Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2017 Jul 1.
Published in final edited form as: Steroids. 2016 Mar 3;111:46–53. doi: 10.1016/j.steroids.2016.02.018

Rapid estrogen actions on ion channels: a survey in search for mechanisms

Lee-Ming Kow 1, Donald W Pfaff 1
PMCID: PMC4929851  NIHMSID: NIHMS764569  PMID: 26939826

Abstract

A survey of nearly two hundred reports shows that rapid estrogenic actions can be detected across a range of kinds of estrogens, a range of doses, on a wide range of tissue, cell and ion channel types. Striking is the fact that preparations of estrogenic agents that do not permeate the cell membrane almost always mimic the actions of the estrogenic agents that do permeate the membrane. All kinds of estrogens, ranging from natural ones, through receptor modulators, endocrine disruptors, phytoestrogens, agonists, and antagonists to novel G-1 and STX, have been reported to be effective. For actions on specific types of ion channels, the possibility of opposing actions, in different cases, is the rule, not the exception. With this variety there is no single, specific action mechanism for estrogens per se, although in some cases estrogens can act directly or via some signaling pathways to affect ion channels. We infer that estrogens can bind a large number of substrates/receptors at the membrane surface. As against the variety of subsequent routes of action, this initial step of the estrogen's binding action is the key.

Keywords: BK, estrogen binding, ion channel kinetics, L-type Ca2+ channel, Selective Estrogen Receptor Modulator (SERM), signaling pathway

Introduction

The very existence of rapid estrogenic actions is, by now, well established beyond question. Hereafter we will refer to these simply as ‘rapid actions’ or ‘rapid effects’. It is difficult to conceptualize these actions because they are wildly diversified, starting from the estrogen receptors involved all the way through the diversity of the tissues and cells they affect. Their effects on ion channels offer no exception to this diversity.

There are very few review articles on this subject, and those focus on certain type of channels. The mechanism of one kind of estrogenic agent on a single type of ion channel may be figured out. But this does not help the understanding of the rapid actions as a whole, because the mechanism found in one particular case may not be applied to others. In view of this, we hope to cover as completely as possible the ion channels investigated for rapid estrogenic actions.

The analyses of the reported results

In this review, we use a simple, straightforward method in the attempt to cover mechanisms of rapid actions. We searched the literature for relevant articles and list the important characteristics, such as the type of estrogen and dose use, the type of channel studied, etc., from each article's content in Excel spread sheets. We then used the Sort function to cross-reference different kinds of characteristics to evaluate the possibility of an existing mechanism. Since many reports used multiple kinds of estrogens on multiple types of ion channels, therefore the basis of incidence of occurrence is ‘a case’ rather an article or report. A case is defined as one kind of estrogenic agent acting on a certain subtype of channel. Hence, a report/article may have many cases; or the number of references cited for a particular incidence may be less than the number of incidences mentioned.

Estrogenic agents employed

As expected, 17β-estradiol (E2) is the most frequently (171/276 cases) employed, followed by Selective Estrogen Receptor Modulator (SERM) (61/276) and Endocrine Disruptors (10/276). More interesting is the comparison of permeable and impermeable versions of the same estrogenic agent. As presented in Table 1, in all but one case, impermeable agents mimicked their permeable version or worked by themselves. This result confirms the well accepted concept that rapid actions are mediated by site(s) of receptors on the outside of the cells.

Table 1.

Results showing membrane-impermeable form of estrogens are as effective as their permeable versions.

number of cases Agent* effects Ion Channel References
26 E2=E-BSA (18);
E2=E-HRP (2);
E2=E-peroxidase (1);
Tmx=EBT (2);
Estrone=estrone oximes (1);
Estrone=Quat DME-estradil (1);
BPA=BPA-Ms (1).
↑ (16);
↓ (10).
Ca (12);
K (10);
Na (1);
Ligand-gated (3).
[9-33]
2 E-BSA;
E-BSA-FITC
↑ (2) Ca-L [33, 34]
1 E2≠E-BSA Kv2 [35]
*

equal sign, “=”, similar in action; number in parentheses indicate the number of cases.

Ion channels investigated

The type/subtypes of ion channels investigated are listed in Table 2. Potassium channels are the most popular, followed by calcium, ligand-gated, sodium and lastly chloride channels. Important and interesting in this table is the lack of consistency of estrogen effects on most types of channels. For example, estrogens could exert stimulatory (↑, increase channel activity, activate or open channels) and inhibitory (↓, decrease, block or abolish channel activity, or channel's open probability) effects on BK (big conductance Ca2+- and voltage-activated K+ channels) and Ca-L (L-type Ca2+ channels), the two most investigated channels, and others. These diversity and often conflicting results are very difficult to be reconciled with the idea of a unified mechanism for rapid actions.

Table 2.

Ion channels investigated.

Ion Channels Effects and numbers of rapid estrogen actions Referencesb
Type Subtype a -- others Total
K+ BK 35 12 2 1 50 [3, 7, 16, 17, 24, 29, 32, 36-56] // [1, 13, 44, 57-62] // [63, 64] // [65].
Kdr (or Kr) 1 13 1 0 15 [19, 35, 66-76].
KATP 7 8 0 0 15 [28, 77-89].
Kir 1 10 2 0 13 [66, 71, 73, 74, 90-97].
KA 0 7 3 0 10 [63, 68, 73, 74, 76, 91, 98-100]
GIRK 0 6 0 0 6 [101-105]
Kv 0 2 2 0 4 [9, 32, 35]
hERG 0 4 0 0 4 [106-109]
KCNQ1 0 4 0 0 4 [107, 108, 110, 111]
Misc 6 6 2 0 14 [6, 57, 58, 91, 97, 112-118].
Total 50 72 12 1 135
Ca2+ Ca-L 7 28 2 0 37 [12, 21, 34, 119-122]/[6, 14, 22, 63, 66, 123-141]/[51, 88].
T-type 0 4 1 0 5 [6, 141, 142] // [22]
N-type 1 2 0 0 3 [143] // [22, 126].
Cav 1 8 0 0 9 [5, 61, 84, 144-149].
[Ca++]i 11 6 1 0 18 [10, 11, 18, 30, 33, 86, 114, 150-153] // [15, 28, 148, 154-156] // [157]
Ca2+ influx 6 3 0 0 9 [23, 158-162] // [26, 163, 164].
Misc 4 6 0 0 10 [2, 84, 165-168] // [72, 169, 170].
Total 30 57 4 0 91
Na+ All 2 10 0 0 12 [75, 171] // [31, 66, 68, 73, 97, 107, 108, 172, 173].
Cl- All 2 5 1 0 8 [174, 175] // [66, 111, 176-178] // [66].
Ligand-gated KAR 3 0 1 0 4 [8, 179-181].
nAChR 1 4 0 0 5 [4, 182-184].
NMDAR 4 0 2 0 6 [8, 25, 144, 185-187].
5-HT3R 0 2 0 0 2 [172, 182]
AMPAR 1 0 0 0 1 [181]
P2X3 0 2 0 0 2 [188, 189].
GABAAR 0 1 0 2 3 [190, 191]
Total 9 9 3 2 23
a

symbols: ↑, denote increase, activation; ↓, reduction, inhibition, blockade; --, no significant effect; others, undefined modulation.

b

References for different effects are presented in order in different clusters separated by //.

Search for causes/explanations for the conflicting rapid effects on BK and Ca-L

In attempts to find out possible explanations for these diverse and conflicting results, we focused on BK and Ca-L and made some detailed analyses. These two types of channels were chosen not only because they are the most intensively investigated but also because of a basic almost all difference – BK is more often “stimulated” by estrogens than Ca-L. The results of these analyses are presented in Table 3. First, we looked at the kinds of estrogenic agents used. The agents that caused ↑ as well as ↓, are Bold-faced. One can see almost all kinds of agents that caused ↓ also caused ↑ in BK. Similarly, all those that caused ↑ in Ca-L also induced ↓. Obviously, the conflicts are not due the estrogens used.

Table 3.

Analyses of the conflicting results from BK and Ca-L.

Ion channel Estrogen action Cases n= Estrogens Signaing path Tissue/cell used (n, %) Ref's
Agent Dose
BK 35 E2: n=18;
E2=Tmx, n=2;
E2=PhytoE, n=2;
phytoE, n=3;
BPA, n=2;
Tmx, n=7;
G-1, ICI 182,780, estrone, DPN, PPT, n=1 each;
E2=E-BSA: n=2;
Tmx=imperm, n=2;
BPA=BPA-Ms, n=1;
100 nM to 100 mM cGMP, n=5;
cAMP/PKA, n=1
Muscle (23, 66%);
Neuron (1, 3%);
HEK-293 (3,8%);
MCF-7 (2, 6%);
Others (6, 17%).
[3, 7, 16, 17, 24, 29, 32, 36-56].
12 E2, n=7;
E2 metab, n=2;
ICI 182,780, n=1;
Tmx, n=2;
E2=E-BSA, n=1;
Tmx=imperm,n=2;
100 pM to 30 mM PKA/PKC, n=1 Muscle (3, 25%);
Neuron (4, 33%);
HEK-293 (2, 17%);
Endothelial cells (3, 25%).
[1, 13, 57-62, 192].
Ca-L 7 E2, n=5;
E-BSA, XE, n=1, each;
E2=E-BSA, n=1;
E2=E-peroxidase, n=1.
1 pM to 100 nM cAMP/PKC, n=1. Muscle, n=2 (29%);
Neuron, n=1 (14%); Others, n=4 (57%).
[12, 21, 34, 119-122]
27 E2, n=19;
EB, EE2, DES, XE, phytoE, ICI 172,780, n=1, each;
SERM, n=2;
E2=E-BSA, n=2;
E2=DES=EE2, n=2;
E2=ICI 172,780, n=1.
1-100 mM * PLC-MAPK-CREB, n=1;
G-protein, n=1.
Muscle (16, 59%);
Neuron (9, 33%);
Others (2, 8%).
[6, 14, 22, 63, 66, 123-137, 139-141].
*

except for one study that used 1-100 pM.

