ArticlePDF AvailableLiterature Review

Potassium Channels in Vascular Smooth Muscle: A Pathophysiological and Pharmacological Perspective

Authors:
  • Gulhane Faculty of Medicine
  • Health Sciences University Gülhane Faculty of Medicine

Abstract

Potassium (K⁺) ion channel activity is an important determinant of vascular tone by regulating cell membrane potential (MP). Activation of K⁺ channels leads to membrane hyperpolarization and subsequently vasodilatation, while inhibition of the channels causes membrane depolarization and then vasoconstriction. So far five distinct types of K⁺ channels have been identified in vascular smooth muscle cells (VSMCs); Ca⁺²‐activated K⁺ channels (BKCa), voltage‐dependent K⁺ channels (KV), ATP‐sensitive K⁺ channels (KATP), inward rectifier K⁺ channels (Kir), and tandem‐two pore K⁺ channels (K2P). The activity and expression of vascular K⁺ channels are changed during major vascular diseases such as hypertension, pulmonary hypertension, hypercholesterolemia, atherosclerosis, and diabetes mellitus. The defective function of K⁺ channels is commonly associated with impaired vascular responses and is likely to become as a result of changes in K⁺ channels during vascular diseases. Increased K⁺ channel function and expression may also help to compensate for increased abnormal vascular tone. There are many pharmacological and genotypic studies which were carried out on the subtypes of K⁺ channels expressed in variable amounts in different vascular beds. Modulation of K⁺ channel activity by molecular approaches and selective drug development may be a novel treatment modality for vascular dysfunction in the future. This review presents the basic properties, physiological functions, pathophysiological and pharmacological roles of the five major classes of K⁺ channels that have been determined in VSMCs. This article is protected by copyright. All rights reserved.
REVIEW
ARTICLE
Potassium channels in vascular smooth
muscle: a pathophysiological and
pharmacological perspective
Muhammed Fatih Dogan
a
, Oguzhan Yildiz
b
*, Seyfullah Oktay
Arslan
a
, Kemal Gokhan Ulusoy
b
a
Department of Pharmacology, Ankara Yildirim Beyazit University, Bilkent, Ankara 06010, Turkey
b
Department of Pharmacology, Gulhane Faculty of Medicine, University of Health Sciences, Etlik, Ankara 06170,
Turkey
Keywords
hypertension,
potassium channels,
vascular diseases,
vascular smooth muscle
cells
Received 14 August 2018;
revised 28 February 2019;
accepted 7 March 2019
*Correspondence and reprints:
oguzhany01@gmail.com
ABSTRACT
Potassium (K
+
) ion channel activity is an important determinant of vascular tone
by regulating cell membrane potential (MP). Activation of K
+
channels leads to
membrane hyperpolarization and subsequently vasodilatation, while inhibition of
the channels causes membrane depolarization and then vasoconstriction. So far
five distinct types of K
+
channels have been identified in vascular smooth muscle
cells (VSMCs): Ca
+2
-activated K
+
channels (BK
Ca
), voltage-dependent K
+
channels
(K
V
), ATP-sensitive K
+
channels (K
ATP
), inward rectifier K
+
channels (K
ir
), and tan-
dem two-pore K
+
channels (K
2
P). The activity and expression of vascular K
+
chan-
nels are changed during major vascular diseases such as hypertension, pulmonary
hypertension, hypercholesterolemia, atherosclerosis, and diabetes mellitus. The
defective function of K
+
channels is commonly associated with impaired vascular
responses and is likely to become as a result of changes in K
+
channels during vas-
cular diseases. Increased K
+
channel function and expression may also help to
compensate for increased abnormal vascular tone. There are many pharmacologi-
cal and genotypic studies which were carried out on the subtypes of K
+
channels
expressed in variable amounts in different vascular beds. Modulation of K
+
channel
activity by molecular approaches and selective drug development may be a novel
treatment modality for vascular dysfunction in the future. This review presents the
basic properties, physiological functions, pathophysiological, and pharmacological
roles of the five major classes of K
+
channels that have been determined in VSMCs.
INTRODUCTION
Vascular smooth muscle cells (VSMCs) are contractile
and relaxative cells of all the vessels in the body. They
determine the diameter of the arteries and veins and
regulate tissue perfusion and venous drainage. Periph-
eral vascular resistance and blood pressure are deter-
mined by the vascular smooth muscle (VSM) tone of
the arteries and arterioles [1,2]. K
+
channels in VSMCs
have important roles in regulation of vascular tone,
but their functions have not yet been fully understood
[3,4]. The current knowledge in the literature clearly
shows that changing the equilibrium between internal
and external K
+
currents in VSMCs has a decisive effect
on MP, Ca
+2
influx, and VSM contraction [5,6]. Thus,
K
+
channels play a crucial role in the regulation of
vasomotor function in pathological conditions and they
are the potential targets of many new drugs for several
cardiovascular diseases [3,7].
The depolarization of cell MP and the continuously
elevated intracellular calcium concentrations ([Ca
+2
]
i
)
are both physiologically and pathologically important
ª2019 Soci
et
e Franc
ßaise de Pharmacologie et de Th
erapeutique
Fundamental & Clinical Pharmacology 1
doi: 10.1111/fcp.12461
factors in the induction of cell proliferation as well as
in the contraction of VSMCs [8,9]. Because of the repo-
larizing effects of VSMCs, changes in the expression or
activity of the K
+
channels often translate into an
abnormal resting membrane potential (MP) associated
with abnormal vascular tone in arterial VSMCs.
Changes in the structure and function of the K
+
chan-
nels contribute to the pathophysiological conditions of
some vascular diseases, including hypertension, pul-
monary hypertension, hypercholesterolemia, and
atherosclerosis [10,11]. Increased vascular tone in
these diseases is often associated with depolarizing
changes in the resting MP of VSMCs, which may cause
abnormal vascular tone and hypersensitivity to vaso-
constrictor substances. Because impaired K
+
channel
activity is associated with vascular diseases, K
+
chan-
nels represent potential targets for the design of novel
therapeutic drugs that regulate abnormal vascular
activity [8,12].
Recently many pharmacological and genotypical
studies have been carried out on the subtypes of K
+
channels in VSMCs. Hence, novel specific drugs modu-
lating these subtypes are being developed from day to
day. In the near future, targeting the K
+
channels
modulation as a promising alternative to treat vascular
diseases may be considered a novel pharmacologic
approach. In this review, the basic pharmacological,
physiological, and pathophysiological properties of K
+
channels in VSMCs are presented.
BASIC PROPERTIES OF POTASSIUM
CHANNELS
Extensive evidence indicates that K
+
conductance is
mainly responsible for modulating cellular excitability
in VSMCs [2]. K
+
channels are a largest family of inte-
gral membrane proteins that form aqueous pores in cell
membranes through which K
+
can flow [13,14]. Func-
tionally, five different types of K
+
channels are
expressed in the vasculature: big conductance Ca
+2
-
activated (BK
Ca
), voltage-activate (K
V
), ATP-sensitive
(K
ATP
), inward rectifier (K
ir
), and two-pore domain
(K
2P
)K
+
channels [4,15,16]. Considering their struc-
tural electrophysiological and pharmacological proper-
ties, they are also classified into three main groups
according to the transmembrane (TM) spanning
domain number of each of the a-subunits forming the
channel: 2 TM, 4 TM, and 6 TM. The first 2 TM
groups consist of K
+
channels with two TM domains in
each of the a-subunits, also known as K
ir
and K
ATP
channels. Those with 6 TM are the largest K
+
channel
class and are divided into two types: K
Ca
and K
V
[16,17]. Unlike the 2 TM and 6 TM families, 4 TM
channels have two rather one pore-forming domains
per subunit [6].
The different subunits (aand b) of the K
+
channels
are encoded by different genes. The a-subunit consti-
tutes the pore structure that comprises the gating
apparatus and selective filter and interacts with auxil-
iary subunits that modulate the effects from different
signaling pathways. The b-subunit accessory part,
which increases sensitivity to Ca
+2
, modulates channel
activation and inhibition and reduces sensitivity to K
+
channels inhibitors, such as iberiotoxin (IbTx) and
tetraethylammonium (TEA) [1820].
K
+
channels activity, an important determinant of
MP in VSMCs, is a major regulator of vessel diameter
and vascular tone. The opening of these channels
induces MP hyperpolarization with K
+
efflux, which
inhibits Ca
+2
entry by the closure of L-type voltage-
activated calcium channels (LVCC), and reduced Ca
+2
entry results in vasodilation [2123]. Closure of K
+
channels leads to MP depolarization, opening of LVCC,
increased [Ca
+2
]
i
, eventually led vasoconstriction.
Therefore, as an important regulator of MP, K
+
chan-
nel activity is an essential determinant of vessel diame-
ter and vascular tone [24,25]. Effects of drugs,
endogenous, or exogenous substances on K
+
channels
in VSMCs are summarized in Table I.
Calcium-activated K
+
channels (K
Ca
)
These channels are expressed virtually in all excitable
tissues and can be classified as three subfamilies: large-
conductance Ca
+2
-activated K
+
channels (BK
Ca
), inter-
mediate-conductance K
+
channels (IK
Ca
), and small-
conductance K
+
channels (SK
Ca
) [51,52]. In general,
vascular endothelial cells express SK
Ca
and IK
Ca
chan-
nels. The single-channel conductivity of BK
Ca
is
between 200 and 250 pS and is significantly different
from the values of SK
Ca
(510 pS) and IK
Ca
(20
40 pS). In the VSM, the most commonly described cur-
rent is a large conductivity (>200 pS). BK
Ca
is known
by pharmacological properties such as apamin (SK
Ca
inhibitor) and clotrimazole (IK
Ca
inhibitor) [53] insensi-
tivity with IbTx high sensitivity, which does not block
IK
Ca
or SK
Ca
[20,54,55].
BK
Ca
, such as K
V
, is voltage-dependent and mem-
brane depolarization activates these channels. BK
Ca
is
specifically activated by Ca
+2
release from the sar-
coplasmic reticulum into the VSMCs [5659].
ª2019 Soci
et
e Franc
ßaise de Pharmacologie et de Th
erapeutique
Fundamental & Clinical Pharmacology
2M.F. Dogan et al.
Ryanodine-sensitive Ca
+2
-release channels (ryanodine
receptors [RyRs]) have a very important function in
controlling arterial tone and diameter through regula-
tion of BK
Ca
. RyRs in sarcoplasmic reticulum and BK
Ca
operate as a functional unit to regulate [Ca
+2
]
i
and
arterial tone through changes in MP and Ca
+2
influx.
The RyRs found in the sarcoplasmic reticulum and
BK
Ca
act as a functional unit regulating arterial tone
and [Ca
+2
]
i
with MP changes [6062]. Hence, BK
Ca
is
target for a number of pharmacological agents that
may have a role in the prevention or reversal of
VSMCs depolarization, contraction, and maintenance of
the resting tone [63,64].
BK
Ca
has been described in almost all VSMCs types
studied so far and play a crucial role in the regulation
of the vascular tone [11,23]. The activation of BK
Ca
causes in a marked K
+
efflux leading to hyperpolariza-
tion in the VSMCs, resulting in LVCC closure and
vasodilation [6567]. Each subunit provides different
pharmacological properties to BK
Ca
, which consists of
the pore-forming asubunit and the accessory bsub-
unit that modulate a-subunit Ca
+2
sensitivity and
channel activity [68,69]. Four b-subunits have been
determined in VSMCs, but the predominant is the b1-
subunit, which enhances the Ca
+2
and voltage sensitiv-
ity of BK
Ca
[16,70]. Many studies have shown
increased myogenic tone and contractility in the
absence or less expression of the b1 subunit. Moreover,
Yang et al. [71] showed that higher ratio of b1 sub-
unit: a-subunit in cerebral VSMCs related to that in
skeletal muscle may lead to higher Ca
+2
sensitivity of
BK
Ca
current in cerebral arteries. It is well known that
BK
Ca
is primarily activated either by an elevation of
the [Ca
+2
]i or by membrane depolarization and main-
tain MP and vascular myogenic tone through negative
feedback modulation [22,7274]. Hence, BK
Ca
affects
as a relaxing negative feedback mechanism after ago-
nist-induced membrane depolarization and extracellular
Table I Effects of drug, endogenous or exogenous substances on K
+
channels in VSMCs.
Substance K
+
Channel Species Vascular bed Conclusion References
Taurine BK
Ca
Human Internal mammary artery Channel activation [26]
Taurine BK
Ca
Human Radial artery Channel activation [27]
Testosteron BK
Ca
Human Internal spermatic vein Channel activation [28]
Testosteron BK
Ca
Human Internal mammary artery Channel activation [29]
Propofol BK
Ca
Human Umbilical artery and vein Channel activation [30]
Propofol BK
Ca
Human Internal mammary artery Channel activation [31]
Hydrochlorothiazide BK
Ca
Human Umbilical artery VSMCs Channel activation [32]
Bupivacaine BK
Ca
Human Umbilical artery VSMCs Channel activation [33]
Epicatechin BK
Ca
,K
ATP
,K
V
Human Internal mammary artery Channel activation [34]
Levosimendan BK
Ca
,K
ATP
Human Umbilical artery Channel activation [35]
Propofol BK
Ca
Human Omental artery and vein Channel activation [36]
ACPA BK
Ca
Rat Mesenteric artery Channel activation [37]
Glabridin BK
Ca
Rat Mesenteric artery Channel activation [38]
Taurine BK
Ca
Rat Mesenteric artery
Renal artery
Channel activation [39]
Propofol BK
Ca
Rat Mesenteric artery Channel activation [40]
Propofol BK
Ca
K
ATP
Rat Mesenteric artery and vein Channel activation [41]
Taurine BK
Ca
K
ATP
Rat Aorta Channel activation [42]
Procyanidin B2 BK
Ca
K
ATP
Human Internal mammary artery Channel activation [43]
YM155 K
V
1.5, K
V
2.1 Human PASMCs Channel activation [44]
Levosimendan K
ATP
Human Internal mammary artery Channel activation [45]
Levosimendan K
ATP
Human PASMCs Channel activation [46]
Propofol K
ATP
Rat Aorta Channel activation [47]
High Salt Diet K
ATP
Rat Mesenteric artery Channel disfunction [21]
Thiopental, Propofol K
ATP
Rat Rat Aorta Decrease in K
ATP
-mediated relaxation [48]
Thiopental, Propofol K
ATP
Dog Pulmonary Vein Decrease in K
ATP
-mediated relaxation [49]
Nitric Oxide K
ATP
Rabbit Mesenteric artery Channel activation [50]
ª2019 Soci
et
e Franc
ßaise de Pharmacologie et de Th
erapeutique
Fundamental & Clinical Pharmacology
Pharmacological perspective of potassium channels 3
Ca
+2
entry in most isolated arteries obtained from ani-
mals and humans [75,76]. Pharmacologically, external
TEA, charybdotoxin, IbTx, ethylene glycol tetraacetic
acid, and Ni
+2
inhibit channel activity [7779]. IbTX
(10100 nM) [80,81] is a potent inhibitor of BK
Ca
that
selectively binds to a specific region on the outside of
the channel and blocks the flow of ions [82,83].
