and IK proteins [86].

2.4. SK and IK Channels in Pregnancy Complications

As described above, serum from pregnant women increases endothelial cell SK3 and IK channel plasma membrane localization compared to cells treated with serum from non-pregnant women. However, the localization of these channels in the plasma membrane was reduced in a clathrin- and caveolin-dependent manner when the endothelial cells were treated with serum from pre-eclamptic women [86]. The findings of Choi and colleagues indicate a role for internalization processes in the regulation of normal endothelial cell excitability and, in turn, vascular function, whereas an increased internalization of these channels may lead to the characteristic endothelial dysfunction observed in pre-eclampsia [87].
Similar to BKCa channels, chronic hypoxia also reduced the activity of SK channels. In a sheep model of pregnancy under hypoxic conditions, the expression of both SK2 and SK3 channel subtypes was reduced in pregnant UtA from animals exposed to chronic hypoxia [85]. Moreover, SK-dependent currents, vasodilatory responses, and a reduced myogenic tone were also abolished in UtA from pregnant sheep in hypoxic conditions compared to pregnant sheep in normoxic conditions [85], suggesting an impaired function of SK channels in the uterine vascular responses under hypoxic conditions during pregnancy.
Using a rat model of diabetes mellitus during pregnancy, Gokina et al. found that endothelial IK-dependent, but not SK-dependent, vasodilatory responses and ionic currents were reduced in radial arteries from diabetic pregnant rats compared to controls [88]. These observations suggest that IK channels are important contributors to the impaired uterine endothelial function, which has been observed in human pregnancies complicated by diabetes [89].
Increased testosterone levels during pregnancy can also be detrimental to fetal development. High levels of testosterone and a reduced UtA blood flow were observed in human pregnancies affected by polycystic ovary syndrome [90]. Rats treated with testosterone during pregnancy showed a decreased pup weight, increased UtA responses to vasoconstrictors, and reduced UtA vasodilatory responses compared to control animals. The lower vasodilatory response was partially attributable to a reduced EDHF component and decreased endothelial SK3 channel expression [91], suggesting a role for these channels in the UtA dysfunction induced by elevated testosterone levels.
Notably, mice overexpressing SK3 channels showed a reduction in the G-protein-mediated UtA contractile response and an increased fetal loss compared to wild-type animals [92]. Thus, special attention should also be given to the observations that exaggerated SK channel expression and activity could result in adverse pregnancy outcomes.

3. ATP-Sensitive K+ Channels

ATP-sensitive K+ (KATP) channels are expressed in many tissues, especially vascular smooth muscle, and are composed of the inwardly rectifying K+ channel family (Kir6.1 and Kir6.2), which forms the ion-conductive pore, and the regulatory ATP-binding sulfonylurea receptors (SUR1, SUR2A, and SUR2B). At least two of each of these subunits form a functional channel. In myometrial and vascular smooth muscle, Kir6.1/SUR2B and Kir6.2/SUR2B are the combinations that result in functional KATP channels [93,94] whose activity is enhanced by intracellular ADP acting on SUR2B and inhibited by intracellular ATP acting on Kir6.1 and Kir6.2 [95,96]. KATP channel activation in vascular beds increases vasodilation and blood flow [22], positioning these channels as important integrators of cellular metabolism with blood flow. This concept has been studied using the metabolic inhibitor, Na-azide, which decreases cellular ATP concentrations and increases KATP channel basal currents [97]. Adenosine, prostacyclin, and β-adrenergic agonists are among several vasodilators that act to enhance KATP channel activity through cyclic-AMP (cAMP) and protein kinase A (PKA) signaling pathways, ultimately resulting in membrane hyperpolarization and vasorelaxation [24,98]. Angiotensin II (Ang II), serotonin, endothelin-1, norepinephrine, and α-adrenergic agonists are vasoconstrictors that decrease KATP channel activity through the activation of PKC, resulting in membrane depolarization and contraction [24,99,100,101,102]. KATP channel activity and function have been studied using several different channel openers and blockers, such as the KATP channel openers pinacidil and diazoxide, as well as glibenclamide, which is a potent KATP channel blocker [103].

3.1. KATP Channels in Healthy Pregnancies

The KATP channel has been shown to play a major role in the modulation of membrane potential by the hyperpolarization and reduction in L-type Ca2+ channel activity leading to a decrease in intracellular Ca2+ concentrations, causing an overall reduction in vascular tone. In sheep, the KATP channel opener, diazoxide, induced UtA vasorelaxation by decreasing intracellular Ca2+ concentrations and attenuating phenylephrine-induced vascular contractions [104]. These effects were observed in both pregnant and nonpregnant sheep but were more potent in pregnant UtA than in nonpregnant UtA [104]. Similarly, the KATP channel opener pinacidil has been shown to oppose norepinephrine-induced vasoconstrictions in non-pregnant human UtA [105]. Moreover, blocking KATP channels with glibenclamide in vivo evoked a larger increase in blood pressure and systemic and uterine vascular resistance in pregnant animals compared to non-pregnant animals [106]. Ang II infusion also induced an increase in systemic and uterine vascular resistance that was further augmented by treatment with glibenclamide in non-pregnant and largely in pregnant guinea pigs [106], suggesting that KATP channels are important regulators of systemic and uterine vascular responses during pregnancy. In a state of metabolic demand, such as is seen during pregnancy, ATP levels decrease because of rapid consumption, which facilitates the opening of the KATP channel due to a lack of inhibition at the Kir subunit family [96]. Hence, the decreased ATP levels may be another contributor to the enhanced KATP channel activity during pregnancy.
KATP channels can also be regulated by other factors. For instance, CGRP activates KATP channels in endothelial and smooth muscle cells [107]. CGRP increases during pregnancy, the magnitude of which is directly correlated to fetal weight [53]. In the uterine vasculature of pregnant rats, CGRP activated KATP channels to induce vasodilation [55,56]. CGRP has also been shown to relax norepinephrine-constricted human UtA, and this vasodilation was inhibited by glibenclamide [105]. Taken together, these results indicate that the KATP channel is a key effector in the CGRP-mediated relaxation of UtA and that this relaxation is increased by pregnancy.