We then looked at doses of estrogens used. For BK, the doses that induced either type of response markedly overlapped. Hence the doses used cannot account for the opposite responses of BK channels to estrogens. The situation is different for Ca-L channels. Here ↑responses were induced by low doses ranging from pM to nM, whereas ↓ responses were induced by higher μM doses in all but one case. Thus, for Ca-L channels, the difference in estrogen doses can be a cause for the opposite responses.

The signaling pathways involved were also examined. Unfortunately, the number of cases that investigated these pathways are too few to allow for making firm conclusions. Still, looking for potential clues, we turned to the tissue/cells used. For the BK channel, the weighted proportion of muscle (and, even fewer, neuronal cases) may together account for the occurrence of more ↑responses. The situation is just the opposite for Ca-L channels, where the weighted muscle proportion is associated with ↓responses. Thus, the response of an ion channel to rapid estrogenic action does not appear to be dependent on the kind of estrogen agents, but may be affected by the dose and/or the preparation employed.

Direct rapid estrogen actions on ion channels

The direct action is one of the most attractive mechanisms proposed for rapid actions. It states that estrogens act by binding directly on parts of ion channels. It was based mostly on precise single-channel recordings (mostly from excised inside-out patches, see references in Table 4), but also on other methods, such as binding studies [1-3], site mutations [4], pharmacological and biophysical analysis [5], kinetics of estrogen effects [6], etc. Results from such studies are presented in Table 4. From the list of estrogen used it appears almost all kinds of estrogenic agents can act directly on ion channels, with a possible exception of G-1. When effective, the direct actions of estrogens are divided about evenly in either direction on the channels, except for BK. Close examination of the results of the three inhibitory cases raised the possibilities that: 1) the E2 metabolite, 2ME2, might act on a “negative” site(s) on BK where other estrogens won't bind; and/or 2) the BKs on endothelial cells are different (perhaps structurally or perhaps by subtype unit) from BKs in other tissue/cells. Of course these possibilities remain to be further evaluated.

Table 4.

Results of experiments designed to investigate the possibility of estrogens acting/binding directly to ion channels.

Methods Number of cases Estrogenic agents Doses Ion channels Effects References
Single-channel recording of BK 21 E2 (10);
ICI 182780 (2);
2ME2 (2);
DPN≠E2 (2);
BPA (1);
BPA=BPA-Ms (1);
Tmx (5);
4-OH Tmx (1);
EBT (1);
PPT+DPN (1).
mM (11);
nM (7).
BK (21) ↑(18);
↓ (3).
[1, 3, 9-27].
Single-channel recording 4 E2 (2);
2ME2;
Tmx.
mM (4) [Ca2+]i;
K;
Slack (2);
Cl-
↑ (2);
↓ (2).
[11, 28-30].
Single-channel recording 3 E2 (2);
G-1.
100 nM (G-1) BK;
NMDA;
KAR.
-- (3) [7, 8]
Single-channel recording and others 2 ERb1 5-20 nM;
Tmx >10 mM
5-20 nM; >10 mM. Ca-RyR;
Cl-
↑ (1);
↓ (1).
[2, 31]
Other than single-channel recording 7 E2 (3); Tmx, EBT;
BPA;
DES=EE2=E2
mM (5) 5-HT3;
Kdr;
Ca-L;
Cav;
nAChR
↑ (2);
↓ (4).
[4-6, 32-34].

Many of the studies on direct actions used high doses of estrogens, in μM ranges, especially when working on muscle preparations. This does not, however, discredit these observations because such studies were usually conducted on stripped down preparations, such as excised, cell-free patches or even artificial bilayers accompanied by no or very little cytoplasm. The reason for using high doses could be that estrogens may also act on something else to work synergistically with the direct actions. As will be further discussed later, rapid activation of signaling systems would be a good candidate for this.

Another interesting result is the lack of direct rapid action in three cases. The lack of an effect on the BK [7] is a big surprise in view of the twenty one positive cases. The one possible reason we could find is that in this particular case G-1, the GPER agonist, was used. Is it possible that G-1 binds only to G-protein coupled receptors? It would be interesting to check out whether G-1 can bind to BK or to other ion channel.

Two of the negative cases come from the same paper [8]. It reported that E2 had no effect on the two glutamate's ligand-gated channels, NMDA or KAR. As far as we can see, this is the only single-channel recording study on these two channels. Since both types of ligand-gated channels are affected by estrogens (see Table 2 and 5), these negative results indicate that the direct action, solidly demonstrated when positive, is not the only mechanism underlying rapid estrogen action.

Table 5.

Rapid estrogen actions involving signaling systems.

Signaling system involved Number of cases Estrogenic agents Doses Ion channels Effects Ref's
cGMP-PKG-NO 11 E2 (10);
Tmx (1);
gisenoside Re (2);
E2=E-HRP (2);
E2=DES=BPA (1)
all in nM [Ca2+]i (3);
BK (5);
KATP (3);
Ca2+ oscillation (1);
Ks (1).
↑ (7);
↓ (7).
[28, 39, 49, 52, 56, 67, 86, 154, 155].
MAPK-ERK1/2-CREB 7 E2 (3);
E2=E-BSA (1);
E-BSA-TITC (1);
E2=G-1=PPT (1);
ICI 182780 (1)
10 to 100 nM Ca-L (3);
[Ca2+]i (1);
BK (1);
P2X3 (1);
ASIC (1).
↑ (4);
↓ (2);
other (1).
[33, 65, 120, 123, 166, 189, 193].
cAMP 7 E2 (4);
E2=G-1=PPT (1);
E-BSA (1);
DES (1).
10 pM to 100 nM Ca: Ca-L, Cav;
K: BK, KATP, K;
Ligand-gated: AMPA/KAR, P2X3.
↑ (5);
↓ (2).
[34, 53, 82, 113, 160, 181, 189].
G-proteins;
G-proteins/PLC
8 E2 (3);
E2=E-BSA (5);
E2=E-BSA=STX (1)
nM (5);
mM (3).
Ca: Ca-L, Ca-T, Cav, [Ca2+]i;
K: K, Kir;
Ligand-gated: KAR.
↑ (4);
↓ (4).
[18, 22, 23, 96, 113, 146, 161, 179].
PIP2 3 Tmx (1);
Tmx=4OH
Tmx=Raloxifen (1);
Tmx=4OH Tmx (1).
nM to mM. Kir (1);
KATP (1);
GIRK (1).
↓ (3) [94, 105]
PKA 5 E2 (3);
E2=G-1 (1);
E2=G-1=PPT (1).
10-100 nM BK (1); [Ca2+]i (1); AMPA/KAR (1); P2X3 (1); Nav (1). ↑ (4);
↓ (1).
[53, 166, 171, 181, 189].
PKC 4 E2 (3);
E2=E-BSA (1);
1-100 nM Ca (1); [Ca2+]i (2); Cl (1). ↑ (3);
↓ (1).
[18, 166, 167, 176].
PKA and PKC 9 E2 (6);
E2=E-BSA (2);
BPA (1).
nM (5);
mM (2).
[Ca2+]i (3); Ca HVA (1); KATP (1); KCNQ1 (1); GIRK (1); Na (2). ↑ (4);
↓ (4);
other (1).
[18, 31, 89, 101, 111, 149, 150, 166, 173].
Misc 5 E2 (1);
E2=E-BSA (3);
CEE (1).
nM (2);
mM (1).
[Ca2+]i (1); Cav (1); NMDA (2). ↑ (3);
↓ (2).
[11, 19, 25, 145, 186].

Rapid estrogen actions via signaling pathways/systems

There are many studies using agonists or, more often, inhibitors to activate or block certain signaling pathways and subsequently observe the effect on rapid estrogen actions. The results of the studies available are listed in Table 5. There are a few things that are obvious. First, a wide variety of signaling systems are involved in mediating rapid estrogen actions. Second, a wide variety of estrogenic agents are effective. They include E2, SERMs, endocrine disruptors, phytoestrogens, ER agonists, the G-1 that did not have direct action, and the novel STX. Third, unlike works on direct actions, studies shown in Table 5 generally use estrogen doses in nM. This may indicate that rapid actions mediated by signaling systems are more sensitive to estrogens. However, since this kind of investigation is usually done with intact cells, estrogens might activate more than one type of rapid actions, e.g., both direct action and signaling path mediated (see more discussion about this below). Fourth, a wide variety of type and subtype of ion channels are affected by rapid actions, including NMDAR and KAR that were not susceptible to direct action (see Table 4). Fifth, the effects of estrogen action are, as usual, pretty evenly stimulatory and inhibitory. We could not find any association between the effects with estrogen types, doses or types of ion channels.

The actual scope of rapid actions via signaling pathways may be wider than that shown in Table 5, because investigators usually attempted to look at the specific pathway they are interested in and only if they already suspected that the pathway was involved.

The origin of the wide-spread rapid action diversity

The most important of our literature survey is the finding that almost all using impermeable estrogens found them effective as permeable ones. This is important because it implies that rapid actions are induced by estrogens working outside the cell. They don't have to, and probably do not, get inside the cell to bind and activate second messengers. Therefore, we can safely assume that the majority, if not all, of rapid actions are originated at estrogen binding sites at membrane's extracellular surface. When substrate is bound, it could trigger a build-in action specific to this substrate. Since estrogens are promiscuous in binding, as indicated by the wide range of estrogens that are effective and the continuous finding of new membrane receptors for them, it is no wonder that rapid estrogen actions are so diverse. Thus, the initial step, the binding of estrogens to membrane substrates, dictate the ensuing estrogen action and the promiscuous binding is the origin of the diversity rapid actions. Hence, this initial step is the key to the understanding of rapid estrogen actions.