BK
Ca
activation relaxes VSM, especially in the con-
duit arteries. Several studies demonstrated that testos-
terone and taurine-induced vasodilation may occur via
activation of BK
Ca
in the human radial artery and the
internal mammary artery [26,27,29]. It has been
shown that the vasodilator effect of propofol on the
human umbilical vessels [30] and the internal mam-
mary artery [31] is related to BK
Ca
. Propofol also
relaxed the mesenteric arterioles via BK
Ca
activation in
rat [40]. Levosimendan-induced relaxation in the
umbilical artery has been reported to occur due to
BK
Ca
and K
V
activation [35]. However, inhibition of
BK
Ca
induces vasoconstriction and Mart
ın et al. [33]
demonstrated using the patch-clamp technique that
bupivacaine, a local anesthetic drug, inhibits BK
Ca
cur-
rents in the umbilical artery. BK
Ca
is the target of both
vasoconstrictors and vasodilators [84,85]. Alterations
in BK
Ca
activity might also initiate or aggravate patho-
physiological states such as vasospasm and ischemia
[86]. It was reported that cytoplasmic ATP inhibits
BK
Ca
, and suggested that these channels may underlie
hypoxia-induced vasodilation [87]. The mechanisms by
which BK
Ca
contributes to vascular dysfunction are still
unclear and further investigation is needed to deter-
mine the role of BK
Ca
in vascular diseases [73,88].
Voltage-dependent K
+
channels (K
V
)
K
+
channels have multiple functions in the VSMCs and
K
V
are postulated to be a major determinant of vascu-
lar tone [7,89]. Broad K
V
expression has been identified
in vascular system. In response to the depolarization of
the MP, K
V
are activated and K
+
efflux occurs, result-
ing in repolarization. The depolarization of VSMCs
leads to Ca
+2
flow into the cells from LVCC and con-
tractile mechanisms are activated. This shows that K
V
maintain the resting vascular tone by limiting mem-
brane depolarization [9092].
K
V
have the widest and most diverse family of 12
subfamilies (K
V
1K
V
12) among the K
+
channels and
consist of four a-subunits and b-subunits. The asub-
units, which forms the K
+
-selective pore, contain six
TM domains (S1S6) and P-loop between the S5 and
S6 domains. The S1S4 region composes the voltage-
sensing domain [55,93,94]. Calcium influx through
LVCC associated with membrane depolarization can
also inhibit K
V
channels, and this may be formed sec-
ondary to [Ca
+2
]
i
release by RyRs. 4-aminopyridine (4-
AP), which is a specific inhibitor, and TEA have an
inhibitory effect against K
V
[29,93,95].
In VSMCs, K
V
have five major subfamilies as K
V
1.0,
K
V
2.0, K
V
3.0, K
V
4.0, and K
V
7; however, the K
V
1.2
and K
V
1.5 are predominantly expressed [12,22,24,96].
The relative abundance of K
V
in rat mesenteric artery
is higher than that in rat tail artery [24,96]. Studies
indicate that K
V
1.3 have positive effects in the prolifer-
ation and cell migration, which can result in neointi-
mal hyperplasia or vascular remodeling. Margatoxin is
a potent, highly selective blocker of human K
V
1.3 and
has no effect on K
Ca
[94,97]. It has been demonstrated
that K
V
current in rat small mesenteric artery myo-
cytes consists of two major parts, K
V
1.21.5b1.2
and K
V
2.19.3, along with less functional K
V
7.4 and
K
V
7.5, which were encoded by the KCNQ4 and
KCNQ5, and play a key functional role in regulating
vascular tone [91]. Initially, K
V
7(K
V
7.1K
V
7.5) identi-
fied in murine portal vein myocytes, aorta, pulmonary,
basilar, cerebral, renal, and mesenteric arteries from
rats, mice, and humans [9,85,98100]. Hence, K
V
7
mostly expressed in VSMCs may be potential of target-
ing to treat vascular diseases such as hypertension and
pulmonary hypertension [94].
ATP-sensitive K
+
channels (K
ATP
channels)
K
ATP
are the important class of ion channels with
expression in many tissues, especially cardiac and
VSM. These channels function in a variety of physio-
logical processes, such as protection of cardiac cells
against ischemic injuries and regulation of vascular
tone [34,87,101]. The density of K
ATP
is relatively
lower in VSM than cardiac myocytes, and the different
expression of K
ATP
subtypes in vascular beds consti-
tutes variable biophysical and the pharmacological
properties [102]. It has been well known that K
ATP
can
directly affect vascular tone and blood flow as a result
of metabolic changes in various tissues and mediate
the effects of various endogenous and pharmacological
vasodilators [2,103].
K
ATP
in VSMCs are insensitive to ATP, and they are
activated by nucleoside diphosphates and inhibited by
glibenclamide (GLI). K
ATP
inhibited by intracellular ATP
were first identified in cardiac myocytes by Noma [104]
and have subsequently been found in the pancreatic b
cells, neurones, skeletal muscle, and vascular and non
ª2019 Soci
et
e Franc
ßaise de Pharmacologie et de Th
erapeutique
Fundamental & Clinical Pharmacology
4M.F. Dogan et al.
VSMs [12,105]. The composition of the K
ATP
channel
structure is clear, made up of four pore-forming K
ir
6 sub-
units (K
ir
6.1 or K
ir
6.2) and four regulatory sulfonylurea
(SUR) receptor subunits. While K
ATP
pore-forming sub-
units were encoded by the KCNJ8 (K
ir
6.1) and KCNJ11
(K
ir
6.2) genes, regulatory subunits of K
ATP
were encoded
by the ABCC8 (SUR1) and ABCC9 (SUR2), which are two
SUR genes. It is known that K
ATP
in VSM is formed of the
K
ir
6.1 and SUR2B subunits [101,106].It have shown
that K
ir
6.1 subunit forms pore and has an crucial role in
the regulation of vascular tone. The overactivity of K
ATP
can cause to decreased vascular tone and blood pressure
[101,107]. K
ir
6.1/SUR2B represent the predominant K
ATP
in VSM, but other subtypes are expressed in specific vascular
beds separately or in combination with K
ir
6.1/SUR2B
[55,102]. K
ATP
activation by drugs and neuropeptides may
be a common mechanism of membrane hyperpolarization
and vasodilation. The opening of K
ATP
results in membrane
hyperpolarization, decreased [Ca
+2
]
i
, and vasodilation
[106,108,109]. K
ATP
have been demonstrated to play a
substantial role in vascular tone regulation by using tradi-
tional pharmacological approaches, including the applica-
tion of K
ATP
openers and K
ATP
blockers. These channels,
which constitute an important component of physiologi-
cal regulation as well as metabolic sensitivity, are opened
vasodilators and are closed by vasoconstrictors. K
ATP
is
opened by some compounds such as lemakalim, minoxi-
dil, cromakalim, pinacidil, and diazoxide. In the treat-
ment of myocardial ischemia, glaucoma, bronchial
asthma, hypertension, and male baldness, K
ATP
openers
exhibit a broad therapeutic effect [110112].
K
ATP
are specifically blocked by SUR derivates such as
GLI. Since GLI-induced vasoconstriction was due to a
blockade of potassium efflux from the smooth muscle
cells, this suggests that under baseline conditions a potas-
sium efflux was responsible for a certain degree of vasore-
laxation [4,97,113]. K
ATP
inhibitor GLI (10 lM)
significantly inhibited both testosterone-induced relax-
ation in internal spermatic vein and levosimendan-
induced relaxation in internal mammary artery [28,45].
In addition, the vasodilation induced by levosimendan
may partially be due to a lowering of [Ca
+2
]
i
, and opening
of K
ATP
[114]. Conversely, Clapp et al. [78] demonstrated
that in guinea-pig aorta, BK
Ca
but not K
ATP
, play a major
role in vasorelaxation induced by the prostanoid agonists,
iloprost and cicaprost.
Inward rectifier K
+
channels (K
ir
)
K
ir
are present in some arterial smooth muscle cells
and in various excitable and nonexcitable cells. The
name of this channel comes from the fact that K
+
ion
current into the cell during changes in MP are larger
than the outward currents. K
ir
are more active at neg-
ative MP and therefore open for regulation the MP in
arteries in the absence of extrinsic depolarizing influ-
ences such as pressure or vasoconstrictors. In contrast
to K
ir
,K
V
and BK
Ca
are inactivated by membrane
hyperpolarization while being activated by membrane
depolarization [2,115].
K
ir
comprise of tetramers where each a-subunit has
two TM domains. The elevation of extracellular K
+
concentration also activates K
ir
and thereby causes
membrane hyperpolarization toward the K
+
equilib-
rium potential [55,77]. Of the large gene family of K
ir
subfamilies (K
ir
1.x to K
ir
7.x), the most relevant K
ir
expressed in the vasculature belong to the K
ir
2.x
(strong inward rectifier), and K
ir
6.x (weakly inward
rectifier) subfamilies. In VSMCs, K
ir
2.1 expression pre-
dominates, particularly in mice cerebral and coronary
arteries, and K
ir
2.1, K
ir
4.1, K
ir
6.x expression predomi-
nates mice mesenteric arteries [4,115,116]. Gollasch
et al. indicated that K
ir
are not functional in VSMCs of
human large epicardial coronary arteries [23].
K
ir
are specifically inhibited by Ba
+2
and Cs
+
,
whereas some of the other K
+
channel inhibitors such
as 4-AP or TEA have little effect on K
ir
. In small-resis-
tance arteries, such as the brain and coronary arteries,
K
ir
is activated by an increase of 15 mMin the extra-
cellular concentration [108,117,118].
Two-pore domain K
+
channels (K
2P
)
K
2P
are the smallest of the K
+
channels and are con-
structed by four TM domains and two pore structures.
They are considered as responsible for background leak
K
+
channels and are open at resting MP therefore
involved in controlling and stabilizing MP [88,119].
Structurally distinct from other vascular K
+
channels,
two pore-forming P loops (P1P2) arrange in tandem,
one between the first and second TM domain and the
other between the third and fourth TM domain. TWIK-
2 (Tandem of P-domains in a weakly inward rectifying
K
+
channel, K
2P
6.1), TREK-1 (TWIK-related K
+
chan-
nel), and TASK-1 (TWIK-related acid-sensitive K
+
channel, K
2P
3.1) from the 15 known members of the
K
2P
family show the largest expression levels for K
2P
in
VSMCs [6,16,120]. Unlike most other K
2P
, TWIK-2 is
highly expressed in all blood vessels studied to date
including resistance sized vessels. Thus, K
2P
6.1 is a
prime candidate for physiological regulation in the vas-
cular system and also regulates systemic blood pressure
ª2019 Soci
et
e Franc
ßaise de Pharmacologie et de Th
erapeutique
Fundamental & Clinical Pharmacology
Pharmacological perspective of potassium channels 5
[16,121]. In addition, TASK-1 channels play a physio-
logical role in regulating basal MP and tone in rat
mesenteric, pulmonary, and rabbit pulmonary arteries.
The contribution of these channels to the regulation of
vascular tone and blood pressure is not fully under-
stood and further research is needed [6,122,123].
The physiological and pathological functions of these
channels are investigated by using animal models
[120,124126]. K
2P
are very weakly sensitive or
insensitive to the classical K
+
channel blockers, such as
TEA, Ba
+2
,Cs
+
, and 4-AP. These channels are sensitive
to pH and are inhibited by acidification of the extracel-
lular fluid [127,128].
As mentioned above, there are currently several
members of K
+
channels and numerous substances
effect their functions. The list of known external block-
ers and openers of K
+
channels in arterial smooth mus-
cle are given in Tables II and III, respectively.
PATHOPHYSIOLOGICAL ROLES OF
POTASSIUM CHANNELS
The activity and expression of vascular K
+
channels are
changed during major vascular diseases such as
Table II External blockers of K
+
channels in arterial smooth muscle.
K
+
channel Blocker Concentration References
K
V
GLY %50 inhibition, 0.21.1 mM[2]
4-AP 1 mM[29]
TEA
+
510 mM[81]
Ba
+2
%50 inhibition >1mM[2]
K
V
1.3 Margatoxin %50 inhibition, 10 nM[22]
Charybdotoxin 0.1 lM[129]
K
V
1.5 Diphenyl phosphine oxide-1 %50 inhibition, 0.31 lM[130]
Kv7 Linopirdine 10 lM[85,99,131]
XE991 10 lM
K
V
7.1 HMR1556 10 lM[100]
K
Ca
TEA %50 inhibition, 0.2 mM[2]
4-AP %50 inhibition >5m
M[132]
Ba
+2
Little effect at 10 mM[2]
BK
Ca
IbTx 0.1100 nM[76,133]
Charybdotoxin 10 nM[100]
Paxilline 1 lM[54,72]
K
ir
Ba
+2
100 lM[134]
4-AP 10% inhibition at 1 mM[2]
TASK-1 Anandamide 10 lM[123]
Zn
+2
100 lM[123,128]
K
ATP
GLY 10 lM[134]
4-AP %50 inhibition, 1 mM[2]
Table III K
+
channel openers in VSMCs.
K
+
Channel Openers References
BK
Ca
NS1619 [83]
NS1608 [20]
Dehydroepiandrosterone [135]
Dehydrosoyasaponin-I [136]
Resveratrol [20]
MaxiKdiol [137]
L-735,334 [137]
NS 004, NS 169 [118]
K
ATP
Nicorandil [137]
Cromakalim [21]
Pinacidil [115]
Minoxidil sulfate [2]
Aprikalim [5]
Diazoxide [112]
PACAP-27 [23]
Lemakalim [87]
K
V
1.1 Acetazolamide [18]
Carbamazepine [18]
K
V
7 Retigabine [138]
Flupirtine [7]
K
ir
Mg
+2
[117]
TASK-1 Halothane [123]
ONO-RS-082 [139]
ª2019 Soci
et
e Franc
ßaise de Pharmacologie et de Th
erapeutique
Fundamental & Clinical Pharmacology
6M.F. Dogan et al.
hypertension, pulmonary hypertension, hypercholes-
terolemia, atherosclerosis, and diabetes mellitus.
Changes in K
+
channels in vascular diseases and differ-
ences among species are summarized in Table IV.
Hypertension
Cardiovascular diseases are the major cause of increas-
ing morbidity in modern societies, and their socio-eco-
nomic consequences are tremendous. Hypertension,
which is caused by diabetes, renal disease, or
unhealthy life style and poor nutrition, is one of, if not,
the major risk factors for myocardial infarction and
stroke [136,164]. Hypertension is generally associated
with increased peripheral resistance in patients with
hypertension and in animal models of hypertension due
in part to enhanced contractile function of VSM
[12,144]. Chronic hypertension is accompanied by a
continuous increase in arterial tone resulting from
coronary artery dysfunction. The increase in the arte-
rial tone is mainly related to depolarization, which may
be due to dysfunction of ion channels responsible for
hyperpolarization of VSMCs [74,165]. The VSM has a
great significance in relation to the pathogenesis of
hypertension [166]. The underlying causes of these
complex pathophysiological processes are probably
VSM remodeling [9]. Abnormal increased vessel tone is
a pathogenic mechanism of hypertension [140]. Both
depolarization and action potential formation are
related to Ca
+2
ion entry into the cell. Increased
[Ca
+2
]
i
in arterial VSMCs during hypertension play a
crucial role in abnormal contraction and phenotypic
changes [167169].