3.2. KATP Channel Impaired Function in Pregnancy Complications

KATP channel dysfunction contributes to the pathological conditions of hypertension, diabetes, obesity, hypercholesterolemia, atherosclerosis, and ischemia [22,108]. The KATP channel has been shown to play a role in the hypoxia-mediated reduction in UtA vasodilation. In pregnant sheep under normoxic conditions, the pharmacological opening of KATP channels decreases intracellular Ca2+ levels and increases UtA vasorelaxation, whereas pregnant animals maintained under long-term hypoxic conditions showed decreased KATP-dependent vasodilation [104], highlighting a role of these channels in the hypoxia-reduced UtA vasodilation. In vascular smooth muscle, KATP channels were inhibited by several vasoconstricting hormones and neurotransmitters through the activation of PKC. In the UtA, PKC activity is reduced in normal pregnancy but enhanced in the state of long-term hypoxia [64]; thus, the pregnancy-mediated increase and chronic hypoxia-mediated decrease in KATP channel activity may be regulated via alterations in PKC signaling.
KATP channels have also been described in fetal vessels. Yin and collaborators found that the mRNA and protein expression of the KATP channel subunit SUR2B were lower in the umbilical artery from cases of severe pre-eclampsia than in healthy pregnancies [109]. These observations imply an association between reduced KATP activity, through a diminished SUR2B expression, and the development of severe pre-eclampsia [109]. SUR2B mRNA and protein expression levels were also lower in early-onset severe pre-eclampsia than in late-onset severe pre-eclampsia, suggesting that the degree of the reduction in SUR2B expression may be related to the occurrence of severe pre-eclampsia [109], perhaps via KATP channel inhibition and reduced vasodilation.
Pinacidil-evoked KATP channel currents and vasodilation are reduced in the umbilical artery smooth muscle of pregnant women diagnosed with gestational diabetes mellitus (GDM) compared to a healthy pregnancy cohort [110]. mRNA and protein levels of KATP subunits Kir6.1, Kir6.2, and SUR2B were also decreased in GDM compared to the healthy control group [110]. However, the responses to adenosine or PKA activation, which activates KATP channels, did not differ, indicating that impaired KATP channel activity and vasorelaxation were due to downregulated KATP channel expression rather than changes in associated signaling [110]. A reduction in Kir6.1 subunit expression has also been observed in umbilical veins of women affected by pregnancy-induced hypertension and GDM [111]. In functional studies, mice with Kir6.1 deletions exhibited hypertension [112], whereas the gain-of-function in this subunit has been linked to hypotension [113], further suggesting its role in the regulation of vascular tone and thus blood pressure control. Furthermore, blocking KATP channels with glibenclamide increased systemic blood pressure in normotensive pregnant rats but did not further increase it in spontaneously hypertensive rats [114]. Thus, the study by Lima and colleagues suggests an impaired regulation of blood pressure by the KATP channel in pregnancies affected by hypertension [114].
IUGR and pre-eclampsia have been associated with decreased CGRP levels in maternal and fetal circulation and reduced placental CGRP expression [53]. Because CGRP is a known vasodilator and modulator of KATP activity in UtA from healthy pregnancies [55,56], a decrease in CGRP could impair the normal uteroplacental blood flow and could be associated with the etiology of IUGR and pre-eclampsia. However, future studies are needed to determine whether reduced CGRP levels lower KATP channel responses in the uterine vasculature of pregnancy complications.

4. Voltage-Gated K+ Channels

Voltage-gated K+ (Kv) channels are important regulators of membrane potential. They allow K+ efflux in response to the depolarization of the membrane. The mammalian Kv channel family is vast, with 12 distinct types (Kv1–12) forming homomers or heteromers [115]. The α subunits form functional channels, which can also be associated with auxiliary proteins [116].
In smooth muscle cells, Kv channels are activated by depolarization and provide a negative feedback mechanism that limits contraction [24,117]. Therefore, the constitutive activation of these channels plays an important role in maintaining myogenic tone in blood vessels. Vasoconstriction results from the inhibition of Kv channels in vascular smooth muscle cells via vasoconstrictors such as phenylephrine and Ang II [118,119,120]. In contrast, known vasodilators such as NO and H2S contribute to the activation of Kv channels in vascular smooth muscle [121,122,123,124]. These channels can also be activated under the conditions of acidosis or hypoxia, or by the influence of anticontractile substances derived from peripheral vascular adipose tissue [125,126,127].

4.1. Kv Channels in Healthy Pregnancies

The physiologic role of Kv channels in uterine vasculature has not been well characterized. Initial studies in non-pregnant sheep found that the in vivo infusion of 4-aminopyridine (4-AP), a selective Kv blocker, did not alter the basal uterine blood flow but did reduce the estradiol-induced rise in uterine blood flow [8]. K+ currents in cultured human UtA smooth muscle cells showed a predominant component attributable to Kv1 channels, whereas protein expression profiles showed the presence of Kv1.2, Kv1.3, Kv1.5, Kv3.4, Kv4.2, and Kv4.3 subtypes in human UtA tissue [128].
The effect of blocking Kv channels in the UtA has been studied in vitro. For instance, ACh-dependent vasodilation is only partially reduced by 4-AP in UtA from pregnant mice [61]. Conversely, exposure to 4-AP enhanced a pressure-evoked increase in intracellular Ca2+ concentrations and myogenic tone in radial UtA, and this effect was greater in non-pregnant rats than late-pregnant rats [129]. This, coupled with the significant decrease in currents through Kv channels in radial UtA smooth muscle cells from late-pregnant rats versus non-pregnant rats, indicates that a reduction in Kv channel activity in smooth muscle cells of small, resistance-size uteroplacental arteries contributes to vasoconstriction in late pregnancy [129]. Additionally, in non-pregnant guinea pigs, ACh-dependent UtA relaxation was not affected by incubation with 4-AP, nor by other K+ channel blockers such as TEA, glibenclamide, or apamin [130]. Consequently, a role for Kv channels in ACh-evoked vasodilation in UtA from pregnant mice, a greater effect of a Kv blocker in enhancing myogenic tone in radial arteries from non-pregnant rats than pregnant rats, and a lack of effect in blocking Kv channels in the ACh-dependent UtA vasodilation in non-pregnant guinea pigs highlight the potential diverse roles of Kv that may differ by species, vessel size, and/or pregnancy state.
The Kv channel subtype Kv3.4, expressed in the human UtA, has been associated with the proliferation of UtA vascular smooth muscle cells in culture [128]. Thus, the direct inhibition of this channel, as well as an increase in extracellular K+ to mimic Kv channel inhibition, resulted in a failure in the vascular smooth muscle cells to enter mitotic growth phases required for proliferation [131], indicating that smooth muscle cell hyperpolarization mediated by Kv channels is directly correlated with vascular smooth muscle proliferation.

4.2. Kv Channels in Pregnancy Complications

The role of Kv channels in the maternal vasculature of pregnancy complications has not been well described, but there are some studies on its role in the fetoplacental circulation. Hypoxia selectively reduced Kv currents measured by the patch clamp technique in smooth muscle cells from peripheral fetoplacental arteries and induced vasoconstriction in placental cotyledons [132]. Another study has shown an increase in the mRNA expression of Kv2.1 in chorionic plate veins from fetal growth-restricted pregnancies compared to healthy controls, whereas Kv9.3 levels were not altered [133].