Acknowledgments

This work was supported by a by a grant HD 05751 from NIH.

Abbreviations

[Ca++]i

intracellular Ca2+ concentration

2ME2

2-Methoxyestradiol, a natural metabolite of estradiol

4-OH-Tmx

4-Hydroxytamoxifen, a metabolite of Tmx

5-HT3R

5-hydroxytryptamine receptor subtype 3

AMPAR

α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor

BBP

Butyl benzyl phthalate, a plasticizer and an environmental pollutant, exerts genomic estrogenic-like effects via estrogen receptors

BK, or Maxi-K or slo-1

big conductance Ca2+- and voltage-activated K+ channel

BPA

Besphenol A

BPA-Ms

BPA monosulfate, membrane-impermeable

Ca-L

L-type Ca2+ channel

cAMP

cyclic adenosine monophosphate

Ca-N

N-type Ca2+ channel

Ca-T

T-type Ca2+ channel

Cav

voltage-gated Ca2+ channel

cGMP

cyclic guanosine monophosphate

CREB

cAMP response element-binding protein

DES

diethylstilbestrol, an endocrine disruptor

DPN

diarylpropionitrile, ERβ agonist

DTP

diethyl terephthalate, has estrogenic actions

E2

17β-estradiol

EB

17β-estradiol benzoate

E-BSA

E2 covalently linked to membrane impermeable BSA (bovine serum albumin

E-BSA-FITC

E-BSA conjugated to fluorescein isothiocyanate

EBT

ethylbromide Tmx, impermeable

Edrp

endocrine disruptor

EE2

ethynylestradiol a derivative of E2

E-HRP

E2 conjugated to horseradish peroxidase

ERK1/2

extracellular signal-regulated kinase 1/2

ERβ1

the long form estrogen receptor subtype β

GABAAR

γ-aminobutyric acid receptor subtype A

GIRK

G protein-coupled inwardly-rectifying potassium channels

hERG

human Ether-à-go-go-Related Gene encoded K+ channel

IsK

slowly activating, voltage-depend K+ current

KA

transient A-type K+ channels

KAR

kainate-gated receptor/channel

KATP

ATP-sensitive K+ channel

KCNQ1

voltage-gated K+ channel subfamily KQT member 1

Kdr (or Kr)

delayed rectifier K+ channel

Kir

inward rectifier K+ channel

Kv

voltage-gated K+ channel

MAPK

mitogen-activated protein kinases

nAChR

nicotinic acetylcholine receptor

NMDAR

N-methyl-D-aspartate receptor

NO

nitric oxide

P2X3

P2X purinoceptor 3 channel

phytoE

phytoestrogen

PIP2

phosphatidylinositol 4,5-bisphosphate or PtdIns(4,5)P2

PKA

protein kinase A

PKC

protein kinase C

PKG

protein kinase G

PLC

phospholipase C

PPT

propylpyrazoletriol, ERα agonist

SERM

selective estrogen receptor modulator

STX

diphenylacrylamide compound (does not bind to classical estrogen receptors)