K
+
conductance plays a pivotal role in the regula-
tion of contractile function in arterial VSM. Disorders
in the functions or activities of the K
+
channels may
be associated with the pathophysiological mechanism
of hypertension leading to vasoconstriction
[73,89,118]. K
+
currents in VSMCs are one of the
most effective factors in preserving membrane stability,
they can suppress the membrane excitability, so less
K
+
current may occur in the hypertension disease
than in the normotensive state [59]. The loss of K
+
efflux causing membrane depolarization results in high
blood pressure in vivo and in vitro in spontaneously
hypertensive rat (SHR) mesenteric arteries [115,169].
It has been shown that humans and animals have an
increase in resting vascular tone in hypertension dis-
ease and that physiological stimulations lead to
increased contractile responses compared to the nor-
motensive control group [77,89].
Alterations of BK
Ca
in VSMCs have been documented
in both physiological (aging, pregnancy) and patho-
physiological conditions (diabetes mellitus) [34,170].
Hyperactivity of the arteries with age progression is the
changes that occur as a result of abnormal expression
and activity of BK
Ca
[141]. Blocking BK
Ca
or RyRs can
cause membrane depolarization, an elevation of
[Ca
+2
]
i
, and vasoconstriction [171]. Although the acti-
vation of the BK
Ca
operates as a negative feedback
mechanism to regulate the level of resting tone [66],
this activation is not enough to abolish the myogenic
tone in the SHRs [52]. Altered activity of the BK
Ca
may also play a role in the increased responsiveness to
TEA and Ba
+2
in SHRs [148]. During the progression
of hypertension, increased expression of BK
Ca
may
occur in order to maintain the resting vascular diame-
ter of arterioles. BK
Ca
current and expression are
increased in the cerebral arteries and aorta of SHRs,
but these increases were not associated with a detect-
able change in mRNA level, suggesting that the
increased expression of BK
Ca
may be mediated primar-
ily by post-transcriptional events [76,147]. The
increased sensitivity to calcium conferred by the b1
subunit is required for the BK
Ca
to translate calcium
sparks to MP hyperpolarization [172]. Impaired b1
subunits inhibited BK
Ca
activation in cerebral arteries
of hypertensive rats, leading to dysfunctional vasodila-
tion [142]. Disruption and knockout of the BK
Ca
-b1
gene (KCNMB1) were associated with elevated blood
pressure in mice, and downregulation of the b1 sub-
unit was been reported in hypertensive rats [72,146].
b1 knockout mice were hypertensive, indicating that
uncoupling BK
Ca
from RyRs by the loss of the b1 sub-
unit alone is sufficient to induce hypertension [145]. It
was demonstrated that b1 knockout mice had higher
systemic blood pressure than control mice (b1-subunit
gene +) [72,173]. It has been also reported that expres-
sion of the pore-forming asubunit of BK
Ca
is increased
in cerebral, mesenteric, and coronary arteries, which
are microcirculatory beds [9].
It has been shown that K
ir
[174] and K
V
currents
decrease in the arterial myocytes of angiotensin II-
induced hypertensive rats [143]. Dilator responses of
the basilar artery to aprikalim, a direct activator of
K
ATP
, are impaired in stroke-prone SHRs in vivo [149].
It has been found that the expression of K
ir
2.1, K
ir
4.1,
K
ir
6.x, and SUR2 mRNA decrease in the isolated
mesenteric arteries of hypertensive mice, and the func-
tional contribution of both K
ir
and K
ATP
decreases in
these mice arteries [115]. There are a significantly
ª2019 Soci
et
e Franc
ßaise de Pharmacologie et de Th
erapeutique
Fundamental & Clinical Pharmacology
Pharmacological perspective of potassium channels 7
Table IV Changes in Potassium Channels in Vascular Diseases.
Disease Type of K
+
channel Species Vascular bed Conclusion References
Hypertension BK
Ca
Rat Mesenteric artery Enhancement of the expression and activity of BK
Ca
increases vasorelaxation [140]
Hypertension BK
Ca
Human Mesentery artery SMCs BK
Ca
activity is decreased [141]
Hypertension BK
Ca
Rat Cerebral artery Increased expression of BK
Ca
channels [76]
Hypertension BK
Ca
-b1 Rat Cerebral artery Impaired b1 subunits and BK
Ca
activation [142]
Hypertension BK
Ca
-b1
K
V
1.2, K
V
1.5
Rat Thoracic aorta Kv1.2/Kv1.5 downregulation and BK
Ca
b1-subunit upregulation [24]
Hypertension K
ir
-K
V
Rat Femoral and cerebral
arteries
Decreased K
+
conductance [143]
Hypertension BK
Ca
Rat Mesenteric artery Increased channel expression and current [66]
Hypertension BK
Ca
Human Mesentery artery BK
Ca
activity is decreased [74]
Hypertension K
ir
2.1, K
ir
4.1 K
ir
6.x Rat Mesenteric artery Decrease in the expression and current [115]
Hypertension BK
Ca
K
V
Rat Mesenteric artery SMCs Increased BK
Ca
current and expression, decreased K
V
current and expression [59]
Hypertension BK
Ca
K
V
Rat Mesenteric artery Increased BK
Ca
current, decreased K
V
current [144]
Hypertension BK
Ca
Rat Cerebral arterial VSMCs Increasing the sensitivity of BK
Ca
to Ca
+2
[145]
Hypertension K
V
1.2, K
V
-b1.1 Rat Thoracic aorta, Tail artery,
Mesenteric artery
Increased K
V
current in SHRs [96]
Hypertension BK
Ca
Human b
1
subunit gene (KCNMB1) KCNMB1 gene polymorphisms may be involved in the pathogenesis of hypertension [146]
Hypertension BK
Ca
Rat Femoral artery BK
Ca
channels are highly activated [52]
Hypertension BK
Ca
Rat Aorta Increased BK
Ca
current and expression [147]
Hypertension K
V
SHRs Isolated myocytes from
thoracic aorta
Decreased K
V
activity, increased BK
Ca
activity [89]
Hypertension BK
Ca
Rat Carotid artery Increased BK
Ca
responsiveness to blockers [148]
Hypertension K
ATP
Rat Basilar artery (in vivo) Impaired K
ATP
activity [149]
Hypertension K
V
7.4 SHRs Renal and mesenteric
arteries
K
V
7.4 protein is downregulated in arteries [98]
Pulmonary hypertension K
2P
(TWIK-2) Mice Pulmonary artery The loss of TWIK-2 function [120]
Pulmonary hypertension K
V
1.5, K
V
2.1 Human PASMCs Decreased expression and activation of K
V
1.5 and K
V
2.1 [44]
Pulmonary hypertension K
V
1.7 Rat PASMCs Decreased expression of K
V
1.7 [150]
Pulmonary hypertension K
V
7.4 Rat Pulmonary artery K
V
7.4 activation prevent hypoxia-induced pulmonary hypertension [131]
Pulmonary hypertension BK
Ca
Rat PASMCs Decreased BK
Ca
currents [135]
Pulmonary hypertension K
V
1.2, K
V
1.5,
K
V
2.1, K
V
9.3
Rat PASMCs Increased the expression of K
V
1.2, K
V
1.5, K
V
2.1, and K
V
9.3 [92]
Pulmonary hypertension K
V
Human Pulmonary Artery Dysfunctional K
V
[151]
Pulmonary hypertension BK
Ca
Human PASMCs Inhibition of the BK
Ca
current [75]
Pulmonary hypertension K
2P
(TASK1) Human PASMCs Decreased channel expression and activity [139]
ª2019 Soci
et
e Franc
ßaise de Pharmacologie et de Th
erapeutique
Fundamental & Clinical Pharmacology
8M.F. Dogan et al.
Table IV Continued
Disease Type of K
+
channel Species Vascular bed Conclusion References
Pulmonary hypertension TASK-1 Human PASMCs Endothelin-1 inhibits channel activity [152]
Pulmonary hypertension K
V
Rat Pulmonary artery Decreased K
V
activity in hypoxia-induced pulmonary hypertension [153]
Atherosclerosis BK
Ca
-b1subunit Human Human aortic VSMCs Decreased b1-subunit expression [154]
Atherosclerosis BK
Ca
Human Cultured coronary
artery SMCs
Increased BK
Ca
activity [155]
Atherosclerosis K
ATP
Monkeys Carotid artery Impaired K
ATP
function [156]
Atherosclerosis BK
Ca
Human Aorta There are kinetically different types of channels [11]
Atherosclerosis K
V
BK
Ca
Atherosclerotic
mouse
Aorta Decreased activity of K
V
and increased BK
Ca
activity [157]
Hypercholesterolemia K
ATP
Rabbit (in vivo) Mesenteric Artery K
ATP
is preserved [158]
Hypercholesterolemia K
ATP
BK
Ca
Rabbit Carotid artery 17b-estradiol protect via K
+
channels [159]
Varicocele K
ATP
Human Internal Spermatic
Vein
Testosterone activates K
ATP
[28]
Diabetes mellitus K
ATP
Rat Mesenteric artery KMUP-1 prevents impairment of K
ATP
[160]
Diabetes mellitus K
V
7.1K
V
7.5 Rat Coronary artery The downregulation of K
V
[138]
Diabetes mellitus K
V
1.1, K
V
1.2, K
V
2.1,
K
V
4.1, K
V
10.1
Rat Mesenteric artery Increased expression of K
+
channels [161]
Diabetes mellitus BK
Ca
Rat Cerebral artery Decreased channel activity and expression [162]
Gestational diabetes K
ATP
Human Umbilical artery Decreased channel expression [163]
ª2019 Soci
et
e Franc
ßaise de Pharmacologie et de Th
erapeutique
Fundamental & Clinical Pharmacology
Pharmacological perspective of potassium channels 9
greater expressions of K
V
1.2 and K
V
b-subunit [96]
and increased K
V
activity with decreased BK
Ca
activity
[89] in SHRs arterial smooth muscle cells. Both BK
Ca
and K
V
are the dominant types of K
+
channels
expressed by VSMCs contributing to a negative feed-
back during agonist-induced vasoconstriction. Pin-
terova et al. have suggested that these K
+
channel
types have impaired function in SHRs [67]. K
V
1.2/
K
V
1.5 downregulation and BK
Ca
b1-subunit upregula-
tion have been demonstrated in SHRs thoracic aortas
[24]. Zhang et al. [59] have shown that current and
expression of the K
V
1.2 and K
V
1.5 were decreased in
mesenteric VSMCs, although BK
Ca
current was dramat-
ically greater in SHRs. Increased gene expression in K
V
may be to normalize resting MP and Ca
+2
influx in
response to compensatory responses such as BK
Ca
[96].
K
V
7 in VSMCs and in baroreceptor neurons have been
demonstrated to serve as important regulators of vas-
cular function [85,99,138]. It has been discovered that
K
V
7.4 protein is downregulated in aorta, renal, and
mesenteric arteries in SHRs when compared with nor-
motensive rats [98,100]. Yıldız et al. [132] demon-
strated that testosterone responses in human internal
mammary, radial, and umbilical arteries altered in
patients with cardiovascular risk factors and/or condi-
tions, such as hyperlipidemia, diabetes, hypertension,
smoking, age, gender, body mass index, and vascular
occlusion.
Pulmonary hypertension
Pulmonary hypertension leading to increased vascular
resistance and right heart dysfunction is a complex,
multifactorial, and refractory pathophysiological condi-
tion characterized by an elevated pulmonary arterial
(PA) pressure and pulmonary vascular resistance at
rest or during exercise [8,44]. Pulmonary circulation
relates to K
+
channels, which are the key determinant
of the resting MP in SMCs and regulate [Ca
+2
]
i
in PA
myocytes that in turn regulate pulmonary vascular
tone and remodeling [175]. Reductions or damages in
the activity and expression of the K
+
channels identi-
fied five major classes in PA are associated with the
pulmonary hypertension in both patients and animal
models [88,176].
BK
Ca
and K
V
indicate prominent contributions in the
physiological regulation of conduit PA smooth muscle
cells (PASMCs); however, K
V
function is predominant
[177]. It has been demonstrated that chronic hypoxia
causes membrane depolarization in human PASMCs
in vivo and in vitro, and the hypoxia-induced
membrane depolarization is partly due to inhibition of
the BK
Ca
[75]. Membrane depolarization resulting in a
decrease in K
+
channel activity may lead to PA vaso-
constriction and eventually to the development of
chronic hypoxic pulmonary hypertension [153]. Patho-
logical and pathophysiological conditions are associated
with abnormal vasoconstriction, vascular injury (in-
duced by ischemia and inflammation), and remodeling
(medial and intimal proliferation) [151]. Hypoxic pul-
monary vasoconstriction (HPV) is a physiological mod-
ulator response in the case of alveolar hypoxia that
distributes the blood flow of pulmonary capillary to
regions where the amount of oxygen is greater.
Hypoxia may occur with the disruption of this mecha-
nism. It reduces perfusion in poorly ventilated alveoli
in order to optimize the oxygenation of arterial blood
[131,178]. Activation of the K
+
channels plays an
important role in the inhibition of HPV. Bonnet et al.
[179] showed that dehydroepiandrosterone administra-
tion in hypoxic rats prevented PA remodeling and
decreased pulmonary hypertension via BK
Ca
stimula-
tion and increased expression levels. Hypoxia has been
reported to inhibit K
V
and cause vasoconstriction as a
result of membrane depolarization in PASMCs [2,135].
Exposure to hypoxia has been demonstrated to inhibit
K
V
1.5 and K
V
2.1 opening, in turn, leads to LVCC acti-
vation and PA vasoconstriction [44,153,180]. It has
been also reported that exposure to hypoxia signifi-
cantly increased expression of the K
V
1.2, K
V
1.5, K
V
2.1,
and K
V
9.3 [92,177]. Mitochondrial-hypoxia-inducible
factor-1aactivation suppresses K
V
1.5 in PASMCs of
spontaneously pulmonary hypertensive fawn-hooded
rats which points out that the mitochondria can be
targeted therapeutically [181]. Mutations, which seem
to increase the severity of the disease, are less frequent
in K
V
1.5 (encoded by KCNA5) [182]. In addition, dehy-
droepiandrosterone reduces hypoxia-inducible factor-1a
accumulation under hypoxia in human PA cells [183].
Changes in K
V
function include regulation of cell prolif-
eration and apoptosis eventually leading to pulmonary
vascular remodeling, and the decreased expression and
activity of K
V
may lead to increased PA vasoconstric-
tion, which causes the pulmonary hypertension [184].