5. Therapeutic Perspectives

Several characteristics of K+ channels make them relevant therapeutic targets for the prevention or treatment of pregnancy complications: (1) their important contribution to the vascular adaptation to pregnancy and their impaired function in pregnancy complications, (2) their role as end-point effectors for many vasodilators or vasoconstrictors, which is beneficial from a clinical point of view, as the modulation of their activity could lead to strong physiological responses, (3) the selective activation of these channels under different conditions (i.e., pregnancy, hypoxia, ischemia, etc.), and (4) their differential expression in distinct cell/tissue types. Currently, several therapeutic interventions using modulators of K+ channels during pregnancy are being assessed in clinical trials [134]. For instance, KATP channel blockers, such as glibenclamide, are being tested to alleviate the effects of GDM on fetal growth and adverse pregnancy outcomes. Furthermore, the KATP channel agonist nicorandil is currently being assessed for the prevention of preterm labor. As reviewed here and summarized in Table 1, multiple K+ channel openers and blockers have specific effects in the uterine vasculature. The role of K+ channels in vascular adaptation to pregnancy and their importance as modulators of a wide variety of signals may position K+ channels as good candidates for therapeutic intervention.

6. Conclusions

Pregnancy induces major changes in maternal uterine vasculature that enable a rise in UtA blood flow to the uteroplacental circulation required to meet the demand for nutrients and oxygen for normal fetal development. K+ channels have been widely described as effectors for multiple vasodilators, many of which are positively regulated during pregnancy. Conversely, in cases of pregnancy complications or under harmful conditions (such as hypoxic conditions), K+ channels in uterine vessels are downregulated and their activity is impaired. As reviewed here, the specific localization of different K+ channels, their selective gestational upregulation, and their modulatory role in the vascular adaptation to pregnancy make them potential therapeutic targets. However, since complications of pregnancy are likely the result of multiple factors (maternal, fetal, placental, genetic, environmental, etc.), additional research is needed for addressing the complex factors involved to prevent or treat pregnancy complications associated with dysfunctional uterine vascular adaptation to pregnancy. We expect that this comprehensive literature review will help researchers to identify the complex nature of the vascular adaptations to pregnancy and use this review as a tool for the development of new approaches.

Author Contributions

Writing—original draft preparation, W.B. and R.A.L.; writing—review and editing, W.B. and R.A.L.; funding acquisition, R.A.L. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by the National Institutes of Health (R03 HD101659 and R21 HD102628 to R.A.L.) and the American Heart Association (20IPA35320027 to R.A.L.) grants.

Institutional Review Board Statement

Not applicable.