Tmx

tamaxifen

TPE

triphenylethylene, a SERM

XE

xenoestrogen

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Korovkina VP, et al. Estradiol binding to maxi-K channels induces their down-regulation via proteasomal degradation. J Biol Chem. 2004;279:1217–23. doi: 10.1074/jbc.M309158200. [DOI] [PubMed] [Google Scholar]
  • 2.Rybalchenko V, et al. The unliganded long isoform of estrogen receptor beta stimulates brain ryanodine receptor single channel activity alongside with cytosolic Ca2+ J Recept Signal Transduct Res. 2009;29:326–341. doi: 10.3109/10799890903295168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Valverde MA, et al. Acute activation of maxi-K channels (hSlo) by estradiol binding to the b subunit. Science. 1999;285:1929–31. doi: 10.1126/science.285.5435.1929. [DOI] [PubMed] [Google Scholar]
  • 4.Paradiso K, Zhang J, Steinbach JH. The C terminus of the human nicotinic a4b2 receptor forms a binding site required for potentiation by an estrogenic steroid. J Neurosci. 2001;21:6561–8. doi: 10.1523/JNEUROSCI.21-17-06561.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Deutschmann A, et al. Bisphenol A inhibits voltage-activated Ca2+ channels in vitro: mechanisms and structural requirements. Mol Pharmacol. 2013;83:501–11. doi: 10.1124/mol.112.081372. [DOI] [PubMed] [Google Scholar]
  • 6.Nakajima T, et al. Antiarrhythmic effect and its underlying ionic mechanism of 17β-estradiol in cardiac myocytes. Br J Pharmacol. 1999;127:429–40. doi: 10.1038/sj.bjp.0702576. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Yu X, et al. Activation of G protein-coupled estrogen receptor induces endothelium-independent relaxation of coronary artery smooth muscle. Am J Physiol Endocrinol Metab. 2011;301:26. doi: 10.1152/ajpendo.00037.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Wong M, Moss RL. Patch-clamp analysis of direct steroidal modulation of glutamate receptor-channels. J Neuroendocrinol. 1994;6:347–55. doi: 10.1111/j.1365-2826.1994.tb00592.x. [DOI] [PubMed] [Google Scholar]
  • 9.Allen MC, et al. Membrane impermeant antioestrogens discriminate between ligand- and voltage-gated cation channels in NG108-15 cells. Biochimica et Biophysica Acta. 2000;1509:229–36. doi: 10.1016/s0005-2736(00)00297-2. [DOI] [PubMed] [Google Scholar]
  • 10.Benten WP, et al. Estradiol binding to cell surface raises cytosolic free calcium in T cells. FEBS Lett. 1998;422:349–53. doi: 10.1016/s0014-5793(98)00039-8. [DOI] [PubMed] [Google Scholar]
  • 11.Beyer C, Raab H. Nongenomic effects of oestrogen: embryonic mouse midbrain neurones respond with a rapid release of calcium from intracellular stores. Eur J Neurosci. 1998;10:255–62. doi: 10.1046/j.1460-9568.1998.00045.x. [DOI] [PubMed] [Google Scholar]
  • 12.Bulayeva NN, et al. Mechanisms of membrane estrogen receptor-alpha-mediated rapid stimulation of Ca2+ levels and prolactin release in a pituitary cell line. Am J Physiol Endocrinol Metab. 2005;288:19. doi: 10.1152/ajpendo.00349.2004. [DOI] [PubMed] [Google Scholar]
  • 13.Carrer HF, Araque A, Buno W. Estradiol regulates the slow Ca2+-activated K+ current in hippocampal pyramidal neurons. J Neurosci. 2003;23:6338–44. doi: 10.1523/JNEUROSCI.23-15-06338.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Chaban VV, et al. Estradiol inhibits atp-induced intracellular calcium concentration increase in dorsal root ganglia neurons. Neuroscience. 2003;118:941–8. doi: 10.1016/s0306-4522(02)00915-6. [DOI] [PubMed] [Google Scholar]
  • 15.Chaban VV, Micevych PE. Estrogen receptor-a mediates estradiol attenuation of ATP-induced Ca2+ signaling in mouse dorsal root ganglion neurons. J Neurosci Res. 2005;81:31–37. doi: 10.1002/jnr.20524. [DOI] [PubMed] [Google Scholar]
  • 16.Coiret G, et al. 17β-estradiol activates maxi-K channels through a non-genomic pathway in human breast cancer cells. FEBS Lett. 2005;579:2995–3000. doi: 10.1016/j.febslet.2005.02.085. [DOI] [PubMed] [Google Scholar]
  • 17.Dick GM, Hunter AC, Sanders KM. Ethylbromide tamoxifen, a membrane-impermeant antiestrogen, activates smooth muscle calcium-activated large-conductance potassium channels from the extracellular side. Mol Pharmacol. 2002;61:1105–13. doi: 10.1124/mol.61.5.1105. [DOI] [PubMed] [Google Scholar]
  • 18.Doolan CM, Harvey BJ. A Gas protein-coupled membrane receptor, distinct from the classical oestrogen receptor, transduces rapid effects of oestradiol on [Ca2+]i in female rat distal colon. Mol Cell Endocrinol. 2003;199:87–103. doi: 10.1016/s0303-7207(02)00303-9. [DOI] [PubMed] [Google Scholar]
  • 19.Fatehi M, Kombian SB, Saleh TM. 17β-estradiol inhibits outward potassium currents recorded in rat parabrachial nucleus cells in vitro. Neuroscience. 2006;135:1075–1086. doi: 10.1016/j.neuroscience.2005.07.024. [DOI] [PubMed] [Google Scholar]
  • 20.Drici MD, et al. Sex hormones prolong the QT interval and downregulate potassium channel expression in the rabbit heart. Circulation. 1996;94:1471–4. doi: 10.1161/01.cir.94.6.1471. [DOI] [PubMed] [Google Scholar]
  • 21.Gao Q, et al. Nonylphenol affects myocardial contractility and L-type Ca2+ channel currents in a non-monotonic manner via G protein-coupled receptor 30. Toxicology. 2015;334:122–9. doi: 10.1016/j.tox.2015.06.004. [DOI] [PubMed] [Google Scholar]
  • 22.Lee DY, et al. 17β-estradiol inhibits high-voltage-activated calcium channel currents in rat sensory neurons via a non-genomic mechanism. Life Sciences. 2002;70:2047–59. doi: 10.1016/s0024-3205(01)01534-x. [DOI] [PubMed] [Google Scholar]
  • 23.Lieberherr M, et al. Cell signaling and estrogens in female rat osteoblasts: a possible involvement of unconventional nonnuclear receptors. J Bone Miner Res. 1993;8:1365–76. doi: 10.1002/jbmr.5650081111. [DOI] [PubMed] [Google Scholar]
  • 24.Maher J, et al. Smooth muscle relaxation and activation of the large conductance Ca++-activated K+ (BK(Ca)) channel by novel oestrogens. Br J Pharmacol. 2013;169:1153–65. doi: 10.1111/bph.12211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.McRoberts JA, et al. Sex-dependent differences in the activity and modulation of N-methyl-d-aspartic acid receptors in rat dorsal root ganglia neurons. Neuroscience. 2007;148:1015–20. doi: 10.1016/j.neuroscience.2007.07.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Morales A, et al. Estradiol modulates acetylcholine-induced Ca2+ signals in LHRH-releasing GT1-7 cells through a membrane binding site. Eur J Neurosci. 2003;18:2505–14. doi: 10.1046/j.1460-9568.2003.02997.x. [DOI] [PubMed] [Google Scholar]
  • 27.Kelly MJ, Qiu J, Ronnekleiv OK. Estrogen modulation of G-protein-coupled receptor activation of potassium channels in the central nervous system. Annals of the New York Academy of Sciences. 2003;1007:6–16. doi: 10.1196/annals.1286.001. [DOI] [PubMed] [Google Scholar]
  • 28.Ripoll C, et al. Rapid regulation of pancreatic alpha- and beta- cell signalling systems by estrogens. Infect Disord Drug Targets. 2008;8:61–4. doi: 10.2174/187152608784139668. [DOI] [PubMed] [Google Scholar]
  • 29.Rottgen TS, et al. Bisphenol A activates BK channels through effects on alpha and beta1 subunits. Channels. 2014;8:249–57. doi: 10.4161/chan.27709. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Stefano GB, et al. Cell-surface estrogen receptors mediate calcium-dependent nitric oxide release in human endothelia. Circulation. 2000;101:1594–7. doi: 10.1161/01.cir.101.13.1594. [DOI] [PubMed] [Google Scholar]
  • 31.Wang Q, et al. Effects of estradiol on voltage-gated sodium channels in mouse dorsal root ganglion neurons. Brain Research. 2013;1512:1–8. doi: 10.1016/j.brainres.2013.02.047. [DOI] [PubMed] [Google Scholar]
  • 32.Wong CM, et al. Differential effects of estrogen and progesterone on potassium channels expressed in Xenopus oocytes. Steroids. 2008;73:272–9. doi: 10.1016/j.steroids.2007.10.010. [DOI] [PubMed] [Google Scholar]
  • 33.Wu TW, Chen S, Brinton RD. Membrane estrogen receptors mediate calcium signaling and MAP kinase activation in individual hippocampal neurons. Brain Res. 2011;16:34–43. doi: 10.1016/j.brainres.2011.01.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Han HJ, Lee YH, Park SH. Estradiol-17β-BSA stimulates Ca2+ uptake through nongenomic pathways in primary rabbit kidney proximal tubule cells: involvement of cAMP and PKC. J Cell Physiol. 2000;183:37–44. doi: 10.1002/(SICI)1097-4652(200004)183:1<37::AID-JCP5>3.0.CO;2-N. [DOI] [PubMed] [Google Scholar]
  • 35.Druzin M, et al. Mechanism of estradiol-induced block of voltage-gated K+ currents in rat medial preoptic neurons. PLoS One. 2011;6:20. doi: 10.1371/journal.pone.0020213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Asano S, Tune JD, Dick GM. Bisphenol A activates Maxi-K (K(Ca)1.1) channels in coronary smooth muscle. Br J Pharmacol. 2010;160:160–70. doi: 10.1111/j.1476-5381.2010.00687.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Behrens R, et al. hKCNMB3 and hKCNMB4, cloning and characterization of two members of the large-conductance calcium-activated potassium channel beta subunit family. FEBS Letters. 2000;474:99–106. doi: 10.1016/s0014-5793(00)01584-2. [DOI] [PubMed] [Google Scholar]
  • 38.Coiret G, et al. The antiestrogen tamoxifen activates BK channels and stimulates proliferation of MCF-7 breast cancer cells. Mol Pharmacol. 2007;71:843–51. doi: 10.1124/mol.106.028290. [DOI] [PubMed] [Google Scholar]