It was shown that decreased K
V
1.5 [130] and K
V
1.7
expression contributes to pulmonary hypertension in
rats [150]. K
V
1.5 is an important O
2
-sensitive channel
and potential therapeutic target in pulmonary hyper-
tension. Pozeg et al. [185] demonstrated that K
V
1.5
gene therapy restored HPV and attenuates pulmonary
hypertension of rats. Also, the loss of K
V
7.4 activity,
ª2019 Soci
et
e Franc
ßaise de Pharmacologie et de Th
erapeutique
Fundamental & Clinical Pharmacology
10 M.F. Dogan et al.
which plays an important role in the pathogenesis of
pulmonary hypertension, can contribute to enhanced
excitation and vasoreactivity. The protective role for
K
V
7.4 in the pulmonary circulation has been shown to
inhibit hypoxia-induced pulmonary hypertension,
restricting increased vasoreactivity to pressure agents
[131,176,178].
The pathogenesis of idiopathic and hypoxic pul-
monary hypertension is related to the dysfunction of
the K
+
channels. Increased [Ca
+2
]
i
stimulates cellular
proliferation and triggers vasoconstriction. Hypoxia
causes a decrease in whole-cell BK
Ca
currents in
PASMCs [135,186,187]. K
2P
3.1 (TASK-1) and K
2P
6.1
(TWIK-2) subunits involved in pulmonary hyperten-
sion, and the K
2P
3.1- and K
2P
6.1-knockout mice exhib-
ited pulmonary vascular remodeling with increased
vessel diameters, accompanied by enhanced right ven-
tricular pressure indicative of pulmonary hypertension
[188,189]. TASK-1 (encoded by KCNK3) also modu-
lates PA tone. It has been demonstrated that TASK-1
probably play an important role in rabbit PASMCs
mediating pulmonary vascular responses to changes in
pH and might be responsible for the regulatory effects
of pH on HPV [123]. Patch-clamp experiments demon-
strated that mutations in this gene produced reduced
KCNK3 current in PA hypertension [190]. Antigny
et al. [139] reported that KCNK3 expression and func-
tion were reduced in monocrotaline-induced pul-
monary hypertension in rats and in human with
pulmonary hypertension. Moreover, in vivo pharmaco-
logical activation with ONO-RS-082 of KCNK3 signifi-
cantly reversed pulmonary hypertension in rats [139]
and partially restored the function of some KCNK3
mutants channels [190]. KCNK3 mutations may be
associated with certain forms of pulmonary hyperten-
sion, and they may cause worsening of the clinical fea-
tures in homozygous patients [191,192]. Higasa et al.
[191] suggested that rare pathogenic variants in
KCNK3 may also increase the risk of familial PA hyper-
tension. Cunningham et al. [193] showed that the
guanylate cyclase activator, riociguat, a novel treat-
ment for pulmonary hypertension, enhances current
through TASK-1 channels but does not recover current
through mutant TASK-1 channels, seen in pulmonary
hypertension patients. While there is compelling evi-
dence that KCNK3 is involved in the pathogenesis of
pulmonary hypertension in humans [194], KCNK3
does not play a key role in initiating HPV of mice pul-
monary hypertension [189,195], but it may participate
in its maintenance [195]. However, KCNK3/TASK-1
channel did not form a functional channel in mice
which is not a suitable model to study KCNK3 in PA
hypertension [196]. Loss of KCNK3 function and
expression is a hallmark of right ventricle hypertrophy/
dysfunction related to pulmonary hypertension [197].
Lambert et al. [198] reported that KCNK3 function is
severely decreased in right ventricle cardiomyocytes
during the development of right ventricle hypertrophy
in rat models of pulmonary hypertension. Ten different
KCNK3 mutations, which cause damaged channel
activity, described in PA hypertension so far, and novel
candidate drugs to restore KCNK3 function should be
investigated [199]. Moreover, TWIK2 (encoded by
KCNK6) is also involved in pulmonary hypertension
since KCNK6-deficient mice showed increase in right
ventricular pressure [120]. On the other hand, it has
been recently observed that ABCC8 mutations in
patients with pulmonary hypertension decrease K
ATP
function, which is pharmacologically recovered in vitro
by the SUR1 activator, diazoxide [200].
K
+
channel openers have been found to be useful by
inhibiting HPV and also decreasing PA resistance in
models of pulmonary hypertension, which are other-
wise resistant to conventional pharmacotherapy [201].
Hypercholesterolemia and atherosclerosis
Hypercholesterolemia is recognized as one of major
independent risk factor for initiating the complex pro-
cess of atherosclerosis [202]. Hypercholesterolemia and
atherosclerosis are both associated with an impairment
of the endothelium-dependent and K
+
channel-
mediated relaxations in VSMCs. Atherosclerosis is a
chronic inflammatory disease resulting from the accu-
mulation of lipids (such as cholesterol crystals, modified
fatty acids, and lysophospholipids) and fibrous elements
in the coronary artery and aorta. It is known that
VSMCs are closely involved in the development of
atherosclerosis [11,154,203]. This disease is particu-
larly associated with the decreased vascular activity of
endothelium-derived nitric oxide, which is likely to play
an important role in the development of atherosclerosis
with aging. As a result, arteries may exhibit increased
vascular tone under basal conditions and may respond
poorly to endothelium-dependent vasodilator agonists.
In the case of changes in vascular activity, the K
+
channel activity or function may also be abnormal.
The proliferation and migration of VSMCs from arterial
media have been recognized as essential steps in the
development of stenosing atherosclerotic plaques in
humans [25,155,164].
ª2019 Soci
et
e Franc
ßaise de Pharmacologie et de Th
erapeutique
Fundamental & Clinical Pharmacology
Pharmacological perspective of potassium channels 11
Vascular smooth muscle is dependent on stabilizing
mechanisms for limiting Ca
+2
entry, and these mecha-
nisms may be a decrease in susceptibility to vasoconstric-
tors or an increase in activation of BK
Ca
. The opening of
K
v
in the atherosclerotic mouse aorta is decreased, and it
is susceptible to be depolarized resulting in Ca
+2
entry
[157]. It has been demonstrated that disruption of the
KCNE2 gene, which encodes a K
V
accessory subunit (b
subunit), causes atherosclerosis in mice [204]. K
V
contri-
bution to adenosine-mediated relaxation abolished by
hypercholesterolemia in porcine coronary arterioles
[205]. Recent studies have shown altered function and
expression of the vascular K
ATP
and b1-subunit of BK
Ca
in
experimental hypercholesterolemia and atherosclerotic
patients [154,206]. Hypercholesterolemia is associated
with impairment of BK
Ca
-mediated vasodilatation in
cholesterol-fed rabbits [207]. BK
Ca
activity is modulated
by membrane cholesterol. As both cholesterol levels and
BK
Ca
involved in pathophysiological mechanism in vascu-
lar disease, identifying disruption of BK
Ca
-cholesterol
interaction may provide new approaches for therapeutic
intervention [208]. Low cholesterol levels suppressed
BK
Ca
-b1 subunit-mediated channel inhibition and vascu-
lar constriction in mouse arteries [209]. Faraci et al.
[156] suggested that in addition to impairment of
endothelium-dependent responses, atherosclerosis impairs
relaxation of the carotid artery in response to activation
of K
ATP
. Altered K
ATP
function in the regulation of coro-
nary vascular tone has been reported in experimental
hypercholesterolemia [210]. 17b-estradiol protects the
hypercholesterolemic rabbit carotid arteries by maintain-
ing both endothelium and K
ATP
and BK
Ca
structures
[159]. Loss of this protective mechanism increases the
risk of thrombosis and the effects of vasospasm-related
vasoconstrictor factors on the arterial vessel wall [211].
Also, in animal models of atherosclerosis or hypercholes-
terolemia, damage of acetylcholine-induced relaxation
responses of endothelium has been associated with rela-
tive protection of endothelium-independent vasodilatation
[158]. Two types of BK
Ca
are present in SMC obtained
from the atherosclerotic aorta. Along with L-type chan-
nels, typical of normal adult human aorta, the channels
of S-type with different kinetic properties were also pre-
sent in these cells. Under similar conditions, S-type chan-
nels had about a four-times-shorter opening time and
considerably lower Ca
+2
sensitivity [11].
Diabetes mellitus
Diabetes mellitus is one of the most prevalent diseases
in the world, and vascular dysfunction is one of the
most important complications in diabetic patients.
Diabetes mellitus is closely related to vascular dysfunc-
tion through both endothelium-independent and
endothelium-dependent mechanisms. Vascular function
abnormalities contribute to the etiology of many dia-
betic complications such as neuropathy, retinopathy,
and myopathy [25,161]. K
ATP
in VSMCs are regulated
in decreasing amounts in diseases like diabetes mellitus
and hypertension, and potentially contribute to the
end-organ problems in these diseases [90,212]. K
ATP
are found in pancreatic-b-cells as well as in VSMCs.
K
ATP
play a crucial role in regulation of blood glucose
level by adjusting insulin release from these cells.
Firstly, K
ATP
was discovered in rabbit mesenteric artery
in 1989 [110,112,201]. Recently, it has been demon-
strated that reduction of K
ATP
current and K
ATP
chan-
nel-induced vasorelaxation in human umbilical artery
smooth muscle are due to the decreased expression of
K
ATP
channels in during gestational diabetes mellitus
[163]. Moreover, KMUP-1, a xanthine derivative, regu-
lates K
ATP
activity and prevents impairment of mesen-
teric artery reactivity and vascular dysfunction in
diabetic rats [160]. The reduction in BK
Ca
-b1 expres-
sion in renal arterioles of type I diabetic patients [70]
and in cerebral artery of type II diabetic rats [162] has
been shown to result in an increase in the myogenic
tone. N-3 polyunsaturated fatty acids protect the coro-
nary BK
Ca
function and vasoreactivity in diabetic rats
as a result of not only increasing BK
Ca
-b1 expressions,
but also decreasing artery tension and [Ca
+2
]
i
[213].
Hyperglycemia increases oxygen production both in
humans and in animals, and vascular K
ATP
inhibition
is formed. Continuous inhibition of K
ATP
may cause
deterioration of the relaxation response of the vessels
[4]. Also, the changes in the K
V
current have been
shown in some diabetic vessels [15,214]. Hong and
colleagues reported that the expression and flow of
many subunits of K
V
(such as K
V
1.1, 1.2, 1.4, and
1.5) are increased in early and chronic stages of
diabetes mellitus in the mesenteric artery of type II
diabetic rats [161].
CONCLUSIONS
K
+
channels are the major ion conductance of the
VSM cell membrane, and they dominantly regulate MP
which is a bright tool for processing vascular tone
quickly, efficiently, and precisely. The impressive com-
plexity of the numerous combinations of K
+
channels
allows precise adjustment of vascular response to direct
ª2019 Soci
et
e Franc
ßaise de Pharmacologie et de Th
erapeutique
Fundamental & Clinical Pharmacology
12 M.F. Dogan et al.
environment and provides a steady blood flow to
compensate for pathological conditions by responding
to various physiological situations.
Regarding altered K
+
channels in presented vascular
diseases, several drugs such as riociguat (pulmonary
hypertension) [215], nicorandil (angina pectoris) [216],
minoxidil, diazoxide, and pinacidil (hypertension) [111]
are used clinical practice. Also, promising new-genera-
tion drugs such as dehydroepiandrosterone [217], ipta-
kalim [218], dichloroacetate [219], levcromakalim
[220], and other benzopyran derivatives [221] have
been developed and/or introduced to the market.
There are many opportunities for future research on
vascular tone regulation through K
+
channels [222
224]. Nonetheless, our understanding is lacking about
the regional heterogeneity in the nature and composi-
tion of K
+
channels in different vascular beds. More-
over, given strong evidence of the critical role of K
+
channels in a variety of vascular pathological condi-
tions including hypertension, PA hypertension,
atherosclerosis, and diabetes mellitus, targeting K
+
channel function represents a novel and promising
strategy for the prevention and treatment of a variety
of diseases [225229]. However, many complex alter-
ations in vascular K
+
channel functions have been
described in diseases, and the understanding of the reg-
ulation of expression and function of the K
+
channels
in these disease states is also limited, especially since
these diseases relate to different vascular beds through-
out the body.
As further progress is made, along with molecular
and electrophysiological approaches, we will clarify the
expression and function of different sequences of K
+
channels contributing to the regulation of VSM con-
tractility in different cell types and in different segmen-
tal regions. This information is crucial for the
understanding of certain K
+
channel abnormalities and
their causes in specific diseases. The future of therapeu-
tic applications by activation of vascular K
+
channels
would seem to be a very promising in numerous vascu-
lar disease states.
REFERENCES
1 Beech D.J. Ion channel switching and activation in smooth-
muscle cells of occlusive vascular diseases. Biochem. Soc.
Trans. (2007) 35 890894.
2 Nelson M.T., Quayle J.M. Physiological roles and properties of
potassium channels in arterial smooth muscle. Am. J.
Physiol. (1995) 268 C799C822.
3 Afsar S., Hemsinli D., Ozyazgan S., Akkan A.G., Arslan C.
The effects of potassium channels in human internal
mammary artery. Pharmacol. J. (2016) 97 7277.
4 Climent B., Simonsen U., Rivera L. Effects of obesity on
vascular potassium channels. Curr. Vasc. Pharmacol. (2014)
12 438452.
5 Cox R.H. Changes in the expression and function of arterial
potassium channels during hypertension. Vascul. Pharmacol.
(2002) 38 1323.
6 Pathan A.R., Rusch N.J. Two-pore domain K+channels
evidence for TWIK-2 in blood pressure regulation.
Hypertension (2011) 58 539541.
7 Sakakibara K., Feng G.G., Li J. et al. Kynurenine causes
vasodilation and hypotension induced by activation of
KCNQ-encoded voltage-dependent K+channels. J. Pharmacol.
Sci. (2015) 129 3137.
8 Hayabuchi Y. The action of smooth muscle cell potassium
channels in the pathology of pulmonary arterial
hypertension. Pediatr. Cardiol. (2017) 38 114.
9 Joseph B.K., Thakali K.M., Moore C.L., Rhee S.W. Ion
channel remodeling in vascular smooth muscle during
hypertension: implications for novel therapeutic approaches.
Pharmacol. Res. (2013) 70 126138.
10 Korovkina V.P., England S.K. Detection and implications of
potassium channel alterations. Vascul. Pharmacol. (2002)
38 312.
11 Bolotina V., Gericke M., Bregestovski P. Kinetic differences
between Ca(2 +)-dependent K+channels in smooth muscle
cells isolated from normal and atherosclerotic human aorta.
Proc. R. Soc. London B (1991) 244 5155.
12 Clapp L.H., Tinker A. Potassium channels in the vasculature.
Curr. Opin. Nephrol. Hypertens. (1998) 79198.
13 Humphries E.S.A., Dart C. Neuronal and cardiovascular
potassium channels as therapeutic drug targets: promise and
pitfalls. J. Biomol. Screen. (2015) 20 10551073.
14 Baranowska M., Kozłowska H., Korbut A., Malinowska B.
Potassium channels in blood vessels: their role in
health and disease. Postep. Hig Med Dosw (2007) 61
596605.
15 Ko E.A., Park W.S., Firth A.L., Kim N., Jason X.-J., Han J.
Pathophysiology of voltage-gated K+channels in vascular
smooth muscle cells: modulation by protein kinases. Prog.