Acknowledgments

We thank Lorna G. Moore and Heather Aldrich for critically reading and editing the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Osol, G.; Moore, L.G. Maternal uterine vascular remodeling during pregnancy. Microcirculation 2014, 21, 38–47. [Google Scholar] [CrossRef] [PubMed]
  2. 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]
  3. van der Heijden, O.W.; Essers, Y.P.; Fazzi, G.; Peeters, L.L.; De Mey, J.G.; van Eys, G.J. Uterine artery remodeling and reproductive performance are impaired in endothelial nitric oxide synthase-deficient mice. Biol. Reprod. 2005, 72, 1161–1168. [Google Scholar] [CrossRef] [PubMed]
  4. Hale, S.A.; Weger, L.; Mandala, M.; Osol, G. Reduced NO signaling during pregnancy attenuates outward uterine artery remodeling by altering MMP expression and collagen and elastin deposition. Am. J. Physiol. Heart Circ. Physiol. 2011, 301, H1266–H1275. [Google Scholar] [CrossRef] [Green Version]
  5. Osol, G.; Barron, C.; Gokina, N.; Mandala, M. Inhibition of nitric oxide synthases abrogates pregnancy-induced uterine vascular expansive remodeling. J. Vasc. Res. 2009, 46, 478–486. [Google Scholar] [CrossRef] [Green Version]
  6. Joyce, J.M.; Phernetton, T.M.; Magness, R.R. Effect of uterine blood flow occlusion on shear stress-mediated nitric oxide production and endothelial nitric oxide synthase expression during ovine pregnancy. Biol. Reprod. 2002, 67, 320–326. [Google Scholar] [CrossRef] [Green Version]
  7. Itoh, S.; Brawley, L.; Wheeler, T.; Anthony, F.W.; Poston, L.; Hanson, M.A. Vasodilation to vascular endothelial growth factor in the uterine artery of the pregnant rat is blunted by low dietary protein intake. Pediatr. Res. 2002, 51, 485–491. [Google Scholar] [CrossRef] [Green Version]
  8. Rosenfeld, C.R.; Roy, T.; Cox, B.E. Mechanisms modulating estrogen-induced uterine vasodilation. Vasc. Pharmacol. 2002, 38, 115–125. [Google Scholar] [CrossRef]
  9. Rosenfeld, C.R.; Cox, B.E.; Roy, T.; Magness, R.R. Nitric oxide contributes to estrogen-induced vasodilation of the ovine uterine circulation. J. Clin. Investig. 1996, 98, 2158–2166. [Google Scholar] [CrossRef]
  10. 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]
  11. Rosenfeld, C.R.; Roy, T. Large conductance Ca2+-activated and voltage-activated K+ channels contribute to the rise and maintenance of estrogen-induced uterine vasodilation and maintenance of blood pressure. Endocrinology 2012, 153, 6012–6020. [Google Scholar] [CrossRef] [PubMed]
  12. 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]
  13. 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]
  14. Ong, S.S.; Moore, R.J.; Warren, A.Y.; Crocker, I.P.; Fulford, J.; Tyler, D.J.; Gowland, P.A.; Baker, P.N. Myometrial and placental artery reactivity alone cannot explain reduced placental perfusion in pre-eclampsia and intrauterine growth restriction. BJOG 2003, 110, 909–915. [Google Scholar] [CrossRef]
  15. Lorca, R.A.; Matarazzo, C.J.; Bales, E.S.; Houck, J.A.; Orlicky, D.J.; Euser, A.G.; Julian, C.G.; Moore, L.G. AMPK activation in pregnant human myometrial arteries from high-altitude and intrauterine growth-restricted pregnancies. Am. J. Physiol. Heart Circ. Physiol. 2020, 319, H203–H212. [Google Scholar] [CrossRef] [PubMed]
  16. Palmer, S.K.; Moore, L.G.; Young, D.; Cregger, B.; Berman, J.C.; Zamudio, S. Altered blood pressure course during normal pregnancy and increased preeclampsia at high altit.tude (3100 meters) in Colorado. Am. J. Obstet. Gynecol. 1999, 180, 1161–1168. [Google Scholar] [CrossRef]
  17. 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]
  18. 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]
  19. Aljunaidy, M.M.; Morton, J.S.; Cooke, C.L.; Davidge, S.T. Maternal vascular responses to hypoxia in a rat model of intrauterine growth restriction. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2016, 311, R1068–R1075. [Google Scholar] [CrossRef]
  20. White, M.M.; Zhang, L. Effects of chronic hypoxia on maternal vasodilation and vascular reactivity in guinea pig and ovine pregnancy. High Alt. Med. Biol. 2003, 4, 157–169. [Google Scholar] [CrossRef]
  21. Ortega, M.A.; Fraile-Martinez, O.; Garcia-Montero, C.; Saez, M.A.; Alvarez-Mon, M.A.; Torres-Carranza, D.; Alvarez-Mon, M.; Bujan, J.; Garcia-Honduvilla, N.; Bravo, C.; et al. The Pivotal Role of the Placenta in Normal and Pathological Pregnancies: A Focus on Preeclampsia, Fetal Growth Restriction, and Maternal Chronic Venous Disease. Cells 2022, 11, 568. [Google Scholar] [CrossRef] [PubMed]
  22. Jackson, W.F. Potassium Channels in Regulation of Vascular Smooth Muscle Contraction and Growth. Adv. Pharmacol. 2017, 78, 89–144. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Korovkina, V.P.; England, S.K. Molecular diversity of vascular potassium channel isoforms. Clin. Exp. Pharmacol. Physiol. 2002, 29, 317–323. [Google Scholar] [CrossRef] [PubMed]
  24. Nelson, M.T.; Quayle, J.M. Physiological roles and properties of potassium channels in arterial smooth muscle. Am. J. Physiol. 1995, 268 Pt 1, C799–C822. [Google Scholar] [CrossRef]
  25. Busse, R.; Edwards, G.; Feletou, M.; Fleming, I.; Vanhoutte, P.M.; Weston, A.H. EDHF: Bringing the concepts together. Trends Pharmacol. Sci. 2002, 23, 374–380. [Google Scholar] [CrossRef]
  26. Behringer, E.J.; Segal, S.S. Membrane potential governs calcium influx into microvascular endothelium: Integral role for muscarinic receptor activation. J. Physiol. 2015, 593, 4531–4548. [Google Scholar] [CrossRef] [Green Version]
  27. Walker, S.D.; Dora, K.A.; Ings, N.T.; Crane, G.J.; Garland, C.J. Activation of endothelial cell IK(Ca) with 1-ethyl-2-benzimidazolinone evokes smooth muscle hyperpolarization in rat isolated mesenteric artery. Br. J. Pharmacol. 2001, 134, 1548–1554. [Google Scholar] [CrossRef] [Green Version]
  28. Kaczmarek, L.K.; Aldrich, R.W.; Chandy, K.G.; Grissmer, S.; Wei, A.D.; Wulff, H. International Union of Basic and Clinical Pharmacology. C. Nomenclature and Properties of Calcium-Activated and Sodium-Activated Potassium Channels. Pharmacol. Rev. 2017, 69, 1–11. [Google Scholar] [CrossRef] [Green Version]
  29. Wei, A.; Solaro, C.; Lingle, C.; Salkoff, L. Calcium sensitivity of BK-type KCa channels determined by a separable domain. Neuron 1994, 13, 671–681. [Google Scholar] [CrossRef]
  30. Wallner, M.; Meera, P.; Toro, L. Determinant for β-subunit regulation in high-conductance voltage-activated and Ca2+-sensitive K+ channels: An additional transmembrane region at the N terminus. Proc. Natl. Acad. Sci. USA 1996, 93, 14922–14927. [Google Scholar] [CrossRef] [Green Version]
  31. 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] [PubMed]