  • 39.Darkow DJ, Lu L, White RE. Estrogen relaxation of coronary artery smooth muscle is mediated by nitric oxide and cGMP. Am J Physiol. 1997;272:H2765–73. doi: 10.1152/ajpheart.1997.272.6.H2765. [DOI] [PubMed] [Google Scholar]
  • 40.De Wet H, et al. Modulation of the BK channel by estrogens: examination at single channel level. Mol Membr Biol. 2006;23:420–9. doi: 10.1080/09687860600802803. [DOI] [PubMed] [Google Scholar]
  • 41.Dick G, Sanders K. (Xeno)estrogen sensitivity of smooth muscle BK channels conferred by the regulatory b1 subunit: a study of b1 knockout mice. J Biol Chem. 2001;276:44835–40. doi: 10.1074/jbc.M106851200. [DOI] [PubMed] [Google Scholar]
  • 42.Dick G, et al. Tamoxifen activates smooth muscle BK channels through the regulatory β1 subunit. J Biol Chem. 2001;276:34594–9. doi: 10.1074/jbc.M104689200. [DOI] [PubMed] [Google Scholar]
  • 43.Dick GM. The pure anti-oestrogen ICI 182,780 (Faslodex) activates large conductance Ca2+-activated K+ channels in smooth muscle. Br J Pharmacol. 2002;136:961–4. doi: 10.1038/sj.bjp.0704807. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Duncan RK. Tamoxifen alters gating of the BK a subunit and mediates enhanced interactions with the avian b subunit. Biochem Pharmacol. 2005;70:47–58. doi: 10.1016/j.bcp.2005.03.026. [DOI] [PubMed] [Google Scholar]
  • 45.Han G, et al. Estrogen receptor alpha mediates acute potassium channel stimulation in human coronary artery smooth muscle cells. J Pharmacol Exp Ther. 2006;316:1025–30. doi: 10.1124/jpet.105.093542. [DOI] [PubMed] [Google Scholar]
  • 46.Kim SC, et al. Relaxation of rabbit cavernous smooth muscle to 17β estradiol: a non-genomic, NO-independent mechanism. Asian J Androl. 2004;6:127–131. [PubMed] [Google Scholar]
  • 47.King JT, et al. β2 and β4 Subunits of BK Channels Confer Differential Sensitivity to Acute Modulation by Steroid Hormones. Journal of Neurophysiology. 2006;95:2878–2888. doi: 10.1152/jn.01352.2005. [DOI] [PubMed] [Google Scholar]
  • 48.Li Z, Hay M. 17β-estradiol modulation of area postrema potassium currents. J Neurophysiol. 2000;84:1385–1391. doi: 10.1152/jn.2000.84.3.1385. [DOI] [PubMed] [Google Scholar]
  • 49.Nakaya Y, et al. The phytoestrogen ginsensoside Re activates potassium channels of vascular smooth muscle cells through PI3K/Akt and nitric oxide pathways. J Med Invest. 2007;54:381–4. doi: 10.2152/jmi.54.381. [DOI] [PubMed] [Google Scholar]
  • 50.Nevala R, et al. Calcium-sensitive potassium channel inhibitors antagonize genistein- and daidzein-induced arterial relaxation in vitro. Life Sci. 2001;69:1407–17. doi: 10.1016/s0024-3205(01)01233-4. [DOI] [PubMed] [Google Scholar]
  • 51.Provence A, et al. Regulation of Guinea Pig Detrusor Smooth Muscle Excitability by 17beta-Estradiol: The Role of the Large Conductance Voltage- and Ca2+-Activated K+ Channels. PLoS One. 2015;10:e0141950. doi: 10.1371/journal.pone.0141950. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Rosenfeld CR, et al. Calcium-activated potassium channels and nitric oxide coregulate estrogen-induced vasodilation. Am J Physiol Heart Circ Physiol. 2000;279:H319–28. doi: 10.1152/ajpheart.2000.279.1.H319. [DOI] [PubMed] [Google Scholar]
  • 53.Suzuki K, et al. Non-genomic action of 17b-estradiol on opening of Ca2+- and voltage-activated K+ channel in lacrimal acinar cells. Tokai J Exp Clin Med. 2004;29:71–8. [PubMed] [Google Scholar]
  • 54.Wang YJ, et al. The activation by estrogen receptor agonists of the BKCa-channel in human cardiac fibroblasts. Biochem Pharmacol. 2007;73:1347–57. doi: 10.1016/j.bcp.2006.12.029. [DOI] [PubMed] [Google Scholar]
  • 55.White RE, Darkow DJ, Lang JL. Estrogen relaxes coronary arteries by opening BKCa channels through a cGMP-dependent mechanism. Circ Res. 1995;77:936–42. doi: 10.1161/01.res.77.5.936. [DOI] [PubMed] [Google Scholar]
  • 56.White RE, et al. Endothelium-independent effect of estrogen on Ca2+-activated K+ channels in human coronary artery smooth muscle cells. Cardiovasc Res. 2002;53:650–61. doi: 10.1016/s0008-6363(01)00428-x. [DOI] [PubMed] [Google Scholar]
  • 57.Chiang HT, Wu SN. Inhibition of large-conductance calcium-activated potassium channel by 2-methoxyestradiol in cultured vascular endothelial (HUV-EC-C) cells. J Memb Biol. 2001;182:203–12. doi: 10.1007/s00232-001-0044-y. [DOI] [PubMed] [Google Scholar]
  • 58.Chu Z, et al. Differential Regulation of Gonadotropin-Releasing Hormone Neuron Activity and Membrane Properties by Acutely Applied Estradio: Dependence on Dose and Estrogen Receptor Subtype. J Neurosci. 2009;29:5616–5627. doi: 10.1523/JNEUROSCI.0352-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Kelly MJ, et al. Estrogen modulation of K+ channel activity in hypothalamic neurons involved in the control of the reproductive axis. Steroids. 2002;67:447–456. doi: 10.1016/s0039-128x(01)00181-7. [DOI] [PubMed] [Google Scholar]
  • 60.Liu YC, et al. Inhibitory action of ICI-182,780, an estrogen receptor antagonist, on BKCa channel activity in cultured endothelial cells of human coronary artery. Biochem Pharmacol. 2003;66:2053–63. doi: 10.1016/s0006-2952(03)00584-7. [DOI] [PubMed] [Google Scholar]
  • 61.Okabe K, Inoue Y, Soeda H. Estradiol inhibits Ca2+ and K+ channels in smooth muscle cells from pregnant rat myometrium. Eur J Pharmacol. 1999;376:101–8. doi: 10.1016/s0014-2999(99)00353-2. [DOI] [PubMed] [Google Scholar]
  • 62.Wagner EJ, Ronnekleiv OK, Kelly MJ. The noradrenergic inhibition of an apamin-sensitive, small-conductance Ca2+-activated K+ channel in hypothalamic gamma-aminobutyric acid neurons: pharmacology, estrogen sensitivity, and relevance to the control of the reproductive axis. J Pharmacol Exptl Ther. 2001;299:21–30. [PubMed] [Google Scholar]
  • 63.Cairrao E, et al. Non-genomic vasorelaxant effects of 17b-estradiol and progesterone in rat aorta are mediated by L-type Ca2+ current inhibition. Acta Pharmacol Sinica. 2012;33:615–24. doi: 10.1038/aps.2012.4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Nishimura I, et al. 17β-estradiol at physiological concentrations augments Ca2+ -activated K+ currents via estrogen receptor b in the gonadotropin-releasing hormone neuronal cell line GT1-7. Endocrinology. 2008;149:774–82. doi: 10.1210/en.2007-0759. [DOI] [PubMed] [Google Scholar]
  • 65.Wu SN, Wu AZ, Lin MW. Pharmacological roles of the large-conductance calcium-activated potassium channel. Curr Top Med Chem. 2006;6:1025–30. doi: 10.2174/156802606777323764. [DOI] [PubMed] [Google Scholar]
  • 66.Borg JJ, et al. Inhibitory effects of the antiestrogen agent clomiphene on cardiac sarcolemmal anionic and cationic currents. J Pharmacol Exptl Therap. 2002;303:282–92. doi: 10.1124/jpet.102.038901. [DOI] [PubMed] [Google Scholar]
  • 67.Furukawa T, et al. Ginsenoside Re, a main phytosterol of Panax ginseng, activates cardiac potassium channels via a nongenomic pathway of sex hormones. Mol Pharmacol. 2006;70:1916–24. doi: 10.1124/mol.106.028134. [DOI] [PubMed] [Google Scholar]
  • 68.Hardy SP, deFelipe C, Valverde MA. Inhibition of voltage-gated cationic channels in rat embryonic hypothalamic neurones and C1300 neuroblastoma cells by triphenylethylene antioestrogens. FEBS Letters. 1998;4343:236–40. doi: 10.1016/s0014-5793(98)00974-0. [DOI] [PubMed] [Google Scholar]
  • 69.Lee JH, Marcus DC. Estrogen acutely inhibits ion transport by isolated stria vascularis. Hearing Res. 2001;158:123–30. doi: 10.1016/s0378-5955(01)00316-1. [DOI] [PubMed] [Google Scholar]
  • 70.Liu H, et al. Raloxifene inhibits transient outward and ultra-rapid delayed rectifier potassium currents in human atrial myocytes. Eur J Pharmacol. 2007;563:61–8. doi: 10.1016/j.ejphar.2007.01.072. [DOI] [PubMed] [Google Scholar]
  • 71.Zhang Y, et al. Inhibitory effects of estradiol on inward rectifier and delayed rectifier K+ currents in guinea pig ventricular myocytes. Acta Pharmacol Sinica. 1999;207:631–4. [PubMed] [Google Scholar]
  • 72.Tanabe S, Hata T, Hiraoka M. Effects of estrogen on action potential and membrane currents in guinea pig ventricular myocytes. Am J Physiol. 1999;277:H826–33. doi: 10.1152/ajpheart.1999.277.2.H826. [DOI] [PubMed] [Google Scholar]
  • 73.He J, et al. Tamoxifen inhibits Na+ and K+ currents in rat ventricular myocytes. Am J Physiol Heart Circ Physiol. 2003;285:17. doi: 10.1152/ajpheart.00686.2002. [DOI] [PubMed] [Google Scholar]
  • 74.Liu XK, et al. The antiestrogen tamoxifen blocks the delayed rectifier potassium current, IKr, in rabbit ventricular myocytes. J Pharmacol Exp Ther. 1998;287:877–83. [PubMed] [Google Scholar]
  • 75.Kow LM, Easton A, Pfaff DW. Acute estrogen potentiates excitatory responses of neurons in rat hypothalamic ventromedial nucleus. Brain Res. 2005;1043:124–131. doi: 10.1016/j.brainres.2005.02.068. [DOI] [PubMed] [Google Scholar]
  • 76.Knock GA, et al. Modulation of potassium current characteristics in human myometrial smooth muscle by 17beta-estradiol and progesterone. Biol Reprod. 2001;64:1526–34. doi: 10.1095/biolreprod64.5.1526. [DOI] [PubMed] [Google Scholar]
  • 77.Das B, Sarkar C. Similarities between ischemic preconditioning and 17β-estradiol mediated cardiomyocyte KATP channel activation leading to cardioprotective and antiarrhythmic effects during ischemia/reperfusion in the intact rabbit heart. J Cardiovasc Pharmacol. 2006;47:277–86. doi: 10.1097/01.fjc.0000202563.54043.d6. [DOI] [PubMed] [Google Scholar]