Biophys. Mol. Biol. (2010) 103 95101.
16 Werner M.E., Ledoux J. K+channels in biological processes:
vascular K+channels in the regulation of blood pressure.
Noteworthy J. (2014) 75160.
17 Boucherat O., Chabot S., Antigny F., Perros F., Provencher
S., Bonnet S. Potassium channels in pulmonary arterial
hypertension. Eur. Respir. J. (2015) 46 11671177.
18 Lawson K., McKay N.G. Modulation of potassium channels
as a therapeutic approach. Curr. Pharm. Des. (2006) 12
459470.
19 Dick G.M., Tune J.D. Role of potassium channels in coronary
vasodilation. Exp. Biol. Med. (2010) 235 1022.
20 Eichhorn B., Dobrev D. Vascular large conductance calcium-
activated potassium channels: functional role and
ª2019 Soci
et
e Franc
ßaise de Pharmacologie et de Th
erapeutique
Fundamental & Clinical Pharmacology
Pharmacological perspective of potassium channels 13
therapeutic potential. Naunyn Schmiedebergs Arch.
Pharmacol. (2007) 376 145155.
21 Whidden M.A., Basgut B., Kirichenko N., Erdos B., T
umer N.
Altered potassium ATP channel signaling in mesenteric
arteries of old high salt-fed rats. J. Exerc. Nutr. Biochem.
(2016) 20 5864.
22 Marinko M., Novakovic A., Nenezic D. et al. Nicorandil
directly and cyclic GMP-dependently opens K+channels in
human bypass grafts. J. Pharmacol. Sci. (2015) 128 5964.
23 Gollasch M., Ried C., Bychkov R., Luft F.C., Haller H. K+
currents in human coronary artery vascular smooth muscle
cells. Circ. Res. (1996) 78 676688.
24 Li Z., Lu N., Shi L. Exercise training reverses alterations in
Kv and BKCa channel molecular expression in thoracic aorta
smooth muscle cells from spontaneously hypertensive rats.
J. Vasc. Res. (2014) 51 447457.
25 Sobey C.G. Potassium channel function in vascular disease.
Arterioscler. Thromb. Vasc. Biol. (2001) 21 2838.
26 Karabacak K., Kaya E., Ulusoy K.G. et al. Effects of taurine
on contractions of human internal mammary artery: a
potassium channel opening action. Eur. Rev. Med.
Pharmacol. Sci. (2015) 19 14981504.
27 Ulusoy K.G., Kaya E., Karabacak K. et al. Taurine relaxes
human radial artery through potassium channel opening
action. Korean J. Physiol. Pharmacol. (2017) 21 617623.
28 Seyrek M., Irkilata H.C., Vural I.M. et al. Testosterone
relaxes human internal spermatic vein through potassium
channel opening action. Urology (2011) 78 233.e1233.e5.
29 Yildiz O., Seyrek M., Gul H., Un I. Testosterone relaxes
human internal mammary artery in vitro. J. Cardiovasc.
Pharmacol. (2005) 45 580585.
30 Caliskan E., Kayhan Z., Tufan H. Propofol inhibits potassium
chloride induced contractions of isolated human umbilical
vessels. Eur. J. Anaesthesiol. (2006) 23 411417.
31 Dogan M.F., Arslan S.O., Yildiz O., Kurtoglu M., Parlar A.
Propofol-induced vasodilation in human internal mammary
artery: role of potassium channels. J. Cardiothorac. Vasc.
Anesth. (2018). https://doi.org/10.1053/j.jvca.2018.12.015.
32 Mart
ın P., Moncada M., Kuntamallappanavar G., Dopico A.M.,
Milesi V. Activation of human smooth muscle BK channels by
hydrochlorothiazide requires cell integrity and the presence of
BK b1subunit. Acta Pharmacol. Sin. (2018) 39 371381.
33 Mart
ın P., Enrique N., Palomo A.R.R., Rebolledo A., Milesi
V. Bupivacaine inhibits large conductance, voltage- and
Ca2 +- activated K+channels in human umbilical artery
smooth muscle cells. Channels (Austin) (2012) 6174180.
34 Novakovic A., Marinko M., Vranic A. et al. Mechanisms
underlying the vasorelaxation of human internal mammary
artery induced by (-)-epicatechin. Eur. J. Pharmacol. (2015)
762 306312.
35 Yildiz O., Nacitarhan C., Seyrek M. Potassium channels in the
vasodilating action of levosimendan on the human umbilical
artery. J. Soc. Gynecol. Investig. (2006) 13 312315.
36 Wallerstedt S.M., Tornebrandt K., Bodelsson M. Relaxant
effects of propofol on human omental arteries and veins. Br.
J. Anaesth. (1998) 80 655659.
37 L
opez-Dyck E., Andrade-Urz
ua F., Elizalde A. et al. ACPA
and JWH-133 modulate the vascular tone of superior
mesenteric arteries through cannabinoid receptors, BKCa
channels, and nitric oxide dependent mechanisms.
Pharmacol. Rep. (2017) 69 11311139.
38 Chanda D., Prieto-Lloret J., Singh A. et al. Glabridin-induced
vasorelaxation: evidence for a role of BKCa channels and
cyclic GMP. Life Sci. (2016) 165 2634.
39 Niu L.G., Zhang M.S., Liu Y. et al. Vasorelaxant effect of
taurine is diminished by tetraethylammonium in rat isolated
arteries. Eur. J. Pharmacol. (2008) 580 169174.
40 Wan H.-J., Wang Y., Si J.-Q., Li L. Propofol-induced
vasodilation of mesenteric arterioles via BKCa channel and gap
junction. Exp. Ther. Med. (2018) 16 29602968.
41 Nagakawa T., Yamazaki M., Hatakeyama N., Stekiel T.A.
The mechanisms of propofol-mediated hyperpolarization of
in situ rat mesenteric vascular smooth muscle. Anesth.
Analg. (2003) 97 16391645.
42 Li Z., Zhang M., Liang Y. Endothelium-independent
vasorelaxant effect of Taurine on rat aorta rings. Zhongguo
Zhong Yao Za Zhi (2009) 34 332335.
43 Novakovic A., Marinko M., Jankovic G. et al. Endothelium-
dependent vasorelaxant effect of procyanidin B2 on human
internal mammary artery. Eur. J. Pharmacol. (2017) 807
7581.
44 Zhang S., Liu B., Fan Z. et al. Targeted inhibition of survivin
with YM155 promotes apoptosis of hypoxic human
pulmonary arterial smooth muscle cells via the upregulation
of voltage-dependent K+channels. Mol. Med. Rep. (2016) 13
34153422.
45 Yildiz O., Seyrek M., Yildirim V., Demirkilic U., Nacitarhan C.
Potassium channel-related relaxation by levosimendan in the
human internal mammary artery. Ann. Thorac. Surg.
(2006) 81 17151719.
46 Revermann M., Schloss M., Mieth A. et al. Levosimendan
attenuates pulmonary vascular remodeling. Intensive Care
Med. (2011) 37 13681377.
47 Lam C.F., Chang P.J., Chen Y.A., Yeh C.Y., Tsai Y.C.
Inhibition of ATP-sensitive potassium channels attenuates
propofol-induced vasorelaxation. Crit. Care Resusc. (2010)
12 186190.
48 Kinoshita H., Ishida K., Ishikawa T. Thiopental and propofol
impair relaxation produced by ATP-sensitive potassium
channel openers in the rat aorta. Br. J. Anaesth. (1998) 81
766770.
49 Roh W.-S., Ding X., Murray P.A. Propofol and thiopental
attenuate adenosine triphosphate-sensitive potassium
channel relaxation in pulmonary veins. Am. J. Physiol. Lung
Cell. Mol. Physiol. (2006) 291 L636L643.
50 Murphy M.E., Brayden J.E. Nitric oxide hyperpolarizes rabbit
mesenteric arteries via ATP-sensitive potassium channels. J.
Physiol. (1995) 486 4758.
51 Gu
eguinou M., Chant^
ome A., Fromont G., Bougnoux P.,
Vandier C., Potier-Cartereau M. KCa and Ca2 +channels:
the complex thought. Biochim. Biophys. Acta (2014) 1843
23222333.
ª2019 Soci
et
e Franc
ßaise de Pharmacologie et de Th
erapeutique
Fundamental & Clinical Pharmacology
14 M.F. Dogan et al.
52 Asano M., Nomura Y., Ito K., Uyama Y., Imaizumi Y.,
Watanabe M. Increased function of voltage-dependent Ca++
channels and Ca(++)-activated K+channels in resting state
of femoral arteries from spontaneously hypertensive rats at
prehypertensive stage. J. Pharmacol. Exp. Ther. (1995) 275
775783.
53 Toyama K., Wulff H., Chandy K.G. et al. The intermediate-
conductance calcium- activated potassium channel KCa3.1
contributes to atherogenesis in mice and humans. J. Clin.
Invest. (2008) 118 30253038.
54 Yamada K. Vascular potassium channels in NVC. Prog.
Brain Res. (2016) 225 6373.
55 Sorensen C., Braunstein T., Holstein-Rathlou N.,
Salomonsson M. Role of vascular potassium channels in the
regulation of renal hemodynamics. Am. J. Physiol. Renal
Physiol. (2012) 302 505518.
56 Brayden J.E. Potassium channels in vascular smooth muscle.
Clin. Exp. Pharmacol. Physiol. (1996) 23 10691076.
57 Kume H., Takagi K., Satake T., Tokuno H., Tomita T. Effects
of intracellular pH on calcium-activated potassium channels
in rabbit tracheal smooth muscle. J. Physiol. (1990) 424
445457.
58 Benham C.D., Bolton T.B., Lang R., Takewaki T. Calcium-
activated potassium channels in single smooth muscle cells
of rabbit jejunum and guinea-pig mesenteric artery. J.
Physiol. (1986) 371 4567.
59 Zhang Y., Gao Y.J., Zuo J., Lee R.M.K.W., Janssen L.J.
Alteration of arterial smooth muscle potassium channel
composition and BKCa current modulation in hypertension.
Eur. J. Pharmacol. (2005) 514 111119.
60 Nelson M.T., Cheng H., Rubart M. et al. Relaxation of
arterial smooth muscle by calcium sparks. Science (1995)
270 633637.
61 Yang Y., Cai F., Li P.Y. et al. Activation of high conductance
Ca2 +-activated K+channels by sodium tanshinoneII-A
sulfonate (DS-201) in porcine coronary artery smooth
muscle cells. Eur. J. Pharmacol. (2008) 598 915.
62 F
urstenau M., L
ohn M., Ried C., Luft F.C., Haller H.,
Gollasch M. Calcium sparks in human coronary artery
smooth muscle cells resolved by confocal imaging. J.
Hypertens. (2000) 18 12151222.
63 Asano M., Masuzawa-Ito K., Matsuda T. et al. Functional
role of charybdotoxin-sensitive K+channels in the resting
state of cerebral, coronary and mesenteric arteries of the
dog. J. Pharmacol. Exp. Ther. (1993) 267 12771285.
64 Gollasch M., L
ohn M., F
urstenau M., Nelson M.T., Luft F.C.,
Haller H. Ca2 +channels, Ca2 +sparks, and regulation of
arterial smooth muscle function. Z. Kardiol. (2000) 89
1519.
65 Dopico A.Μ., Kirber M.T., Singer J.J., Walsh J.V. Jr
Membrane stretch directly activates large conductance
Ca2 +-activated K+channels in mesenteric artery smooth
muscle cells. Am. J. Hypertens. (1994) 78289.
66 Hu Z., Ma A., Tian H., Xi Y., Fan L., Wang T. Effects of age
on expression of BKca channel in vascular smooth muscle
cells from mesenteric arteries of spontaneously hypertensive
rats. J. Physiol. Biochem. (2013) 69 945955.
67 Pint
erov
a M., Behuliak M., Kune
s J., Zicha J. Involvement of
BKCa and KV potassium channels in cAMP-induced
vasodilatation: their insufficient function in genetic
hypertension. Physiol. Res. (2014) 63 275285.
68 Tanaka Y., Meera P., Song M., Knaus H.G., Toro L.Molecular
constituents of maxi K(Ca) channels in human coronary
smooth muscle: predominant a +B subunit complexes. J.
Physiol. (1997) 502 545557.
69 Meera P., Wallner M., Toro L. A neuronal beta subunit
(KCNMB4) makes the large conductance, voltage- and
Ca2 +-activated K+channel resistant to charybdotoxin and
iberiotoxin. Proc. Natl Acad. Sci. USA (2000) 97 5562
5567.
70 Grimm P.R., Sansom S.C. BK channels and a new form of
hypertension. Kidney Int. (2010) 78 956962.
71 Yang Y., Sohma Y., Nourian Z. et al. Mechanisms
underlying regional differences in the Ca2 +sensitivity of
BKCa current in arteriolar smooth muscle. J. Physiol. (2013)
591 12771293.
72 Xu H., Garver H., Fernandes R. et al. BK channel b1-subunit
deficiency exacerbates vascular fibrosis and remodeling but
does not promote hypertension in high fat fed obesity in
mice. J. Hypertens. (2015) 33 16111623.
73 Carvalho-de-Souza J.L., Varanda W.A., Tostes R.C., Chignalia
A.Z. BK channels in cardiovascular diseases and aging.
Aging Dis. (2013) 43849.
74 Yang Y., Li P.-Y., Cheng J. et al. Function of BKCa channels
is reduced in human vascular smooth muscle cells from Han
Chinese patients with hypertension. Hypertension (2013) 61
519525.
75 Peng W., Hoidal J.R., Farrukh I.S. Role of a novel KCa
opener in regulating K+channels of hypoxic human
pulmonary vascular cells. Am. J. Respir. Cell Mol. Biol.
(1999) 20 737745.
76 Liu Y., Pleyte K., Knaus H., Rusch N.J. Increased expression
of Ca +2 sensitive K+channels in the cerebral
microcirculation of genetically hypertensive rats. Exp. Anim.
(1998) 82 729737.
77 Martens J.R., Gelband C.H. Ion channels in vascular smooth
muscle: alterations in essential hypertension. Proc. Soc. Exp.
Biol. Med. (1998) 218 192203.
78 Clapp L.H., Turcato S., Hall S., Baloch M. Evidence that
Ca2 +-activated K+channels play a major role in mediating
the vascular effects of iloprost and cicaprost. Eur. J.
Pharmacol. (1998) 356 215224.
79 Xu H., Garver H., Galligan J.J., Fink G.D. Large-conductance
Ca +2 activated K+channel b1-subunit knockout mice are
not hypertensive. Am J Physiol Hear. Circ Physiol (2011)
300 H476H485.
80 Candia S., Garcia M.L., Latorre R. Mode of action of
iberiotoxin, a potent blocker of the large conductance Ca
(2 +)-activated K+channel. Biophys. J . (1992) 63 583
590.
ª2019 Soci
et
e Franc
ßaise de Pharmacologie et de Th
erapeutique
Fundamental & Clinical Pharmacology
Pharmacological perspective of potassium channels 15
81 Malinowski M., Deja M.A., Janusiewicz P., Golba K.S.,
Roleder T., Wos S. Mechanisms of vasodilatatory effect of
perivascular tissue of human internal thoracic artery. J.