  32. Evanson, K.W.; Bannister, J.P.; Leo, M.D.; Jaggar, J.H. LRRC26 is a functional BK channel auxiliary γ subunit in arterial smooth muscle cells. Circ. Res. 2014, 115, 423–431. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. 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 β1 subunit of the calcium-activated potassium channel. Nature 2000, 407, 870–876. [Google Scholar] [CrossRef]
  34. Tanaka, Y.; Meera, P.; Song, M.; Knaus, H.G.; Toro, L. Molecular constituents of maxi KCa channels in human coronary smooth muscle: Predominant α+β subunit complexes. J. Physiol. 1997, 502 Pt 3, 545–557. [Google Scholar] [CrossRef]
  35. 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] [PubMed] [Green Version]
  36. 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]
  37. 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 γ1-subunit modulate mouse uterine artery function during pregnancy. J. Physiol. 2018, 596, 1019–1033. [Google Scholar] [CrossRef] [Green Version]
  38. 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]
  39. 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. [Google Scholar] [CrossRef] [Green Version]
  40. 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]
  41. Nagar, D.; Liu, X.T.; Rosenfeld, C.R. Estrogen regulates β1-subunit expression in Ca2+-activated K+ channels in arteries from reproductive tissues. Am. J. Physiol. Heart Circ. Physiol. 2005, 289, H1417. [Google Scholar] [CrossRef] [PubMed]
  42. 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] [PubMed] [Green Version]
  43. 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] [PubMed]
  44. Lorca, R.A.; Prabagaran, M.; England, S.K. Functional insights into modulation of BKCa channel activity to alter myometrial contractility. Front. Physiol. 2014, 5, 289. [Google Scholar] [CrossRef] [Green Version]
  45. Barman, S.A.; Zhu, S.; White, R.E. Protein kinase C inhibits BKCa channel activity in pulmonary arterial smooth muscle. Am. J. Physiol. Lung Cell. Mol. Physiol. 2004, 286 Pt 1, L149–L155. [Google Scholar] [CrossRef] [Green Version]
  46. 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]
  47. Farley, D.B.; Ford, S.P. Evidence for declining extracellular calcium uptake and protein kinase C activity in uterine arterial smooth muscle during gestation in gilts. Biol. Reprod. 1992, 46, 315–321. [Google Scholar] [CrossRef]
  48. 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] [Green Version]
  49. 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]
  50. 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]
  51. Shen, Y.T.; Pittman, T.J.; Buie, P.S.; Bolduc, D.L.; Kane, S.A.; Koblan, K.S.; Gould, R.J.; Lynch, J.J., Jr. Functional role of α-calcitonin gene-related peptide in the regulation of the cardiovascular system. J. Pharmacol. Exp. Ther. 2001, 298, 551–558. [Google Scholar] [PubMed]
  52. Stevenson, J.C.; Macdonald, D.W.; Warren, R.C.; Booker, M.W.; Whitehead, M.I. Increased concentration of circulating calcitonin gene related peptide during normal human pregnancy. Br. Med. J. 1986, 293, 1329–1330. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Dong, Y.L.; Chauhan, M.; Green, K.E.; Vegiraju, S.; Wang, H.Q.; Hankins, G.D.; Yallampalli, C. Circulating calcitonin gene-related peptide and its placental origins in normotensive and preeclamptic pregnancies. Am. J. Obstet. Gynecol. 2006, 195, 1657–1667. [Google Scholar] [CrossRef] [PubMed]
  54. Chauhan, M.; Gangula, P.R.; Wimalawansa, S.J.; Yallampalli, C. Studies on the effects of the N-terminal domain antibodies of calcitonin receptor-like receptor and receptor activity-modifying protein 1 on calcitonin gene-related peptide-induced vasorelaxation in rat uterine artery. Biol. Reprod. 2004, 70, 1658–1663. [Google Scholar] [CrossRef] [Green Version]
  55. 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] [Green Version]
  56. Vedernikov, Y.P.; Fulep, E.E.; Saade, G.R.; Garfield, R.E. Calcitonin gene-related peptide dilates the pregnant rat uterine vascular bed via guanylate cyclase, ATP- and Ca-sensitive potassium channels and gap junctions. Curr. Med. Res. Opin. 2002, 18, 465–470. [Google Scholar] [CrossRef]
  57. Di Iorio, R.; Marinoni, E.; Letizia, C.; Alo, P.; Villaccio, B.; Cosmi, E.V. Adrenomedullin, a new vasoactive peptide, is increased in preeclampsia. Hypertension 1998, 32, 758–763. [Google Scholar] [CrossRef] [Green Version]
  58. Jerat, S.; Kaufman, S. Effect of pregnancy and steroid hormones on plasma adrenomedullin levels in the rat. Can. J. Physiol. Pharmacol. 1998, 76, 463–466. [Google Scholar] [CrossRef]
  59. 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]
  60. Soliman, A.; Lacasse, A.A.; Lanoix, D.; Sagrillo-Fagundes, L.; Boulard, V.; Vaillancourt, C. Placental melatonin system is present throughout pregnancy and regulates villous trophoblast differentiation. J. Pineal Res. 2015, 59, 38–46. [Google Scholar] [CrossRef] [Green Version]
  61. 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] [PubMed]
  62. 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]
  63. 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]
  64. 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] [Green Version]
  65. 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]
  66. 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]
  67. 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]
  68. Chen, M.; Xiao, D.; Hu, X.Q.; Dasgupta, C.; Yang, S.; Zhang, L. Hypoxia Represses ER-α 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]
  69. 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]
  70. 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] [Green Version]
  71. Bavelloni, A.; Ramazzotti, G.; Poli, A.; Piazzi, M.; Focaccia, E.; Blalock, W.; Faenza, I. MiRNA-210: A Current Overview. Anticancer Res. 2017, 37, 6511–6521. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. 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] [PubMed]
  73. Enquobahrie, D.A.; Abetew, D.F.; Sorensen, T.K.; Willoughby, D.; Chidambaram, K.; Williams, M.A. Placental microRNA expression in pregnancies complicated by preeclampsia. Am. J. Obstet. Gynecol. 2011, 204, 178.e12–178.e21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Jairajpuri, D.S.; Malalla, Z.H.; Sarray, S.; Mahmood, N. Analysis of differential expression of hypoxia-inducible microRNA-210 gene targets in mild and severe preeclamptic patients. Noncoding RNA Res. 2021, 6, 51–57. [Google Scholar] [CrossRef] [PubMed]
  75. Hayder, H.; Fu, G.; Nadeem, L.; O’Brien, J.A.; Lye, S.J.; Peng, C. Overexpression of miR-210-3p Impairs Extravillous Trophoblast Functions Associated with Uterine Spiral Artery Remodeling. Int. J. Mol. Sci. 2021, 22, 3961. [Google Scholar] [CrossRef]