  • 78.Lee TM, et al. Effect of estrogen on ventricular repolarization in menopausal patients with syndrome X and effects of nicorandil. Amer J Cardiol. 1999;84:65–9. doi: 10.1016/s0002-9149(99)00193-9. [DOI] [PubMed] [Google Scholar]
  • 79.Lee TM, et al. Cardioprotective effects of 17β-estradiol produced by activation of mitochondrial ATP-sensitive K+ Channels in canine hearts. J Mol Cellu Cardiol. 2000;32:1147–58. doi: 10.1006/jmcc.2000.1167. [DOI] [PubMed] [Google Scholar]
  • 80.Lee TM, et al. Pharmacologic preconditioning of estrogen by activation of the myocardial adenosine triphosphate-sensitive potassium channel in patients undergoing coronary angioplasty. J Am Coll Cardiol. 2002;39:871–7. doi: 10.1016/s0735-1097(01)01816-2. [DOI] [PubMed] [Google Scholar]
  • 81.Lee TM, Chou TF, Tsai CH. Differential role of KATP channels activated by conjugated estrogens in the regulation of myocardial and coronary protective effects. Circulation. 2003;107:49–54. doi: 10.1161/01.cir.0000043243.49875.2e. [DOI] [PubMed] [Google Scholar]
  • 82.Martinez C, et al. Involvement of KATP channels in diethylstilbestrol-induced relaxation in rat aorta. Eur J Pharmacol. 2001;413:109–16. doi: 10.1016/s0014-2999(01)00749-x. [DOI] [PubMed] [Google Scholar]
  • 83.Nadal A, et al. Rapid insulinotropic effect of 17β-estradiol via a plasma membrane receptor. Faseb J. 1998;12:1341–8. doi: 10.1096/fasebj.12.13.1341. [DOI] [PubMed] [Google Scholar]
  • 84.Nadal A, et al. Estrogen and xenoestrogen actions on endocrine pancreas: from ion channel modulation to activation of nuclear function. Steroids. 2004;69:531–6. doi: 10.1016/j.steroids.2004.05.010. [DOI] [PubMed] [Google Scholar]
  • 85.Ogata R, et al. Inhibitory effects of genistein on ATP-sensitive K+ channels in rabbit portal vein smooth muscle. Br J Pharmacol. 1997;122:1395–404. doi: 10.1038/sj.bjp.0701532. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Ropero AB, et al. Non-genomic actions of 17β-oestradiol in mouse pancreatic β-cells are mediated by a cGMP-dependent protein kinase. J Physiol. 1999;521:397–407. doi: 10.1111/j.1469-7793.1999.00397.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Soriano S, et al. Rapid regulation of KATP channel activity by 17β-estradiol in pancreati β-cells involves the estrogen receptor b and the atrial natriuretic peptide receptor. Mol Endocrinol. 2009;23:1973–82. doi: 10.1210/me.2009-0287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Tsang SY, et al. Effect of 17β-estradiol exposure on vasorelaxation induced by K+ channel openers and Ca2+ channel blockers. Pharmacology. 2002;65:26–31. doi: 10.1159/000056182. [DOI] [PubMed] [Google Scholar]
  • 89.Zhang C, Kelly MJ, Ronnekleiv OK. 17β-Estradiol rapidly increases KATP activity in GnRH via a protein knase signaling pathway. Endocrinology. 2010;151:4477–84. doi: 10.1210/en.2010-0177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Brubaker KD, Gay CV. Depolarization of osteoclast plasma membrane potential by 17β-estradiol. J Bone Mineral Res. 1999;14:1861–6. doi: 10.1359/jbmr.1999.14.11.1861. [DOI] [PubMed] [Google Scholar]
  • 91.El Gebeily G, Fiset C. 4-Hydroxytamoxifen inhibits K+ currents in mouse ventricular myocytes. Eur J Pharmacol. 2010;629:96–103. doi: 10.1016/j.ejphar.2009.12.006. [DOI] [PubMed] [Google Scholar]
  • 92.Nabekura J, et al. Mechanism of the rapid effect of 17β-estradiol on medial amygdala neurons. Science. 1986;233:226–8. doi: 10.1126/science.3726531. [DOI] [PubMed] [Google Scholar]
  • 93.Okabe K, et al. Estrogen directly acts on osteoclasts via inhibition of inward rectifier K+ channels. Naunyn Schmiedebergs Arch Pharmacol. 2000;361:610–20. doi: 10.1007/s002100000243. [DOI] [PubMed] [Google Scholar]
  • 94.Ponce-Balbuena D, et al. Tamoxifen inhibits inward rectifier K+ 2.x family of inward rectifier channels by interfering with phosphatidylinositol 4,5-bisphosphate-channel interactions. J Pharmacol Exp Ther. 2009;331:563–73. doi: 10.1124/jpet.109.156075. [DOI] [PubMed] [Google Scholar]
  • 95.Ponce-Balbuena D, et al. Tamoxifen inhibits cardiac ATP-sensitive and acetylcholine-activated K+ currents in part by interfering with phosphatidylinositol 4,5-bisphosphate-channel interaction. J Pharmacol Sci. 2010;113:66–75. doi: 10.1254/jphs.10024fp. [DOI] [PubMed] [Google Scholar]
  • 96.Qiu J, et al. Rapid signaling of estrogen in hypothalamic neurons involves a novel G-protein-coupled estrogen receptor that activates protein kinase C. J Neurosci. 2003;23:9529–40. doi: 10.1523/JNEUROSCI.23-29-09529.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Smitherman KA, Sontheimer H. Inhibition of glial Na+ and K+ currents by tamoxifen. J Memb Biol. 2001;181:125–35. doi: 10.1007/s00232-001-0016-2. [DOI] [PubMed] [Google Scholar]
  • 98.Berger F, et al. Effects of 17b-estradiol on action potentials and ionic currents in male rat ventricular myocytes. Naunyn Schmiedebergs Arch Pharmacol. 1997;356:788–96. doi: 10.1007/pl00005119. [DOI] [PubMed] [Google Scholar]
  • 99.Kunz L, et al. Voltage-dependent K+ channel acts as sex steroid sensor in endocrine cells of the human ovary. J Cell Physiol. 2006;206:167–74. doi: 10.1002/jcp.20453. [DOI] [PubMed] [Google Scholar]
  • 100.Zhang ZH, Wang Q. Modulation of a cloned human A-type voltage-gated potassium channel hKv1.4) by the protein tyrosine kinase inhibitor genistein. Pflugers Arch. 2000;440:784–92. doi: 10.1007/s004240000360. [DOI] [PubMed] [Google Scholar]
  • 101.Kelly MJ, et al. Rapid effects of estrogen to modulate G protein-coupled receptors via activation of protein kinase A and protein kinase C pathways. Steroids. 1999;64:64–75. doi: 10.1016/s0039-128x(98)00095-6. [DOI] [PubMed] [Google Scholar]
  • 102.Kelly MJ, et al. Rapid effects of estrogen on G protein-coupled receptor activation of potassium channels in the central nervous system CNS) J Steroid Biochem Mol Biol. 2002;83:187–93. doi: 10.1016/s0960-0760(02)00249-2. [DOI] [PubMed] [Google Scholar]
  • 103.Lagrange AH, et al. Estrogen rapidly attenuates a GABAB response in hypothalamic neurons. Neuroendocrinology. 1996;64:114–23. doi: 10.1159/000127106. [DOI] [PubMed] [Google Scholar]
  • 104.Lagrange AH, Ronnekleiv OK, Kelly MJ. Modulation of G protein-coupled receptors by an estrogen receptor that activates protein kinase A. Mol Pharmacol. 1997;51:605–12. doi: 10.1124/mol.51.4.605. [DOI] [PubMed] [Google Scholar]
  • 105.Vanheiden S, Pott L, Kienitz MC. Voltage-dependent open-channel block of G protein-gated inward-rectifying K+) GIRK) current in rat atrial myocytes by tamoxifen. Naunyn Schmiedebergs Arch Pharmacol. 2012;385:1149–60. doi: 10.1007/s00210-012-0801-8. [DOI] [PubMed] [Google Scholar]
  • 106.Kurokawa J, et al. Acute effects of oestrogen on the guinea pig and human IKr channels and drug-induced prolongation of cardiac repolarization. J Physiol. 2008;586:2961–73. doi: 10.1113/jphysiol.2007.150367. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Liu H, et al. The selective estrogen receptor modulator raloxifene inhibits cardiac delayed rectifier potassium currents and voltage-gated sodium current without QTc interval prolongation. Pharmacol Res. 2010;62:384–90. doi: 10.1016/j.phrs.2010.07.008. [DOI] [PubMed] [Google Scholar]
  • 108.Moller C, Netzer R. Effects of estradiol on cardiac ion channel currents. Eur J Pharmacol. 2006;532:44–9. doi: 10.1016/j.ejphar.2006.01.006. [DOI] [PubMed] [Google Scholar]
  • 109.Thomas D, et al. Inhibition of cloned HERG potassium channels by the antiestrogen tamoxifen. Naunyn Schmiedebergs Archives of Pharmacology. 2003;368:41–8. doi: 10.1007/s00210-003-0766-8. [DOI] [PubMed] [Google Scholar]
  • 110.Betts V, Lapaix F, Harvey BJ. The colonic crypt potassium channel, KCNQ1, is a target of estrogen-activated PKA and PKC delta kinase. FASEB Journal. 2004;18 Abst. 846.13. [Google Scholar]
  • 111.O'Mahony F, et al. Female gender-specific inhibition of KCNQ1 channels and chloride secretion by 17b-estradiol in rat distal colonic crypts. J Biol Chem. 2007;282:24563–24573. doi: 10.1074/jbc.M611682200. [DOI] [PubMed] [Google Scholar]
  • 112.Lagrange AH, Ronnekleiv OK, Kelly MJ. Estradiol-17β and m-opioid peptides rapidly hyperpolarize GnRH neurons: a cellular mechanism of negative feedback? Endocrinology. 1995;136:2341–4. doi: 10.1210/endo.136.5.7720682. [DOI] [PubMed] [Google Scholar]
  • 113.Minami T, et al. 17 β-estradiol depolarization of hypothalamic neurons is mediated by cyclic AMP. Brain Res. 1990;519:301–7. doi: 10.1016/0006-8993(90)90092-p. [DOI] [PubMed] [Google Scholar]
  • 114.Rusko J, Li L, van Breemen C. 17β-Estradiol stimulation of endothelial K+ channels. Biochem Biophys Res Comm. 1995;214:367–72. doi: 10.1006/bbrc.1995.2297. [DOI] [PubMed] [Google Scholar]
  • 115.Tsang SY, et al. Contribution of K+ channels to relaxation induced by 17β-estradiol but not by progesterone in isolated rat mesenteric artery rings. J Cardiovascular Pha rmacol. 2003;41:4–13. doi: 10.1097/00005344-200301000-00002. [DOI] [PubMed] [Google Scholar]
  • 116.Waldegger S, et al. Inhibition of minK protein induced K+ channels in Xenopus oocytes by estrogens. N-S Arch Pharmacol. 1996;354:698–702. doi: 10.1007/BF00166894. [DOI] [PubMed] [Google Scholar]
  • 117.Wong M, Moss RL. Electrophysiological evidence for a rapid membrane action of the gonadal steroid, 17β-estradiol, on CA1 pyramidal neurons of the rat hippocampus. Brain Res. 1991;543:148–52. doi: 10.1016/0006-8993(91)91057-8. [DOI] [PubMed] [Google Scholar]