Physiol. Pharmacol. (2013) 64 309316.
82 Hilgers R.H.P., Todd J., Webb R.C. Regional heterogeneity in
acetylcholine-induced relaxation in rat vascular bed: role of
calcium-activated K+channels. Am. J. Physiol. (2006) 291
H216H222.
83 Holland M., Langton P.D., Standen N.B., Boyle J.P. Effects of
the BKCa channel activator, NS1619, on rat cerebral artery
smooth muscle. Br. J. Pharmacol. (1996) 117 119129.
84 Dong D.-L., Bai Y.-L., Cai B.-Z. Calcium-activated potassium
channels: potential target for cardiovascular diseases. Adv.
Protein Chem. Struct. Biol. (2016) 104 233261.
85 Zhong X.Z., Harhun M.I., Olesen S.P. et al. Participation of
KCNQ (Kv7) potassium channels in myogenic control of
cerebral arterial diameter. J. Physiol. (2010) 588 3277
3293.
86 Brayden J.E., Nelson M.T. Regulation of arterial tone by
activation of calcium-dependent potassium channels. Science
(1992) 256 532535.
87 Silberberg S.D., van Breemen C. A potassium current
activated by lemakalim and metabolic inhibition in rabbit
mesenteric artery. Eur. J. Physiol. (1992) 420 118120.
88 Das M., Fessel J., Tang H., West J. A process-based review of
mouse models of pulmonary hypertension. Pulm. Circ.
(2012) 2415433.
89 Cox R.H. Comparison of K+channel properties in freshly
isolated myocytes from thoracic aorta of WKY and SHR.
Am. J. Hypertens. (1996) 9884894.
90 Ko E.A., Han J., Jung I.D., Park W.S. Physiological roles of
K+channels in vascular smooth muscle cells. J. Smooth
Muscle Res. (2008) 44 6581.
91 Cox R.H., Fromme S. Comparison of voltage gated K+
currents in arterial myocytes with heterologously expressed
Kv subunits. Cell Biochem. Biophys. (2016) 74 499511.
92 Dong Q., Zhao N., Xia C., Du L., Fu X.X., Du Y.M. Hypoxia
induces voltage-gated K+(Kv) channel expression in
pulmonary arterial smooth muscle cells through hypoxia-
inducible factor-1 (HIF-1). Bosn. J. Basic Med. Sci. (2012) 12
158163.
93 Stott J.B., Jepps T.A., Greenwood I.A. KV7 potassium
channels: a new therapeutic target in smooth muscle
disorders. Drug Discov. Today (2014) 19 413424.
94 Jepps T.A., Olesen S.P., Greenwood I.A. One man’s side effect
is another man’s therapeutic opportunity: targeting Kv7
channels in smooth muscle disorders. Br. J. Pharmacol.
(2013) 168 1927.
95 Cox R.H., Petrou S. Ca2 +influx inhibits voltage-dependent
and augments Ca2 +-dependent K+currents in arterial
myocytes. Am. J. Physiol. Cell Physiol. (1999) 277
C51C63.
96 Cox R.H., Folander K., Swanson R. Differential expression of
voltage-gated K+channel genes in arteries from
spontaneously hypertensive and Wistar-Kyoto Rats.
Hypertension (2001) 37 13151322.
97 Novakovic A., Pavlovic M., Stojanovic I. et al. Different K+
channels are involved in relaxation of arterial and venous
graft induced by nicorandil. J. Cardiovasc. Pharmacol.
(2011) 58 602608.
98 Carr G., Barrese V., Stott J.B. et al. MicroRNA-153
targeting of KCNQ4 contributes to vascular dysfunction
in hypertension. Cardiovasc. Res. (2016) 112 581
589.
99 Mackie A.R., Byron K.L. Cardiovascular KCNQ (Kv7)
potassium channels: physiological regulators and new
targets for therapeutic intervention. Mol. Pharmacol. (2008)
74 11711179.
100 Stott J.B., Barrese V., Jepps T.A., Leighton E.V., Greenwood
I.A. Contribution of Kv7 channels to natriuretic peptide
mediated vasodilation in normal and hypertensive rats.
Hypertension (2015) 65 622628.
101 Aziz Q., Thomas A.M., Gomes J. et al. The ATP-sensitive
potassium channel subunit, Kir6.1, in vascular smooth
muscle plays a major role in blood pressure control.
Hypertension (2014) 64 523529.
102 Nichols C.G., Singh G.K., Grange D.K. K ATP channels and
cardiovascular disease : suddenly a syndrome. Circ. Res.
(2014) 112 10591072.
103 Rubaiy H.N. The therapeutic agents that target ATP-sensitive
potassium channels. Acta Pharm. (2016) 66 2334.
104 Noma A. ATP-regulated K+channels in cardiac muscle.
Nature (1983) 305 147148.
105 Satake N., Shibata M., Shibata S. The inhibitory effects of
iberiotoxin and 4-aminopyridine on the relaxation induced
by beta 1- and beta 2-adrenoceptor activation in rat aortic
rings. Br. J. Pharmacol. (1996) 119 505510.
106 Aguilar-Bryan L., Clement J.P., Gonzalez G., Kunjilwar K.,
Babenko A., Bryan J. Toward understanding the assembly
and structure of K(ATP) channels. Physiol. Rev. (1998) 78
227245.
107 Li A., Knutsen R.H., Zhang H. et al. Hypotension due to
Kir6.1 gain-of-function in vascular smooth muscle. J. Am.
Heart Assoc. (2013) 2113.
108 Nelson M., Patlak J., Worley J., Standen N. Calcium channels,
potassium channels, and voltage dependence of arterial smooth
muscle tone. Am. J. Physiol. (1990) 259 C3C18.
109 Brayden J.E., Huang Y., Nelson M.T. Hyperpolarizing
vasodilators activate ATP-sensitive K+channels in arterial
smooth muscle. Science (1989) 245 177180.
110 Shi W.-W., Yang Y., Shi Y., Jiang C. K(ATP) channel action
in vascular tone regulation: from genetics to diseases. Sheng
Li Xue Bao (2012) 64 113.
111 Seino S., Miki T. Physiological and pathophysiological roles
of ATP-sensitive K+channels. Prog. Biophys. Mol. Biol.
(2003) 81 133176.
112 Standen N.B. KATP channels in vascular smooth muscle:
structure, regulation and functional roles. J. Clin. Basic
Cardiol. (2003) 6714.
113 Moreau R., Komeichi H., Kirstetter P. et al. Effects of
glibenclamide on systemic and splanchnic haemodynamics
in conscious rats. Br. J. Pharmacol. (1994) 112 649653.
ª2019 Soci
et
e Franc
ßaise de Pharmacologie et de Th
erapeutique
Fundamental & Clinical Pharmacology
16 M.F. Dogan et al.
114 Yildiz O. Vasodilating mechanisms of levosimendan:
involvement of K+channels. J. Pharmacol. Sci. (2007) 104
15.
115 Tajada S., Cidad P., Moreno-Domınguez A., Perez-Garcıa
M.T., L
opez-L
opez J.R. High blood pressure associates with
the remodelling of inward rectifier K+channels in mice
mesenteric vascular smooth muscle cells. J. Physiol. (2012)
23 60756091.
116 Zaritsky J.J., Eckman D.M., Wellman G.C., Nelson M.T.,
Schwarz T.L. Targeted disruption of Kir2.1 and Kir2.2 genes
reveals the essential role of the inwardly rectifying K+
current in K +-mediated vasodilation. Circ. Res. (2000) 87
160167.
117 Gonz
alez C., Baez-Nieto D., Valencia I. et al. K+channels:
function-structural overview. Compr. Physiol. (2012) 2
20872149.
118 Pint
erov
a M., Kune
s J., Zicha J. Altered neural and vascular
mechanisms in hypertension. Physiol. Res. (2011) 60 381
402.
119 Coetzee W.A., Amarillo Y., Chiu J. et al. Molecular diversity
of K+channels. Ann. N. Y. Acad. Sci. (1999) 868 233
255.
120 Pandit L.M., Lloyd E.E., Reynolds J.O. et al. TWIK-2 channel
deficiency leads to pulmonary hypertension through a Rho-
kinase mediated process. Hypertension (2015) 64 1260
1265.
121 Lloyd E.E., Crossland R.F., Phillips S.C. et al. Disruption of
K2P 6.1 produces vascular dysfunction and hypertension in
mice. Hypertension (2010) 58 672678.
122 Gardener M.J., Johnson I.T., Burnham M.P., Edwards G.,
Heagerty A.M., Weston A.H. Functional evidence of a role for
two-pore domain potassium channels in rat mesenteric and
pulmonary arteries. Br. J. Pharmacol. (2004) 142 192202.
123 Gurney A.M., Osipenko O.N., MacMillan D., McFarlane K.M.,
Tate R.J., Kempsill F.E.J. Two-pore domain K channel,
TASK-1, in pulmonary artery smooth muscle cells. Circ. Res.
(2003) 93 957964.
124 Pokojski S., Busch C., Grgic I. et al. TWIK-related two-pore
domain potassium channel TREK-1 in carotid endothelium
of normotensive and hypertensive mice. Cardiovasc. Res.
(2008) 79 8088.
125 Muhammad S., Aller M.I., Maser-Gluth C., Schwaninger M.,
Wisden W. Expression of the kcnk3 potassium channel gene
lessens the injury from cerebral ischemia, most likely by a
general influence on blood pressure. Neuroscience (2010)
167 758764.
126 Nielsen G., Wandall-Frostholm C., Sadda V. et al. Alterations
of N-3 polyunsaturated fatty acid-activated K2P channels in
hypoxia-induced pulmonary hypertension. Basic Clin.
Pharmacol. Toxicol. (2013) 113 250258.
127 Feliciangeli S., Chatelain F.C., Bichet D., Lesage F. The
family of K2P channels: salient structural and functional
properties. J. Physiol. (2015) 593 25872603.
128 Olschewski A., Veale E.L., Nagy B.M. et al. TASK-1 (KCNK3)
channels in the lung: from cell biology to clinical
implications. Pulm. Circ. Physiopathol. (2017) 50 1700754.
129 Dias F.M.V., Ribeiro R.F., Fernandes A.A. et al. Na+K+-
ATPase activity and K+channels differently contribute to
vascular relaxation in male and female rats. PLoS ONE
(2014) 918.
130 Mondejar-Parre~
no G., Callejo M., Barreira B. et al. miR-1 is
increased in pulmonary hypertension and downregulates
Kv1.5 channels in rat pulmonary arteries. J. Physiol. (2019)
597 11851197
131 Sedivy V., Joshi S., Ghaly Y. et al. Role of Kv7
channels in responses of the pulmonary circulation to
hypoxia. Am. J. Physiol. Lung Cell. Mol. Physiol.
(2015) 308 L48L57.
132 Yildiz O., Seyrek M., Un I., Gul H., Candemir G., Yildirim V.
The relationship between risk factors and testosterone-
induced relaxations in human internal mammary artery. J.
Cardiovasc. Pharmacol. (2005) 45 47.
133 Koschak A., Koch R.O., Liu J. et al. [125I] Iberiotoxin-
D19Y/Y36F, the first selective, high specific activity
radioligand for high-conductance calcium-activated
potassium channels. Biochemistry (1997) 36 19431952.
134 Li R.W.S., Yang C., Shan L. et al. Relaxation effect of a novel
Danshensu/tetramethylpyrazine derivative on rat mesenteric
arteries.Eur.J.Pharmacol.(2015)761 153160.
135 Yan J., Chen R., Liu P., Gu Y. Docosahexaenoic acid
attenuates hypoxic pulmonary vasoconstriction by
activating the large conductance Ca2 +-activated K+
currents in pulmonary artery smooth muscle cells. Pulm.
Pharmacol. Ther. (2014) 28 916.
136 K
ohler R., Kaistha B.P., Wulff H. Vascular KCa-channels as
therapeutic targets in hypertension and restenosis disease.
Expert Opin. Ther. Targets (2010) 14 143155.
137 Lawson K. Potassium channel openers as potential
therapeutic weapons in ion channel disease. Kidney Int.
(2000) 57 838845.
138 Morales-Cano D., Moreno L., Barreira B. et al. Kv7 channels
critically determine coronary artery reactivity: left-right
differences and down-regulation by hyperglycaemia.
Cardiovasc. Res. (2015) 106 98108.
139 Antigny F., Hautefort A., Meloche J. et al. Potassium
channel subfamily K member 3 (KCNK3) contributes to the
development of pulmonary arterial hypertension. Circulation
(2016) 133 13711385.
140 Guan Y., Li N., Tian Y.M. et al. Chronic intermittent
hypobaric hypoxia antagonizes renal vascular hypertension
by enhancement of vasorelaxation via activating BKCa. Life
Sci. (2016) 157 7481.
141 Cheng J., Mao L., Wen J. et al. Different effects of
hypertension and age on the function of large conductance
calcium- and voltage-activated potassium channels in
human mesentery artery smooth muscle cells. J. Am. Heart
Assoc. (2016) 5111.
142 Leo M.D., Zhai X., Yin W., Jaggar J.H. Impaired trafficking of
b1 subunits inhibits BK channels in cerebral arteries of
hypertensive rats. Hypertension (2018) 72 765775.
143 Seo E.Y., Kim H.J., Zhao Z.H. et al. Low K+current in
arterial myocytes with impaired K +-vasodilation and its
ª2019 Soci
et
e Franc
ßaise de Pharmacologie et de Th
erapeutique
Fundamental & Clinical Pharmacology
Pharmacological perspective of potassium channels 17
recovery by exercise in hypertensive rats. Pfl
ugers Arch. J.
Physiol. (2014) 466 21012111.
144 Cox R.H., Lozinskaya I., Dietz N.J. Calcium exerts a larger
regulatory effect on potassium+channels in small
mesenteric artery myocytes from spontaneously hypertensive
rats compared to Wistar-Kyoto Rats Robert. Am. J.
Hypertens. (2003) 16 2127.
145 Amberg G., Bonev A., Rossow C.F., Nelson M.T., Santana
L.F.. Modulation of the molecular composition of large
conductance, Ca
2+
activated K+channels in vascular
smooth muscle during hypertension. J. Clin. Invest. (2003)
112 717724.
146 Fern
andez-Fern
andez J.M., Tom
as M., V
azquez E. et al. Gain-
of-function mutation in the KCNMB1 potassium channel
subunit is associated with low prevalence of diastolic
hypertension. J. Clin. Invest. (2004) 113 10321039.
147 Liu Y., Pleyte K., Knaus H.G., Rusch N.J. Increased
expression of Ca2 +-sensitive K+channels in aorta of
hypertensive rats. Hypertension (1997) 30 14031409.
148 Kolias T.J., Chai S., Webb R.C. Potassium channel
antagonists and vascular reactivity in stroke-prone
spontaneously hypertensive rats. Am. J. Hypertens. (1993) 6
528533.