  76. Korkes, H.A.; De Oliveira, L.; Sass, N.; Salahuddin, S.; Karumanchi, S.A.; Rajakumar, A. Relationship between hypoxia and downstream pathogenic pathways in preeclampsia. Hypertens. Pregnancy 2017, 36, 145–150. [Google Scholar] [CrossRef] [Green Version]
  77. Luo, R.; Shao, X.; Xu, P.; Liu, Y.; Wang, Y.; Zhao, Y.; Liu, M.; Ji, L.; Li, Y.X.; Chang, C.; et al. MicroRNA-210 contributes to preeclampsia by downregulating potassium channel modulatory factor 1. Hypertension 2014, 64, 839–845. [Google Scholar] [CrossRef] [Green Version]
  78. Biro, O.; Fothi, A.; Alasztics, B.; Nagy, B.; Orban, T.I.; Rigo, J., Jr. Circulating exosomal and Argonaute-bound microRNAs in preeclampsia. Gene 2019, 692, 138–144. [Google Scholar] [CrossRef] [Green Version]
  79. 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]
  80. Chauhan, M.; Betancourt, A.; Balakrishnan, M.; Mishra, A.; Espinosa, J.; Shamshirsaz, A.A.; Fox, K.; Belfort, M.; Yallampalli, C. Calcitonin Gene Related Peptide, Adrenomedullin, and Adrenomedullin 2 Function in Uterine Artery During Human Pregnancy. Endocrinology 2022, 163, bqab204. [Google Scholar] [CrossRef]
  81. 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] [Green Version]
  82. Marshall, S.A.; Senadheera, S.N.; Jelinic, M.; O’Sullivan, K.; Parry, L.J.; Tare, M. Relaxin Deficiency Leads to Uterine Artery Dysfunction During Pregnancy in Mice. Front. Physiol. 2018, 9, 255. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Nakade, U.P.; Sharma, A.; Kumari, P.; Bhatiya, S.; Nair, S.V.; Karikaran, K.N.; Sharma, V.; Choudhury, S.; Garg, S.K. Functional and molecular characterization of endothelium-dependent and endothelium-independent relaxant pathways in uterine artery of non-pregnant buffaloes. Naunyn Schmiedebergs Arch. Pharmacol. 2020, 393, 225–241. [Google Scholar] [CrossRef] [PubMed]
  84. Murphy, T.V.; Kanagarajah, A.; Toemoe, S.; Bertrand, P.P.; Grayson, T.H.; Britton, F.C.; Leader, L.; Senadheera, S.; Sandow, S.L. TRPV3 expression and vasodilator function in isolated uterine radial arteries from non-pregnant and pregnant rats. Vasc. Pharmacol. 2016, 83, 66–77. [Google Scholar] [CrossRef]
  85. 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]
  86. 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]
  87. Opichka, M.A.; Rappelt, M.W.; Gutterman, D.D.; Grobe, J.L.; McIntosh, J.J. Vascular Dysfunction in Preeclampsia. Cells 2021, 10, 3055. [Google Scholar] [CrossRef]
  88. 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] [Green Version]
  89. 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]
  90. Palomba, S.; Falbo, A.; Russo, T.; Battista, L.; Tolino, A.; Orio, F.; Zullo, F. Uterine blood flow in pregnant patients with polycystic ovary syndrome: Relationships with clinical outcomes. BJOG Int. J. Obstet. Gynaecol. 2010, 117, 711–721. [Google Scholar] [CrossRef]
  91. 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]
  92. 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]
  93. Babenko, A.P.; Aguilar-Bryan, L.; Bryan, J. A view of sur/KIR6.X, KATP channels. Annu. Rev. Physiol. 1998, 60, 667–687. [Google Scholar] [CrossRef] [PubMed]
  94. Du, Q.; Jovanovic, S.; Tulic, L.; Tulic, I.; Jovanovic, A. Pregnancy-induced hypertension is associated with down-regulation of Kir6.1 in human myometrium. Pregnancy Hypertens. 2019, 18, 96–98. [Google Scholar] [CrossRef]
  95. Yamada, M.; Isomoto, S.; Matsumoto, S.; Kondo, C.; Shindo, T.; Horio, Y.; Kurachi, Y. Sulphonylurea receptor 2B and Kir6.1 form a sulphonylurea-sensitive but ATP-insensitive K+ channel. J. Physiol. 1997, 499 Pt 3, 715–720. [Google Scholar] [CrossRef]
  96. Yoshida, H.; Bao, L.; Kefaloyianni, E.; Taskin, E.; Okorie, U.; Hong, M.; Dhar-Chowdhury, P.; Kaneko, M.; Coetzee, W.A. AMP-activated protein kinase connects cellular energy metabolism to KATP channel function. J. Mol. Cell. Cardiol. 2012, 52, 410–418. [Google Scholar] [CrossRef] [Green Version]
  97. Li, C.G.; Cui, W.Y.; Wang, H. Sensitivity of KATP channels to cellular metabolic disorders and the underlying structural basis. Acta Pharmacol. Sin. 2016, 37, 134–142. [Google Scholar] [CrossRef] [Green Version]
  98. Dart, C.; Standen, N.B. Adenosine-activated potassium current in smooth muscle cells isolated from the pig coronary artery. J. Physiol. 1993, 471, 767–786. [Google Scholar] [CrossRef]
  99. Cole, W.C.; Malcolm, T.; Walsh, M.P.; Light, P.E. Inhibition by protein kinase C of the K(NDP) subtype of vascular smooth muscle ATP-sensitive potassium channel. Circ. Res. 2000, 87, 112–117. [Google Scholar] [CrossRef] [Green Version]
  100. Miyoshi, Y.; Nakaya, Y. Angiotensin II blocks ATP-sensitive K+ channels in porcine coronary artery smooth muscle cells. Biochem. Biophys. Res. Commun. 1991, 181, 700–706. [Google Scholar] [CrossRef]
  101. Miyoshi, Y.; Nakaya, Y.; Wakatsuki, T.; Nakaya, S.; Fujino, K.; Saito, K.; Inoue, I. Endothelin blocks ATP-sensitive K+ channels and depolarizes smooth muscle cells of porcine coronary artery. Circ. Res. 1992, 70, 612–616. [Google Scholar] [CrossRef] [Green Version]
  102. Quayle, J.M.; Nelson, M.T.; Standen, N.B. ATP-sensitive and inwardly rectifying potassium channels in smooth muscle. Physiol. Rev. 1997, 77, 1165–1232. [Google Scholar] [CrossRef]
  103. Standen, N.B.; Quayle, J.M.; Davies, N.W.; Brayden, J.E.; Huang, Y.; Nelson, M.T. Hyperpolarizing vasodilators activate ATP-sensitive K+ channels in arterial smooth muscle. Science 1989, 245, 177–180. [Google Scholar] [CrossRef]
  104. Xiao, D.; Longo, L.D.; Zhang, L. Role of KATP and L-type Ca2+ channel activities in regulation of ovine uterine vascular contractility: Effect of pregnancy and chronic hypoxia. Am. J. Obstet. Gynecol. 2010, 203, 596.e6–596.e12. [Google Scholar] [CrossRef] [Green Version]
  105. Nelson, S.H.; Suresh, M.S.; Dehring, D.J.; Johnson, R.L. Relaxation by calcitonin gene-related peptide may involve activation of K+ channels in the human uterine artery. Eur. J. Pharmacol. 1993, 242, 255–261. [Google Scholar] [CrossRef]
  106. Keyes, L.; Rodman, D.M.; Curran-Everett, D.; Morris, K.; Moore, L.G. Effect of K+ATP channel inhibition on total and regional vascular resistance in guinea pig pregnancy. Am. J. Physiol. 1998, 275, H680–H688. [Google Scholar] [CrossRef]
  107. Norton, C.E.; Segal, S.S. Calcitonin gene-related peptide hyperpolarizes mouse pulmonary artery endothelial tubes through KATP channel activation. Am. J. Physiol. Lung Cell. Mol. Physiol. 2018, 315, L212–L226. [Google Scholar] [CrossRef] [Green Version]