  • 118.Zhang L, et al. Direct binding of estradiol enhances Slack sequence like a calcium-activated potassium channel) channels?activity. Neuroscience. 2005;131:275–282. doi: 10.1016/j.neuroscience.2004.10.042. [DOI] [PubMed] [Google Scholar]
  • 119.Azenabor AA, Chaudhry AU. 17β-estradiol induces L-type Ca2+ channel activation and regulates redox function in macrophages. J Reprod Immunol. 2003;59:17–28. doi: 10.1016/s0165-0378(02)00084-0. [DOI] [PubMed] [Google Scholar]
  • 120.Hao J, et al. Ca2+ channel subunit α 1D promotes proliferation and migration of endometrial cancer cells mediated by 17β-estradiol via the G protein-coupled estrogen receptor. Faseb J. 2015;29:2883–93. doi: 10.1096/fj.14-265603. [DOI] [PubMed] [Google Scholar]
  • 121.Johnson BD, et al. Increased expression of the cardiac L-type calcium channel in estrogen receptor-deficient mice. J Gen Physiol. 1997;110:135–140. doi: 10.1085/jgp.110.2.135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Sarkar SN, et al. Estrogens directly potentiate neuronal L-type Ca2+ channels. Proc Natl Acad Sci U S A. 2008;105:15148–15153. doi: 10.1073/pnas.0802379105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Boulware MI, et al. Estradiol activates group I and II metabotropic glutamate receptor signaling, leading to opposing influences on cAMP response element-binding protein. J Neurosci. 2005;25:5066–5078. doi: 10.1523/JNEUROSCI.1427-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Cairrao E, et al. Non-genomic vasorelaxant effects of 17β-estradiol and progesterone in rat aorta are mediated by L-type Ca2+ current inhibition. Acta Pharmacol Sin. 2012;33:615–24. doi: 10.1038/aps.2012.4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Grohe C, et al. Modulation of hypertensive heart disease by estrogen. Steroids. 1996;61:201–204. doi: 10.1016/0039-128x(96)00014-1. [DOI] [PubMed] [Google Scholar]
  • 126.Kim YJ, et al. Nongenomic inhibition of catecholamine secretion by 17β-estradiol in PC12 cells. J Neurochem. 2000;74:2490–2496. doi: 10.1046/j.1471-4159.2000.0742490.x. [DOI] [PubMed] [Google Scholar]
  • 127.Kitazawa T, et al. Non-genomic mechanism of 17 β-oestradiol-induced inhibition of contraction in mammalian vascular smooth muscle. J Physiol. 1997;499:497–511. doi: 10.1113/jphysiol.1997.sp021944. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Kumar A, Foster TC. 17β-estradiol benzoate decreases the AHP amplitude in CA1 pyramidal neurons. J Neurophysiol. 2002;88:621–6. doi: 10.1152/jn.2002.88.2.621. [DOI] [PubMed] [Google Scholar]
  • 129.Kurata K, et al. Effect of beta-estradiol on voltage-gated Ca2+ channels in rat hippocampal neurons: a comparison with dehydroepiandrosterone. European Journal of Pharmacology. 2001;416:203–12. doi: 10.1016/s0014-2999(01)00880-9. [DOI] [PubMed] [Google Scholar]
  • 130.Liew R, et al. Raloxifene acutely suppresses ventricular myocyte contractility through inhibition of the L-type calcium current. Br J Pharmacol. 2004 doi: 10.1038/sj.bjp.0705736. Epub 2004 Mar 15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Liu B, et al. Effects of 17 beta-estradiol on early afterdepolarizations and L-type Ca2+ currents induced by endothelin-1 in guinea pig papillary muscles and ventricular myocytes. Methods & Findings in Experimental & Clinical Pharmacology. 1997;19:19–25. [PubMed] [Google Scholar]
  • 132.Liu H, Li K, Sperelakis N. Tyrosine kinase inhibitor, genistein, inhibits macroscopic L-type calcium current in rat portal vein smooth muscle cells. Can J Physiol Pharmacol. 1997;75:1058–62. doi: 10.1139/cjpp-75-9-1058. [DOI] [PubMed] [Google Scholar]
  • 133.Mermelstein PG, Becker JB, Surmeier DJ. Estradiol reduces calcium currents in rat neostriatal neurons via a membrane receptor. J Neurosci. 1996;16:595–604. doi: 10.1523/JNEUROSCI.16-02-00595.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Montano LM, et al. Relaxation of androgens on rat thoracic aorta: testosterone concentration dependent agonist/antagonist L-type Ca2+ channel activity, and 5beta-dihydrotestosterone restricted to L-type Ca2+ channel blockade. Endocrinology. 2008 doi: 10.1210/en.2007-1288. Epub 2008 Feb 14. [DOI] [PubMed] [Google Scholar]
  • 135.Nakajima T, et al. 17β-Estradiol inhibits the voltage-dependent L-type Ca2+ currents in aortic smooth muscle cells. Eur J Pharmacol. 1995;294:625–35. doi: 10.1016/0014-2999(95)00602-8. [DOI] [PubMed] [Google Scholar]
  • 136.Ruehlmann DO, et al. Environmental estrogenic pollutants induce acute vascular relaxation by inhibiting L-type Ca2+ channels in smooth muscle cells. Faseb J. 1998;12:613–9. doi: 10.1096/fasebj.12.7.613. [DOI] [PubMed] [Google Scholar]
  • 137.Sanchez JC, Lopez-Zapata DF, Pinzon OA. Effects of 17β-estradiol and IGF-1 on L-type voltage-activated and stretch-activated calcium currents in cultured rat cortical neurons. Neuro Endocrinol Lett. 2014;35:724–32. [PubMed] [Google Scholar]
  • 138.Song J, et al. Tamoxifen estrogen antagonist) inhibits voltage-gated calcium current and contractility in vascular smooth muscle from rats. Journal of Pharmacology & Experimental Therapeutics. 1996;277:1444–53. [PubMed] [Google Scholar]
  • 139.Ullrich ND, Koschak A, MacLeod KT. Oestrogen directly inhibits the cardiovascular L-type Ca2+ channel Cav1.2. Biochem Biophys Res Commun. 2007;361:522–7. doi: 10.1016/j.bbrc.2007.07.054. [DOI] [PubMed] [Google Scholar]
  • 140.Wu TW, et al. 17Beta-estradiol induced Ca2+ influx via L-type calcium channels activates the Src/ERK/cyclic-AMP response element binding protein signal pathway and BCL-2 expression in rat hippocampal neurons: a potential initiation mechanism for estrogen-induced neuroprotection. Neuroscience. 2005;135:59–72. doi: 10.1016/j.neuroscience.2004.12.027. [DOI] [PubMed] [Google Scholar]
  • 141.Zhang F, et al. 17 beta-Estradiol attenuates voltage-dependent Ca2+ currents in A7r5 vascular smooth muscle cell line. Am J Physiol. 1994;266:C975–80. doi: 10.1152/ajpcell.1994.266.4.C975. [DOI] [PubMed] [Google Scholar]
  • 142.Wang Q, et al. Effects of raloxifene on voltage-dependent T-type Ca2+ channels in mouse spermatogenic cells. Pharmacology. 2011;87:70–80. doi: 10.1159/000321726. [DOI] [PubMed] [Google Scholar]
  • 143.Zhao X, et al. Calcium flux in neuroblastoma cells is a coupling mechanism between non-genomic and genomic modes of estrogens. Neuroendocrinology. 2005;81:174–182. doi: 10.1159/000087000. [DOI] [PubMed] [Google Scholar]
  • 144.Huang Y, et al. Raloxifene acutely reduces glutamate-induced intracellular calcium increase in cultured rat cortical neurons via inhibition of high-voltage-activated calcium current. Neuroscience. 2007;147:334–41. doi: 10.1016/j.neuroscience.2007.04.037. [DOI] [PubMed] [Google Scholar]
  • 145.Muck AO, et al. Does melatonin affect calcium influx in human aortic smooth muscle cells and estradiol-mediated calcium antagonism? Journal of Pineal Research. 1996;20:145–7. doi: 10.1111/j.1600-079x.1996.tb00250.x. [DOI] [PubMed] [Google Scholar]
  • 146.Ogata R, et al. Oestradiol-induced relaxation of rabbit basilar artery by inhibition of voltage-dependent Ca channels through GTP-binding protein. Br J Pharmacol. 1996;117:351–359. doi: 10.1111/j.1476-5381.1996.tb15198.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Salas E, et al. Endothelium-independent relaxation by 17 α-estradiol of pig coronary arteries. Eur J Pharmacol. 1994;258:47–55. doi: 10.1016/0014-2999(94)90056-6. [DOI] [PubMed] [Google Scholar]
  • 148.Shan J, et al. Vascular effects of 17β-estradiol in male Sprague-Dawley rats. Am J Physiol. 1994;266:H967–73. doi: 10.1152/ajpheart.1994.266.3.H967. [DOI] [PubMed] [Google Scholar]
  • 149.Wang Q, et al. Effects of estradiol on high-voltage-activated Ca2+ channels in cultured rat cortical neurons. Endocr Res. 2014;39:44–9. doi: 10.3109/07435800.2013.799485. [DOI] [PubMed] [Google Scholar]
  • 150.Doolan CM, Condliffe SB, Harvey BJ. Rapid non-genomic activation of cytosolic cyclic AMP-dependent protein kinase activity and [Ca2+]i by 17β-oestradiol in female rat distal colon. Br J Pharmacol. 2000;129:1375–1386. doi: 10.1038/sj.bjp.0703193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Irnaten M, Blanchard-Gutton N, Harvey BJ. Rapid effects of 17β-estradiol on epithelial TRPV6 Ca2+ channel in human T84 colonic cells. Cell Calcium. 2008;44:441–452. doi: 10.1016/j.ceca.2008.02.007. [DOI] [PubMed] [Google Scholar]
  • 152.Jeng YJ, Kochukov M, Watson CS. Combinations of physiologic estrogens with xenoestrogens alter calcium and kinase responses, prolactin release, and membrane estrogen receptor trafficking in rat pituitary cells. Environ Health. 2010;9:9–61. doi: 10.1186/1476-069X-9-61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Morley P, et al. A new, nongenomic estrogen action: the rapid release of intracellular calcium. Endocrinology. 1992;131:1305–1312. doi: 10.1210/endo.131.3.1505465. [DOI] [PubMed] [Google Scholar]
  • 154.Alonso-Magdalena P, et al. Low doses of bisphenol A and diethylstilbestrol impair Ca2+ signals in pancreatic alpha-cells through a nonclassical membrane estrogen receptor within intact islets of Langerhans. Environmental health perspectives. 2005;113:969–77. doi: 10.1289/ehp.8002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Morales A, et al. Rapid modulatory effect of estradiol on acetylcholine-induced Ca2+ signal is mediated through cyclic-GMP cascade in LHRH-releasing GT1-7 cells. Eur J Neurosci. 2005;22:2207–15. doi: 10.1111/j.1460-9568.2005.04432.x. [DOI] [PubMed] [Google Scholar]