149 Kitazono T., Heistad D.D., Faraci F.M. ATP-sensitive
potassium channels in the basilar artery during chronic
hypertension. Hypertension (1993) 22 677681.
150 Lee H., Kim K.C., Hong Y.M. Change of voltage-gated
potassium channel 1.7 expressions in monocrotaline-
induced pulmonary arterial hypertension rat model. Korean
J. Pediatr. (2018) 61 271278.
151 Yuan J.X., Aldinger A.M., Juhaszova M. et al. Dysfunctional
voltage-gated K+channels in pulmonary artery smooth
muscle cells of patients with primary pulmonary
hypertension. Circulation (1998) 98 14001407.
152 Tang B., Li Y., Nagaraj C. et al. Endothelin-1 inhibits
background two-pore domain channel TASK-1 in primary
human pulmonary artery smooth muscle cells. Am. J.
Respir. Cell Mol. Biol. (2009) 41 476483.
153 Ke Q., Wu J., Tian L., Li W., Du Y.-M. Role of voltage-gated
potassium channels in pathogenesis of chronic pulmonary
heart disease. J. Huazhong Univ. Sci. Technol. (2013) 33
644649.
154 Son Y., Chun W., Ahn Y.T. et al. 7-Ketocholesterol induces
the reduction of KCNMB1 in atherosclerotic blood vessels.
Biochem. Biophys. Res. Commun. (2015) 457 324327.
155 Wiecha J., Schlager B., Voisard R., Hannekum A., Mattfeldt
T., Hombach V. Ca(+2)-activated K+channels in human
smooth muscle cells of coronary atherosclerotic plaques and
coronary media segments. Basic Res. Cardiol. (1997) 92
233239.
156 Faraci F.M., Orgren K., Heistad D.D. Impaired relaxation of
the carotid artery during activation of ATP-sensitive
potassium channels in atherosclerotic monkeys. J. Stroke
(1994) 25 178182.
157 Jiang J., Thor
en P., Caligiuri G., Hansson G.K., Pernow J.
Enhanced phenylephrine-induced rhythmic activity in the
atherosclerotic mouse aorta via an increase in opening of
KCa channels: relation to Kv channels and nitric oxide. Br.
J. Pharmacol. (1999) 128 637646.
158 Rozsa Z., Pataricza J., Nemeth J., Papp J.G. Differential
efficacy of vasodilators in hypercholesterolaemic rabbits. J.
Pharm. Pharmacol. (1998) 50 10351044.
159 Ghanam K., Lavagna C., Burgaud J.-L., Javellaud J., Oudart
N. Involvement of potassium channels in the protective
effect of 17b-estradiol on hypercholesterolemic rabbit carotid
artery. Atherosclerosis (2000) 152 5967.
160 Lee C.H., Dai Z.K., Yen C.T., Hsieh S.L., Wu B.N. KMUP-1
protects against streptozotocin-induced mesenteric artery
dysfunction via activation of ATP-sensitive potassium
channels. Pharmacol. Reports (2018) 70 746752.
161 Hong D.H., Li H., Kim H.W. et al. Alterations of voltage-
dependent K+channels in the mesenteric artery during the
early and chronic phases of diabetes. Clin. Exp. Pharmacol.
Physiol. (2016) 43 808817.
162 Wang Y., Zhang H.-T., Su X.-L. et al. Experimental diabetes
mellitus down-regulates large-conductance Ca2 +-activated K+
channels in cerebral artery smooth muscle and alters functional
conductance. Curr. Neurovasc. Res. (2010) 77584.
163 Li H., Shin S.E., Seo M.S. et al. Alterations of ATP-sensitive
K+channels in human umbilical arterial smooth muscle
during gestational diabetes mellitus. Pfl
ugers Arch. - Eur. J.
Physiol. (2018) 470 13251333.
164 Yildiz O. Vascular smooth muscle and endothelial
functions in aging. Ann. N. Y. Acad. Sci. (2007) 1100
353360.
165 Fransen P., Van Hove C.E., Leloup A.J.A., Schrijvers D.M.,
De Meyer G.R.Y., De Keulenaer G.W. Effect of angiotensin II-
induced arterial hypertension on the voltage-dependent
contractions of mouse arteries. Pflugers Arch. Eur. J.
Physiol. (2016) 468 257267.
166 Cox R.H., Lozinskaya I.M. Augmented calcium currents
in mesenteric artery branches of the spontaneously
hypertensive rat. Hypertension (1995) 26 10601064.
167 Wellman G.C., Cartin L., Eckman D.M. et al. Membrane
depolarization, elevated Ca(2 +) entry, and gene expression
in cerebral arteries of hypertensive rats. Am. J. Physiol.
Hear. Circ. Physiol. (2001) 281 H2559H2567.
168 Harder D.R., Smeda J., Lombard J. Enhanced myogenic
depolarization in hypertensive cerebral arterial muscle. Circ.
Res. (1985) 57 319322.
169 Sonkusare S., Palade P.T., Marsh J.D., Telemaque S., Pesic
A., Rusch N.J. Vascular calcium channels and high blood
pressure: pathophysiology and therapeutic implications.
Vasc. Pharmacol. (2006) 44 131142.
170 Hald B.O., Jacobsen J.C.B., Braunstein T.H. et al. BKCa and
KV channels limit conducted vasomotor responses in rat
mesenteric terminal arterioles. Pflugers Arch. Eur. J. Physiol.
(2012) 463 279295.
171 Nelson M.T., Bonev A.D. The b1 subunit of the Ca 2 +-
sensitive K +channel protects against hypertension. J. Clin.
Invest. (2004) 113 955957.
ª2019 Soci
et
e Franc
ßaise de Pharmacologie et de Th
erapeutique
Fundamental & Clinical Pharmacology
18 M.F. Dogan et al.
172 Brenner R., Perz G.J., Bonev A.D. et al. Vasoregulation by
the b1 subunit of the calcium-activated potassium channel.
Nature (2000) 407 870876.
173 Pl
uger S., Faulhaber J., F
urstenau M. et al. Mice with
disrupted BK channel b1 subunit gene feature abnormal
Ca +2 Spark/STOC coupling and elevated blood pressure.
Circ. Res. (2000) 87 E53E60.
174 Park W.S., Kim N., Youm J.B. et al. Angiotensin II inhibits
inward rectifier K+channels in rabbit coronary arterial
smooth muscle cells through protein kinase Ca. Biochem.
Biophys. Res. Commun. (2006) 341 728735.
175 Bonnet S., Archer S.L. Potassium channel diversity in the
pulmonary arteries and pulmonary veins: implications for
regulation of the pulmonary vasculature in health and
during pulmonary hypertension. Pharmacol. Ther. (2007)
115 5669.
176 Jepps T.A., Chadha P.S., Davis A.J. et al. Downregulation of
Kv7.4 channel activity in primary and secondary
hypertension. Circulation (2011) 124 602611.
177 Archer S.L., Wu X.C., Th
ebaud B. et al. Preferential
expression and function of voltage-gated, O 2-sensitive K+
channels in resistance pulmonary arteries explains regional
heterogeneity in hypoxic pulmonary vasoconstriction: ionic
diversity in smooth muscle cells. Circ. Res. (2004) 95 308
318.
178 Sommer N., Dietrich A., Schermuly R.T. et al. Regulation of
hypoxic pulmonary vasoconstriction: basic mechanisms. Eur.
Respir. J. (2008) 32 16391651.
179 Bonnet S., Dumas-de-La-Roque E., Begueret H. et al.
Dehydroepiandrosterone (DHEA) prevents and reverses
chronic hypoxic pulmonary hypertension. Proc. Natl Acad.
Sci. (2003) 100 94889493.
180 Fan Z., Liu B., Zhang S. et al. YM155, a selective survivin
inhibitor, reverses chronic hypoxic pulmonary hypertension
in rats via upregulating voltage-gated potassium channels.
Clin. Exp. Hypertens. (2015) 37 381387.
181 Bonnet S., Michelakis E.D., Porter C.J. et al. An abnormal
mitochondrial-hypoxia inducible factor-1a-Kv channel
pathway disrupts oxygen sensing and triggers pulmonary
arterial hypertension in fawn hooded rats: similarities to
human pulmonary arterial hypertension. Circulation (2006)
113 26302641.
182 Pousada G., Baloira A., Vilari~
no C., Cifrian J.M., Valverde
D., Ahmad S. Novel mutations in BMPR2, ACVRL1 and
KCNA5 genes and hemodynamic parameters in patients
with pulmonary arterial hypertension. PLoS ONE (2014) 9
110.
183 Dessouroux A., Akwa Y., Baulieu E.E. DHEA decreases HIF-
1aaccumulation under hypoxia in human pulmonary
artery cells: potential role in the treatment of pulmonary
arterial hypertension. J. Steroid Biochem. Mol. Biol. (2008)
109 8189.
184 Park W.S., Firth A.L., Han J., Ko E.A. Patho-, physiological
roles of voltage-dependent K+channels in pulmonary
arterial smooth muscle cells. J. Smooth Muscle Res. (2010)
46 89105.
185 Pozeg Z.I., Michelakis E.D., McMurtry S. et al. In vivo gene
transfer of the O2-sensitive potassium channel Kv1.5
reduces pulmonary hypertension and restores hypoxic
pulmonary vasoconstriction in chronically hypoxic rats.
Circulation (2003) 107 20372044.
186 Firth A.L., Remillard C.V., Platoshyn O., Fantozzi I., Ko E.A.,
Yuan J.X.Y. Functional ion channels in human pulmonary
artery smooth muscle cells: voltage-dependent cation
channels. Pulm. Circ. (2011) 14871.
187 Mandegar M., Yuan J.X.J. Role of K+channels in pulmonary
hypertension. Vascul. Pharmacol. (2002) 38 2533.
188 Wiedmann F., Schmidt C., Lugenbiel P. et al. Therapeutic
targeting of two-pore-domain potassium (K2P) channels in
the cardiovascular system. Clin. Sci. (2016) 130 643
650.
189 Kitagawa M.G., Reynolds J.O., Wehrens X.H.T., Bryan R.M.,
Pandit L.M. Hemodynamic and pathologic characterization
of the TASK-1/mouse does not demonstrate pulmonary
hypertension. Front. Med. (2017) 417.
190 Ma L., Roman-Campos D., Austin E.D. et al. A novel
channelopathy in pulmonary arterial hypertension. N. Engl.
J. Med. (2013) 369 351361.
191 Higasa K., Ogawa A., Terao C. et al. A burden of rare
variants in BMPR2 and KCNK3 contributes to a risk of
familial pulmonary arterial hypertension. BMC Pulm. Med.
(2017) 17 57.
192 Best D.H., Sumner K.L., Smith B.P. et al. EIF2AK4
mutations in patients diagnosed with pulmonary arterial
hypertension. Chest (2017) 151 821828.
193 Cunningham K.P., Holden R.G., Escribano-Subias P.M.,
Cogolludo A., Veale E.L., Mathie A. Characterisation and
regulation of wild type and mutant TASK-1 two pore
domain potassium channels indicated in pulmonary
arterial hypertension. J. Physiol. (2019) 597, 1087
1101.
194 Bohnen M.S., Roman-Campos D., Terrenoire C. et al. The
impact of heterozygous KCNK3 mutations associated with
pulmonary arterial hypertension on channel function and
pharmacological recovery. J. Am. Heart Assoc. (2017) 6
e006465. https://doi.org/10.1161/JAHA.117.006465
195 Murtaza G., Mermer P., Goldenberg A. et al. TASK-1
potassium channel is not critically involved in mediating
hypoxic pulmonary vasoconstriction of murine intra-
pulmonary arteries. PLoS ONE (2017) 12 118.
196 Manoury B., Lamalle C., Oliveira R., Reid J., Gurney A.M.
Contractile and electrophysiological properties of pulmonary
artery smooth muscle are not altered in TASK-1 knockout
mice. J. Physiol. (2011) 589 32313246.
197 Chaudhary K.R., Stewart D.J. Taking the right ventricle
to ‘task’ in pulmonary hypertension: role of TASK1/
KCNK3 in RV dysfunction. Cardiovasc. Res. (2018) 114
776778.
198 Lambert M., Boet A., Rucker-Martin C. et al. Loss of KCNK3
is a hallmark of RV hypertrophy/dysfunction associated with
pulmonary hypertension. Cardiovasc. Res. (2018) 114 880
893.
ª2019 Soci
et
e Franc
ßaise de Pharmacologie et de Th
erapeutique
Fundamental & Clinical Pharmacology
Pharmacological perspective of potassium channels 19
199 Lambert M., Capuano V., Olschewski A. et al. Ion channels
in pulmonary hypertension: a therapeutic interest? Int. J.
Mol. Sci. (2018) 19 3162.
200 Bohnen M.S., Ma L., Zhu N. et al. Loss-of-function ABCC8
mutations in pulmonary arterial hypertension. Circ.
Genomic Precis. Med. (2018) 11 e002087.
201 Jahangir A., Terzic A. KATP channel therapeutics at the
bedside. J. Mol. Cell. Cardiol. (2005) 39 99112.
202 Ren Y.J., Xu X.H., Zhong C.B., Feng N., Wang X.L.
Hypercholesterolemia alters vascular functions and
gene expression of potassium channels in rat aortic
smooth muscle cells. Acta Pharmacol. Sin. (2001) 22
274278.
203 Mach F., Steffens S. The role of the endocannabinoid
system in atherosclerosis. J. Neuroendocrinol. (2008) 20
5357.
204 Lee S.M., Nguyen D., Hu Z., Abbott G.W. Kcne2 deletion
promotes atherosclerosis and diet-dependent sudden death.
J. Mol. Cell. Cardiol. (2015) 87 148151.
205 Heaps C.L., Tharp D.L., Bowles D.K. Hypercholesterolemia
abolishes voltage-dependent K+channel contribution to
adenosine-mediated relaxation in porcine coronary
arterioles. Am. J. Physiol. Heart Circ. Physiol. (2005) 288
H568H576.
206 Lee T., Lin M., Chou T., Tsai C. et al. Effect of pravastatin on
left ventricular mass by activation of myocardial KATP
channels in hypercholesterolemic rabbits. Atherosclerosis
(2004) 176 273278.
207 Jeremy R.W., McCarron H. Effect of hypercholesterolemia on
Ca(2 +)-dependent K(+) channel-mediated vasodilatation
in vivo. Am. J. Physiol. Heart Circ. Physiol. (2000) 279
H1600H1608.
208 Dopico A.M., Bukiya A.N., Singh A.K. Large conductance,
calcium- and voltage-gated potassium (BK) channels:
regulation by cholesterol. Pharmacol. Ther. (2012) 135
133150.
209 Bukiya A.N., Vaithianathan T., Kuntamallappanavar G.,
Asuncion-Chin M., Dopico A.M. Smooth muscle cholesterol
enables BK-b1 subunit-mediated channel inhibition and
subsequent vasoconstriction evoked by alcohol. Arterioscler.
Thromb. Vasc. Biol. (2011) 31 24102423.
210 Mathew V., Lerman A. Altered effects of potassium channel
modulation in the coronary circulation in experimental
hypercholesterolemia. Atherosclerosis (2001) 154 329335.