  108. Dogan, M.F.; Yildiz, O.; Arslan, S.O.; Ulusoy, K.G. Potassium channels in vascular smooth muscle: A pathophysiological and pharmacological perspective. Fundam Clin. Pharmacol. 2019, 33, 504–523. [Google Scholar] [CrossRef]
  109. Yin, B.; Zhang, Y.; Wei, X.; Pang, C.; Hou, T.; Yang, C.; Ning, Y.; Fu, X. The expression of ATP-sensitive potassium channels in human umbilical arteries with severe pre-eclampsia. Sci. Rep. 2021, 11, 7955. [Google Scholar] [CrossRef]
  110. Li, H.; Shin, S.E.; Seo, M.S.; An, J.R.; Ha, K.S.; Han, E.T.; Hong, S.H.; Kim, J.; Yim, M.J.; Lee, J.M.; et al. Alterations of ATP-sensitive K+ channels in human umbilical arterial smooth muscle during gestational diabetes mellitus. Pflugers. Arch. 2018, 470, 1325–1333. [Google Scholar] [CrossRef]
  111. Djokic, V.; Jankovic-Raznatovic, S.; Novakovic, R.; Kostic, M.; Rajkovic, J.; Labudovic-Borovic, M.; Rakocevic, J.; Stanisic, J.; Djuric, M.; Gojkovic-Bukarica, L. Effect of gestational diabetes mellitus and pregnancy-induced hypertension on human umbilical vein smooth muscle KATP channels. Exp. Mol. Pathol. 2019, 111, 104323. [Google Scholar] [CrossRef]
  112. Aziz, Q.; Thomas, A.M.; Gomes, J.; Ang, R.; Sones, W.R.; Li, Y.; Ng, K.E.; Gee, L.; Tinker, A. The ATP-sensitive potassium channel subunit, Kir6.1, in vascular smooth muscle plays a major role in blood pressure control. Hypertension 2014, 64, 523–529. [Google Scholar] [CrossRef] [Green Version]
  113. Li, A.; Knutsen, R.H.; Zhang, H.; Osei-Owusu, P.; Moreno-Dominguez, A.; Harter, T.M.; Uchida, K.; Remedi, M.S.; Dietrich, H.H.; Bernal-Mizrachi, C.; et al. Hypotension due to Kir6.1 gain-of-function in vascular smooth muscle. J. Am. Heart Assoc. 2013, 2, e000365. [Google Scholar] [CrossRef] [Green Version]
  114. Lima, R.; Tardim, J.C.; Barros, M.E.; Boim, M.A. Role of ATP-sensitive potassium channels in normal and hypertension-associated pregnancy in rats. Clin. Exp. Pharmacol. Physiol. 2006, 33, 780–786. [Google Scholar] [CrossRef]
  115. Gutman, G.A.; Chandy, K.G.; Adelman, J.P.; Aiyar, J.; Bayliss, D.A.; Clapham, D.E.; Covarriubias, M.; Desir, G.V.; Furuichi, K.; Ganetzky, B.; et al. International Union of Pharmacology. XLI. Compendium of voltage-gated ion channels: Potassium channels. Pharmacol. Rev. 2003, 55, 583–586. [Google Scholar] [CrossRef]
  116. Torres, Y.P.; Morera, F.J.; Carvacho, I.; Latorre, R. A marriage of convenience: β-subunits and voltage-dependent K+ channels. J. Biol. Chem. 2007, 282, 24485–24489. [Google Scholar] [CrossRef] [Green Version]
  117. Jackson, W.F. KV channels and the regulation of vasc.cular smooth muscle tone. Microcirculation 2018, 25, e12421. [Google Scholar] [CrossRef]
  118. Mistry, D.K.; Garland, C.J. The influence of phenylephrine outward potassium currents in single smooth muscle cells from the rabbit mesenteric artery. Gen. Pharmacol. 1999, 33, 389–399. [Google Scholar] [CrossRef]
  119. Hayabuchi, Y.; Standen, N.B.; Davies, N.W. Angiotensin II inhibits and alters kinetics of voltage-gated K+ channels of rat arterial smooth muscle. Am. J. Physiol. Heart Circ. Physiol. 2001, 281, H2480–H2489. [Google Scholar] [CrossRef] [Green Version]
  120. Kidd, M.W.; Bulley, S.; Jaggar, J.H. Angiotensin II reduces the surface abundance of KV 1.5 channels in arterial myocytes to stimulate vasoconstriction. J. Physiol. 2017, 595, 1607–1618. [Google Scholar] [CrossRef]
  121. Sobey, C.G.; Faraci, F.M. Effect of nitric oxide and potassium channel agonists and inhibitors on basilar artery diameter. Am. J. Physiol. 1997, 272, H256–H262. [Google Scholar] [CrossRef]
  122. Tanaka, Y.; Tang, G.; Takizawa, K.; Otsuka, K.; Eghbali, M.; Song, M.; Nishimaru, K.; Shigenobu, K.; Koike, K.; Stefani, E.; et al. Kv channels contribute to nitric oxide- and atrial natriuretic peptide-induced relaxation of a rat conduit artery. J. Pharmacol. Exp. Ther. 2006, 317, 341–354. [Google Scholar] [CrossRef] [Green Version]
  123. Cheang, W.S.; Wong, W.T.; Shen, B.; Lau, C.W.; Tian, X.Y.; Tsang, S.Y.; Yao, X.; Chen, Z.Y.; Huang, Y. 4-aminopyridine-sensitive K+ channels contributes to NaHS-induced membrane hyperpolarization and relaxation in the rat coronary artery. Vasc. Pharmacol. 2010, 53, 94–98. [Google Scholar] [CrossRef]
  124. Martelli, A.; Testai, L.; Breschi, M.C.; Lawson, K.; McKay, N.G.; Miceli, F.; Taglialatela, M.; Calderone, V. Vasorelaxation by hydrogen sulphide involves activation of Kv7 potassium channels. Pharmacol. Res. 2013, 70, 27–34. [Google Scholar] [CrossRef]
  125. Hedegaard, E.R.; Nielsen, B.D.; Kun, A.; Hughes, A.D.; Kroigaard, C.; Mogensen, S.; Matchkov, V.V.; Frobert, O.; Simonsen, U. KV 7 channels are involved in hypoxia-induced vasodilatation of porcine coronary arteries. Br. J. Pharmacol. 2014, 171, 69–82. [Google Scholar] [CrossRef] [Green Version]
  126. Berger, M.G.; Vandier, C.; Bonnet, P.; Jackson, W.F.; Rusch, N.J. Intracellular acidosis differentially regulates KV channels in coronary and pulmonary vascular muscle. Am. J. Physiol. 1998, 275, H1351–H1359. [Google Scholar] [CrossRef] [Green Version]
  127. Tano, J.Y.; Schleifenbaum, J.; Gollasch, M. Perivascular adipose tissue, potassium channels, and vascular dysfunction. Arter. Thromb. Vasc. Biol. 2014, 34, 1827–1830. [Google Scholar] [CrossRef] [Green Version]
  128. Miguel-Velado, E.; Moreno-Dominguez, A.; Colinas, O.; Cidad, P.; Heras, M.; Perez-Garcia, M.T.; Lopez-Lopez, J.R. Contribution of Kv channels to phenotypic remodeling of human uterine artery smooth muscle cells. Circ. Res. 2005, 97, 1280–1287. [Google Scholar] [CrossRef] [Green Version]
  129. Telezhkin, V.; Goecks, T.; Bonev, A.D.; Osol, G.; Gokina, N.I. Decreased function of voltage-gated potassium channels contributes to augmented myogenic tone of uterine arteries in late pregnancy. Am. J. Physiol. Heart Circ. Physiol. 2008, 294, H272–H284. [Google Scholar] [CrossRef]
  130. Jovanovic, A.; Grbovic, L.; Jovanovic, S. K+ channel blockers do not modify relaxation of guinea-pig uterine artery evoked by acetylcholine. Eur. J. Pharmacol. 1995, 280, 95–100. [Google Scholar] [CrossRef]
  131. Miguel-Velado, E.; Perez-Carretero, F.D.; Colinas, O.; Cidad, P.; Heras, M.; Lopez-Lopez, J.R.; Perez-Garcia, M.T. Cell cycle-dependent expression of Kv3.4 channels modulates proliferation of human uterine artery smooth muscle cells. Cardiovasc. Res. 2010, 86, 383–391. [Google Scholar] [CrossRef] [Green Version]
  132. Hampl, V.; Bibova, J.; Stranak, Z.; Wu, X.; Michelakis, E.D.; Hashimoto, K.; Archer, S.L. Hypoxic fetoplacental vasoconstriction in humans is mediated by potassium channel inhibition. Am. J. Physiol. Heart Circ. Physiol. 2002, 283, H2440–H2449. [Google Scholar] [CrossRef] [Green Version]