  • 156.Rossi AM, et al. Evidence on the operation of ATP-induced capacitative calcium entry in breast cancer cells and its blockade by 17β-estradiol. J Cellu Biochem. 2002;87:324–33. doi: 10.1002/jcb.10303. [DOI] [PubMed] [Google Scholar]
  • 157.Farkas I, et al. Estrogen receptor a and b differentially mediate C5aR agonist evoked Ca2+-influx in neurons through L-type voltage-gated Ca2+ channels. Neurochem Int. 2012;60:631–639. doi: 10.1016/j.neuint.2012.02.024. [DOI] [PubMed] [Google Scholar]
  • 158.Al-Majed HT, et al. Effect of 17β-estradiol on insulin secretion and cytosolic calcium in Min6 mouse insulinoma cells and human islets of Langerhans. Pancreas. 2005;30:307–13. doi: 10.1097/01.mpa.0000161886.17492.22. [DOI] [PubMed] [Google Scholar]
  • 159.Dayanithi G, Tapia-Arancibia L. Rise in intracellular calcium via a nongenomic effect of allopregnanolone in fetal rat hypothalamic neurons. Journal of Neuroscience. 1996;16:130–6. doi: 10.1523/JNEUROSCI.16-01-00130.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Picotto G, Massheimer V, Boland R. Acute stimulation of intestinal cell calcium influx induced by 17β-estradiol via the cAMP messenger system. Mol Cell Endocrinol. 1996;119:129–34. doi: 10.1016/0303-7207(96)03799-9. [DOI] [PubMed] [Google Scholar]
  • 161.Picotto G, Vazquez G, Boland R. 17β-Oestradiol increases intracellular Ca2+ concentration in rat enterocytes. Potential role of phospholipase C-dependent store-operated Ca2+ influx. Biochem J. 1999;339:71–7. [PMC free article] [PubMed] [Google Scholar]
  • 162.Pietras RJ, Szego CM. Endometrial cell calcium and oestrogen action. Nature. 1975;253:357–9. doi: 10.1038/253357a0. [DOI] [PubMed] [Google Scholar]
  • 163.Crews JK, Khalil RA. Gender-specific inhibition of Ca2+ entry mechanisms of arterial vasoconstriction by sex hormones. Clinical & Experimental Pharmacology & Physiology. 1999;26:707–15. doi: 10.1046/j.1440-1681.1999.03110.x. [DOI] [PubMed] [Google Scholar]
  • 164.Han SZ, et al. 17 beta-Estradiol inhibits Ca2+ influx and Ca2+ release induced by thromboxane A2 in porcine coronary artery. Circulation. 1995;91:2619–26. doi: 10.1161/01.cir.91.10.2619. [DOI] [PubMed] [Google Scholar]
  • 165.Blanchard-Gutton N, Irnaten M, Harvey BJP. Rapid effects of oestrogen targeting epithelial Ca2+channels in kidney and colon. FASEB Journal. 2005;19:A1183. [Google Scholar]
  • 166.Muchekehu RW, Harvey BJ. 17β-estradiol rapidly mobilizes intracellular calcium from ryanodine-receptor-gated stores via a PKC-PKA-Erk-dependent pathway in the human eccrine sweat gland cell line NCL-SG3. Cell Calcium. 2008;44:276–88. doi: 10.1016/j.ceca.2007.12.001. [DOI] [PubMed] [Google Scholar]
  • 167.Perret S, Dockery P, Harvey BJ. 17β-oestradiol stimulates capacitative Ca2+ entry in human endometrial cells. Mol Cell Endocrinol. 2001;176:77–84. doi: 10.1016/s0303-7207(01)00464-6. [DOI] [PubMed] [Google Scholar]
  • 168.Sheng JZ, et al. Estrogen and the Ca2+-mobilizing agonist ATP evoke acute NO synthesis via distinct pathways in an individual human vascular endothelium-derived cell. Am J Physiol Cell Physiol. 2008;294:26. doi: 10.1152/ajpcell.00561.2007. [DOI] [PubMed] [Google Scholar]
  • 169.Sheldon JH, Argentieri TM. Acute administration of 17β-estradiol inhibits calcium currents in isolated guinea pig detrusor myocytes. J Pharmacol Experimental Therapeut. 1995;274:723–9. [PubMed] [Google Scholar]
  • 170.Yamamoto T. Effects of estrogens on Ca channels in myometrial cells isolated from pregnant rats. Am J Physiol. 1995;268:C64–9. doi: 10.1152/ajpcell.1995.268.1.C64. [DOI] [PubMed] [Google Scholar]
  • 171.Fraser SP, et al. Estrogen and non-genomic upregulation of voltage-gated Na+ channel activity in MDA-MB-231 human breast cancer cells: role in adhesion. J Cell Physiol. 2010;224:527–39. doi: 10.1002/jcp.22154. [DOI] [PubMed] [Google Scholar]
  • 172.Barann M, et al. Inhibition by steroids of [14C]-guanidinium flux through the voltage-gated sodium channel and the cation channel of the 5-HT3 receptor of N1E-115 neuroblastoma cells. Naunyn Schmiedebergs Arch Pharmacol. 1999;360:234–41. doi: 10.1007/s002109900089. [DOI] [PubMed] [Google Scholar]
  • 173.Wang Q, et al. Inhibition of voltage-gated sodium channels by bisphenol A in mouse dorsal root ganglion neurons. Brain Research. 2011;1378:1–8. doi: 10.1016/j.brainres.2011.01.022. [DOI] [PubMed] [Google Scholar]
  • 174.Hardy SP, Valverde MA. Novel plasma membrane action of estrogen and antiestrogens revealed by their regulation of a large conductance chloride channel. Faseb J. 1994;8:760–5. doi: 10.1096/fasebj.8.10.8050676. [DOI] [PubMed] [Google Scholar]
  • 175.Li Z, et al. Estrogen modulates a large conductance chloride channel in cultured porcine aortic endothelial cells. J Cardiovasc Pharmacol. 2000;35:506–10. doi: 10.1097/00005344-200003000-00023. [DOI] [PubMed] [Google Scholar]
  • 176.Condliffe SB, Doolan CM, Harvey BJ. 17β-Oestradiol acutely regulates Cl-secretion in rat distal colonic epithelium. J Physiol. 2001;530:47–54. doi: 10.1111/j.1469-7793.2001.0047m.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Valverde MA, Hardy SP, Diaz M. Activation of Maxi Cl- channels by antiestrogens and phenothiazines in NIH3T3 fibroblasts. Steroids. 2002;67:439–45. doi: 10.1016/s0039-128x(01)00174-x. [DOI] [PubMed] [Google Scholar]
  • 178.Zhang JJ, et al. Tamoxifen blocks chloride channels: A possible mechanism for cataract formation. J Clinical Investigation. 1994;94:1690–1697. doi: 10.1172/JCI117514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Gu Q, Moss RL. 17β-Estradiol potentiates kainate-induced currents via activation of the cAMP cascade. J Neurosci. 1996;16:3620–3629. doi: 10.1523/JNEUROSCI.16-11-03620.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Gu Q, Moss RL. Novel mechanism for non-genomic action of 17β-oestradiol on kainate-induced currents in isolated rat CA1 hippocampal neurones. J Physiol. 1998;506:745–54. doi: 10.1111/j.1469-7793.1998.745bv.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Gu Q, Korach KS, Moss RL. Rapid action of 17β-estradiol on kainate-induced currents in hippocampal neurons lacking intracellular estrogen receptors. Endocrinology. 1999;140:660–6. doi: 10.1210/endo.140.2.6500. [DOI] [PubMed] [Google Scholar]
  • 182.Allen MC, et al. Inhibition of ligand-gated cation-selective channels by tamoxifen. Eur J Pharmacol. 1998;354:261–9. doi: 10.1016/s0014-2999(98)00454-3. [DOI] [PubMed] [Google Scholar]
  • 183.Liu PS, et al. Butyl benzyl phthalate blocks Ca2+ signaling and catecholamine secretion coupled with nicotinic acetylcholine receptors in bovine adrenal chromaffin cells. Neurotoxicology. 2003;24:97–105. doi: 10.1016/s0161-813x(02)00154-7. [DOI] [PubMed] [Google Scholar]
  • 184.Nakazawa K, Ohno Y. Modulation by estrogens and xenoestrogens of recombinant human neuronal nicotinic receptors. European Journal of Pharmacology. 2001;430:175–83. doi: 10.1016/s0014-2999(01)01389-9. [DOI] [PubMed] [Google Scholar]
  • 185.Foy MR, et al. 17β-Estradiol enhances NMDA receptor-mediated EPSPs and long-term potentiation. J Neurophysiol. 1999;81:925–9. doi: 10.1152/jn.1999.81.2.925. [DOI] [PubMed] [Google Scholar]
  • 186.Nilsen J, Chen S, Brinton RD. Dual action of estrogen on glutamate-induced calcium signaling: mechanisms requiring interaction between estrogen receptors and src/mitogen activated protein kinase pathway. Brain Res. 2002;930:216–34. doi: 10.1016/s0006-8993(02)02254-0. [DOI] [PubMed] [Google Scholar]
  • 187.Smith SS. Estrogen administration increases neuronal responses to excitatory amino acids as a long-term effect. Brain Research. 1989;503:354–357. doi: 10.1016/0006-8993(89)91691-0. [DOI] [PubMed] [Google Scholar]
  • 188.Cario-Toumaniantz C, et al. Non-genomic inhibition of human P2X7 purinoceptor by 17β-oestradiol. J Physiol. 1998;508:659–66. doi: 10.1111/j.1469-7793.1998.659bp.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Lu Y, et al. 17β-estradiol rapidly attenuates P2X3 receptor-mediated peripheral pain signal transduction via ERalpha and GPR30. Endocrinology. 2013;154:2421–33. doi: 10.1210/en.2012-2119. [DOI] [PubMed] [Google Scholar]
  • 190.Aoshima H, et al. Effects of bisphenol A and its derivatives on the response of GABAA) receptors expressed in Xenopus oocytes. Biosci Biotechnol Biochem. 2001;65:2070–7. doi: 10.1271/bbb.65.2070. [DOI] [PubMed] [Google Scholar]
  • 191.Choi IS, et al. Multiple effects of bisphenol A, an endocrine disrupter, on GABAA receptors in acutely dissociated rat CA3 pyramidal neurons. Neuroscience Research. 2007;59:8–17. doi: 10.1016/j.neures.2007.05.003. [DOI] [PubMed] [Google Scholar]
  • 192.Duncan RK. Tamoxifen alters gating of the BK alpha subunit and mediates enhanced interactions with the avian beta subunit. Biochem Pharmacol. 2005;70:47–58. doi: 10.1016/j.bcp.2005.03.026. [DOI] [PubMed] [Google Scholar]
  • 193.Qu ZW, et al. 17β-Estradiol enhances ASIC activity in primary sensory neurons to produce sex difference in acidosis-induced nociception. Endocrinology. 2015;156:4660–71. doi: 10.1210/en.2015-1557. [DOI] [PubMed] [Google Scholar]

RESOURCES