  133. Corcoran, J.; Lacey, H.; Baker, P.N.; Wareing, M. Altered potassium channel expression in the human placental vasculature of pregnancies complicated by fetal growth restriction. Hypertens. Pregnancy 2008, 27, 75–86. [Google Scholar] [CrossRef]
  134. U.S. National Library of Medicine. Clinical Trials Involving Potassium Channels during Pregnancy. Available online: https://clinicaltrials.gov/ct2/results?cond=Pregnancy%3B+pregnancy+complications&term=potassium&cntry=&state=&city=&dist=&Search=Search&recrs=a&recrs=b&recrs=d&recrs=e&recrs=f (accessed on 2 August 2022).
Figure 1. Mechanisms underlying the modulation of K+ channels in the uterine vasculature during normal pregnancy. Pregnancy increases uterine blood flow by augmenting vasodilation and/or remodeling of uterine, radial, and myometrial arteries. A critical mechanism for increased vasodilation is greater activation of K+ channels in the endothelium (top panel) and smooth muscle (bottom panel) of these uterine blood vessels. Pink arrows show the effects of pregnancy. ACh, acetylcholine; ADM, adrenomedullin; ADM2, adrenomedullin 2; ATP, adenosine triphosphate; BKCa, large-conductance Ca2+-activated K+ channel; β1, BKCa β1 subunit; cGMP, cyclic guanosine monophosphate; CGRP, calcitonin gene-related peptide; EDHF, endothelial-derived hyperpolarizing factor; ERα, estrogen receptor α; γ1 and γ3, BKCa γ1 and γ3 subunits; H2S, hydrogen sulfide; IK, intermediate-conductance Ca2+-activated K+ channel; KATP, ATP-sensitive K+ channel; KCNMB1, BKCa β1 subunit gene; Kir6.1/6.2, inward-rectifier K+ channel type 6.1 and 6.2; Kv, voltage-gated K+ channel; Kv3.4, voltage-gated K+ channel type 3.4; PKC, protein kinase C; PKG, protein kinase G; RyR1/2, ryanodine receptor type 1 and 2; SK, small-conductance Ca2+-activated K+ channel; SUR2B, sulfonylurea receptor type 2B; TRPV3, transient receptor potential vanilloid type 3.
Figure 1. Mechanisms underlying the modulation of K+ channels in the uterine vasculature during normal pregnancy. Pregnancy increases uterine blood flow by augmenting vasodilation and/or remodeling of uterine, radial, and myometrial arteries. A critical mechanism for increased vasodilation is greater activation of K+ channels in the endothelium (top panel) and smooth muscle (bottom panel) of these uterine blood vessels. Pink arrows show the effects of pregnancy. ACh, acetylcholine; ADM, adrenomedullin; ADM2, adrenomedullin 2; ATP, adenosine triphosphate; BKCa, large-conductance Ca2+-activated K+ channel; β1, BKCa β1 subunit; cGMP, cyclic guanosine monophosphate; CGRP, calcitonin gene-related peptide; EDHF, endothelial-derived hyperpolarizing factor; ERα, estrogen receptor α; γ1 and γ3, BKCa γ1 and γ3 subunits; H2S, hydrogen sulfide; IK, intermediate-conductance Ca2+-activated K+ channel; KATP, ATP-sensitive K+ channel; KCNMB1, BKCa β1 subunit gene; Kir6.1/6.2, inward-rectifier K+ channel type 6.1 and 6.2; Kv, voltage-gated K+ channel; Kv3.4, voltage-gated K+ channel type 3.4; PKC, protein kinase C; PKG, protein kinase G; RyR1/2, ryanodine receptor type 1 and 2; SK, small-conductance Ca2+-activated K+ channel; SUR2B, sulfonylurea receptor type 2B; TRPV3, transient receptor potential vanilloid type 3.
Ijms 23 09446 g001
Table 1. K+ channel openers and blockers, and their effect on uterine vasculature.
Table 1. K+ channel openers and blockers, and their effect on uterine vasculature.
DrugMechanismEffect on Uterine VasculatureRef.
NS-1619BKCa openerRelaxed pregnant UtA[85]
TetraethylammoniumBKCa blockerDecreased UtA blood flow[38]
Increased UtA myogenic tone, reduced BKCa-evoked currents[36]
Diminished ADM-induced UtA vasodilation[59]
Reduced CGRP-dependent UtA vasodilation and prevented CGRP-induced decrease in UtA perfusion pressure [55,56]
IberiotoxinBKCa blockerDecreased late pregnant UtA diameter in wild-type but not BKCa γ1 subunit knockout mouse[37]
Reduced ACh-elicited UtA vasodilation [61]
Diminished H2S-induced UtA vasodilation [40]
NS-309SK openerInduced vasodilation in non-pregnant UtA[83]
ApaminSK blockerReduced EDHF-dependent myometrial artery vasodilation (in combination with TRAM-34 or charybdotoxin)[79]
Decreased UtA vasodilation induced by CGRP, ADM, and ADM2 (in combination with charybdotoxin)[80]
Diminished ACh-induced UtA vasodilation and increased Ca2+ levels in smooth muscle cells (in combination with charybdotoxin)[81]
Reduced ACh-induced vasodilation in non-pregnant UtA (with or without TRAM-34)[83]
Relaxed pregnant UtA[85]
SKA-31IK openerInduced vasodilation in radial UtA (greater in pregnant than non-pregnant) [84]
TRAM-34IK blockerPrevented ACh-induced vasodilation in non-pregnant UtA (with or without apamin)[83]
Decreased TRPV3-evoked vasodilation in radial UtA[84]
CharybdotoxinSK/IK blockerAttenuated EDHF-dependent myometrial artery vasodilation[79]
DiazoxideKATP openerInduced vasodilation of UtA, decreased Ca2+ levels[104]
PinacidilKATP openerReduced PE-dependent UtA vasoconstriction[105]
GlibenclamideKATP blockerIncreased uterine vascular resistance, exacerbated the increase in uterine vascular resistance induced by AngII[106]
4-AminopyridineKv blockerPartially reduced ACh-induced vasodilation of UtA[61]
Increased myogenic tone and Ca2+ levels in radial UtA[129]
BDS-IKv3.4 blockerPrevented proliferation in UtA smooth muscle cells [131]
NS-1619, 1,3-dihydro-1-[2-hydroxy-5-(trifluoromethyl)phenyl]-5-(trifluoromethyl)-2H-benzimidazol-2-one; NS-309, 6,7-dichloro-1H-indole-2,3-dione 3-oxime; SKA-31, naphtho[1,2-d]thiazol-2-ylamine; TRAM-34, 1-[(2-chlorophenyl)diphenylmethyl]-1H-pyrazole; BDS-I, blood-depressing substance-1; UtA, uterine artery; ADM, adrenomedullin; CGRP, calcitonin gene-related peptide; ACh, acetylcholine; H2S, hydrogen sulfide; EDHF, endothelial-derived hyperpolarizing factor; ADM2, adrenomedullin 2; TRPV3, transient receptor potential vanilloid 3; PE, phenylephrine; AngII, angiotensin II.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Bresnitz, W.; Lorca, R.A. Potassium Channels in the Uterine Vasculature: Role in Healthy and Complicated Pregnancies. Int. J. Mol. Sci. 2022, 23, 9446. https://doi.org/10.3390/ijms23169446

AMA Style

Bresnitz W, Lorca RA. Potassium Channels in the Uterine Vasculature: Role in Healthy and Complicated Pregnancies. International Journal of Molecular Sciences. 2022; 23(16):9446. https://doi.org/10.3390/ijms23169446

Chicago/Turabian Style

Bresnitz, Wyanet, and Ramón A. Lorca. 2022. "Potassium Channels in the Uterine Vasculature: Role in Healthy and Complicated Pregnancies" International Journal of Molecular Sciences 23, no. 16: 9446. https://doi.org/10.3390/ijms23169446

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