211 Chester A.H., O’Neil G.S., Tadjkarimi S., Yacoub M.H.,
Moncada S. Low basal and stimulated release of nitric oxide
in atherosclerotic epicardial coronary arteries. Lancet J.
(1990) 336 897900.
212 Jackson W.F. Potassium channels in the peripheral
microcirculation. Microcirc. J. (2005) 12 113127.
213 Tang X., Qian L.L., Wang R.X. et al. Regulation of coronary
arterial large conductance Ca2 +-activated K+channel protein
expression and function by n-3 polyunsaturated fatty acids in
diabetic rats. J. Vasc. Res. (2017) 54 329343.
214 Cox R.H. Molecular determinants of voltage-gated potassium
currents in vascular smooth muscle. Cell Biochem. Biophys.
(2005) 42 167195.
215 Khaybullina D., Patel A., Zerilli T. Riociguat (adempas): a
novel agent for the treatment of pulmonary arterial
hypertension and chronic thromboembolic pulmonary
hypertension. P T (2014) 39 749758.
216 Tarkin J.M., Kaski J.C. Nicorandil and long-acting nitrates:
vasodilator therapies for the management of chronic stable
angina pectoris. Eur. Cardiol. (2018) 13 2328.
217 Dumas de la Roque E., Savineau J.P., Bonnet S.
Dehydroepiandrosterone: a new treatment for vascular
remodeling diseases including pulmonary arterial
hypertension. Pharmacol. Ther. (2010) 126 186199.
218 Pan Z., Huang J., Cui W., Long C., Zhang Y., Wang H.
Targeting hypertension with a new adenosine triphosphate
sensitive potassium channel opener iptakalim. J. Cardiovasc.
Pharmacol. (2010) 56 215228.
219 McMurtry M.S., Bonnet S., Wu X. et al. Dichloroacetate
prevents and reverses pulmonary hypertension by inducing
pulmonary artery smooth muscle cell apoptosis. Circ. Res.
(2004) 95 830840.
220 Suzuki S., Yano K., Kusano S., Hashimoto T.
Antihypertensive effect of levcromakalim in patients with
essential hypertension. Study by 24-h ambulatory blood
pressure monitoring. Arzneimittelforschung (1995) 45 859
864.
221 Bano M., Barot K., Ahmed S., Nikolova S., Ivanov I., Ghate
M. Benzopyran derivatives as cardio-selective ATP-sensitive
potassium channel openers: a review. Mini-Reviews Med.
Chem. (2013) 13 17441760.
222 Jackson W.F. Potassium channels in regulation of vascular
smooth muscle contraction and growth. Adv. Pharmacol.
(2017) 78 89144.
223 Jackson W.F. KV channels and the regulation of vascular
smooth muscle tone. Microcirculation (2018) 25 e12421.
224 Dopico A.M., Bukiya A.N., Jaggar J.H. Calcium- and voltage-
gated BK channels in vascular smooth muscle. Pflugers
Arch. Eur. J. Physiol. (2018) 470 12711289.
225 Szeto V., Chen N.-H., Sun H.-S., Feng Z.-P. The role of KATP
channels in cerebral ischemic stroke and diabetes. Acta
Pharmacol. Sin. (2018) 39 683694.
226 Severino P., D’Amato A., Netti L. et al. Diabetes mellitus and
ischemic heart disease: the role of ion channels. Int. J. Mol.
Sci. (2018) 10 E802.
227 Zhu Y., Ye P., Chen S., Zhang D.-M. Functional
regulation of large conductance Ca 2 +-activated K+
channels in vascular diseases. Metab., Clin. Exp. (2018)
83 7580.
228 Barrese V., Stott J.B., Greenwood I.A. KCNQ-encoded
potassium channels as therapeutic targets. Annu. Rev.
Pharmacol. Toxicol. (2018) 58 625648.
229 Fosmo A.L., Skraastad Ø.B. The Kv7 channel and cardiovascular
risk factors. Front. Cardiovasc. Med. (2017) 475.
ª2019 Soci
et
e Franc
ßaise de Pharmacologie et de Th
erapeutique
Fundamental & Clinical Pharmacology
20 M.F. Dogan et al.
... The extract was evaluated on acetylcholine-contracted rat tracheal rings in the presence of glibenclamide, which inhibits the ATP-sensitive potassium channels, to determine if the extract's bronchorelaxant process is related to these channels. In fact, K + channel activation generated membrane hyperpolarization, which led to dilation, while K+ channel inhibition resulted in membrane depolarization, which led to constriction [48,49]. The relaxing impact of HMAE was stronger in the presence of glibenclamide than it was in its absence. ...
... Because the uterine arteries of pregnant rats showed greater vascular relaxation than those of nonpregnant rats, we investigated the mechanisms of H 2 S relaxation in these arterial rings. A high K + concentration (80 mM), which inhibits the opening of K channels involved in smooth muscle relaxation [27], completely blocked the relaxation response to NaHS and shifted the H 2 S response to slight vasoconstriction ( Figure 2). ...
Article
Full-text available
Hydrogen sulfide (H2S) is a cardiovascular signaling molecule that causes vasodilation in vascular smooth muscle cells, but its mechanism is unclear. We examined how H2S affects mesenteric and uterine arteries without endothelium in nonpregnant and pregnant rats and the underlying mechanisms. H2S donors GYY4137 and NaHS relaxed uterine arteries more than mesenteric arteries in both pregnant and nonpregnant rats. GYY4137 and NaHS caused greater relaxation in the uterine artery of pregnant versus nonpregnant rats. High extracellular K⁺ abolished NaHS relaxation in pregnant uterine arteries, indicating potassium channel involvement. NaHS relaxation was unaffected by voltage-gated potassium channel blockers, reduced by ATP-sensitive potassium channel blockers, and abolished by calcium-activated potassium (BKCa) channel blockers. Thiol-reductant dithiothreitol also prevented NaHS relaxation. Thus, H2S has region-specific and pregnancy-enhanced vasodilator effects in the uterine arteries, mainly mediated by BKCa channels and sulfhydration.
Article
Piezo are mechanosensitive non‐selective cation channels that are suggested to be involved in vascular development and function. The aim of our study was to determine any sex‐specific contributions of the mechanosensitive Piezo 1 channels on blood vessel wall stiffness. Composite Young modulus (CYM) was determined using pressure myograph experimental approach using third‐order mesenteric arteries (intact and denuded) from Dahl salt‐sensitive male and female normotensive and hypertensive rats (n = 6–8). The effects of Piezo 1 agonist (Yoda 1; 10 μM), and antagonist (GsMTx‐4; 2 μM) were studied in intact and denuded vessels. The distribution of Piezo 1 was identified using immunohistochemistry. In intact blood vessels, there were no differences in CYM between the experimental groups, however, removal of the endothelium unmasked significant increases in CYM in normotensive males and female groups compared to hypertensive males. The presence of Yoda 1 did not affect CYM in any groups. In the intact tissues, GsMTx‐4 led to significant increases in CYM in hypertensive females, and normotensive males and females, but not in hypertensive males. In the denuded vessels, GsMTx‐4, produced a significant increase in CYM but only in the female normotensives. Differential expression of Piezo 1 were found in male versus female blood vessels. Our findings support a greater contribution of Piezo 1 mechanoceptors to vascular biomechanics of male hypertensive compared to male normotensive or female groups. The evidence also points to a possible differential vasoregulatory role for Piezo 1 in endothelial versus vascular smooth cells, with a greater contribution in males than females.
Article
Background The endocannabinoid system plays important roles in various systems, including the genitourinary system; however, its mechanism of action is not fully understood. Objectives This study aimed to investigate the direct relaxant effects of anandamide and its possible mechanisms in isolated rat bladder and vas deferens tissues. Methods Twenty‐one adult male Wistar albino rats were used. Bladder and vas deferens (prostatic and epididymal portions) tissues were mounted in 10 mL of organ baths. Relaxation responses to anandamide were recorded at 3 and 10 μM concentrations. After the rest period, the procedures were repeated in the presence of cannabinoid (CB) and vanilloid receptor antagonists, various potassium channel blockers, cyclo‐oxygenase, and nitric oxide synthase inhibitors. In different tissues to investigate the Ca ²⁺ ‐channel antagonistic effect of anandamide, concentration–response curves to CaCl 2 were obtained in the absence and presence of anandamide. Results Anandamide caused a significant relaxation response in the bladder and epididymal vas deferens tissues, but not in the prostatic portion. The effect of anandamide was antagonized in the presence of the CB 1 antagonist AM251 or the non‐selective potassium channel blocker tetraethylammonium in bladder tissue. In the epididymal vas deferens, anandamide significantly inhibited the calcium contraction responses, especially at high concentrations. The CB 2 antagonist AM630 reversed this inhibition. Conclusions The results show that anandamide has a direct relaxant effect on the isolated rat bladder and epididymal vas deferens. Anandamide triggers different mechanisms in different types of tissues, and further studies are needed to elucidate the mechanism of action of anandamide.
Article
Studies have examined the therapeutic effect of levosimendan on cardiovascular diseases such as heart failure, perioperative cardiac surgery, and septic shock, but the specific mechanism in mice remains largely unknown. This study aimed to investigate the relaxation mechanism of levosimendan in the thoracic aorta smooth muscle of mice. Levosimendan-induced relaxation of isolated thoracic aortic rings that were precontracted with norepinephrine (NE) or KCl was recorded in an endothelium-independent manner. Vasodilatation by levosimendan was not associated with the production of the endothelial relaxation factors NO and PGI 2 . The voltage-dependent K ⁺ channel (K V ) blocker (4-aminopyridine) and selective K Ca blocker (tetraethylammonium) had no effect on thoracic aortas treated with levosimendan, indicating that K V and K Ca channels may not be involved in the levosimendan-induced relaxation mechanism. Although the inwardly rectifying K ⁺ channel (K ir ) blocker (barium chloride) and the K ATP channel blocker (glibenclamide) significantly inhibited levosimendan-induced vasodilation in the isolated thoracic aorta, barium chloride had a much stronger inhibitory effect on levosimendan-induced vasodilation than glibenclamide, suggesting that levosimendan-induced vasodilation may be mediated by K ir channels. The vasodilation effect and expression of K ir 2.1 induced by levosimendan were further enhanced by the PKC inhibitor staurosporine. Extracellular calcium influx was inhibited by levosimendan without affecting intracellular Ca ²⁺ levels in the isolated thoracic aorta. These results suggest that K ir channels play a more important role than K ATP channels in regulating vascular tone in larger arteries and that the activity of the K ir channel is enhanced by the PKC pathway.
Article
Full-text available
Background Pulmonary arterial hypertension (PAH), Group 1 pulmonary hypertension (PH), is a type of pulmonary vascular disease characterized by abnormal contraction and remodeling of the pulmonary arterioles, manifested by pulmonary vascular resistance (PVR) and increased pulmonary arterial pressure, eventually leading to right heart failure or even death. The mechanisms involved in this process include inflammation, vascular matrix remodeling, endothelial cell apoptosis and proliferation, vasoconstriction, vascular smooth muscle cell proliferation and hypertrophy. In this study, we review the mechanisms of action of prostaglandins and their receptors in PAH. Main body PAH-targeted therapies, such as endothelin receptor antagonists, phosphodiesterase type 5 inhibitors, activators of soluble guanylate cyclase, prostacyclin, and prostacyclin analogs, improve PVR, mean pulmonary arterial pressure, and the six-minute walk distance, cardiac output and exercise capacity and are licensed for patients with PAH; however, they have not been shown to reduce mortality. Current treatments for PAH primarily focus on inhibiting excessive pulmonary vasoconstriction, however, vascular remodeling is recalcitrant to currently available therapies. Lung transplantation remains the definitive treatment for patients with PAH. Therefore, it is imperative to identify novel targets for improving pulmonary vascular remodeling in PAH. Studies have confirmed that prostaglandins and their receptors play important roles in the occurrence and development of PAH through vasoconstriction, vascular smooth muscle cell proliferation and migration, inflammation, and extracellular matrix remodeling. Conclusion Prostacyclin and related drugs have been used in the clinical treatment of PAH. Other prostaglandins also have the potential to treat PAH. This review provides ideas for the treatment of PAH and the discovery of new drug targets.
Article
Full-text available
Key points The TASK‐1 channel gene (KCNK3) has been identified as a possible disease‐causing gene in heritable pulmonary arterial hypertension (PAH). In the present study, we show that novel mutated TASK‐1 channels, seen in PAH patients, have a substantially reduced current compared to wild‐type TASK‐1 channels. These mutated TASK‐1 channels are located at the plasma membrane to the same degree as wild‐type TASK‐1 channels. ONO‐RS‐082 and alkaline pH 8.4 both activate TASK‐1 channels but do not recover current through mutant TASK‐1 channels. We show that the guanylate cyclase activator, riociguat, a novel treatment for PAH, enhances current through TASK‐1 channels but does not recover current through mutant TASK‐1 channels. Abstract Pulmonary arterial hypertension (PAH) affects ∼15–50 people per million. KCNK3, the gene that encodes the two pore domain potassium channel TASK‐1 (K2P3.1), has been identified as a possible disease‐causing gene in heritable PAH. Recently, two new mutations have been identified in KCNK3 in PAH patients: G106R and L214R. The present study aimed to characterize the functional properties and regulation of wild‐type (WT) and mutated TASK‐1 channels and determine how these might contribute to PAH and its treatment. Currents through WT and mutated human TASK‐1 channels transiently expressed in tsA201 cells were measured using whole‐cell patch clamp electrophysiology. Localization of fluorescence‐tagged channels was visualized using confocal microscopy and quantified with in‐cell and on‐cell westerns. G106R or L214R mutated channels were located at the plasma membrane to the same degree as WT channels; however, their current was markedly reduced compared to WT TASK‐1 channels. Functional current through these mutated channels could not be restored using activators of WT TASK‐1 channels (pH 8.4, ONO‐RS‐082). The guanylate cyclase activator, riociguat, enhanced current through WT TASK‐1 channels; however, similar to the other activators investigated, riociguat did not have any effect on current through mutated TASK‐1 channels. Thus, novel mutations in TASK‐1 seen in PAH substantially alter the functional properties of these channels. Current through these channels could not be restored by activators of TASK‐1 channels. Riociguat enhancement of current through TASK‐1 channels could contribute to its therapeutic benefit in the treatment of PAH.
Article
Full-text available
Pulmonary arterial hypertension (PAH) is a multifactorial and severe disease without curative therapies. PAH pathobiology involves altered pulmonary arterial tone, endothelial dysfunction, distal pulmonary vessel remodeling, and inflammation, which could all depend on ion channel activities (K+, Ca2+, Na+ and Cl−). This review focuses on ion channels in the pulmonary vasculature and discusses their pathophysiological contribution to PAH as well as their therapeutic potential in PAH.
Article
Full-text available
Purpose: Abnormal potassium channels expression affects vessel function, including vascular tone and proliferation rate. Diverse potassium channels, including voltage-gated potassium (Kv) channels, are involved in pathological changes of pulmonary arterial hypertension (PAH). Since the role of the Kv1.7 channel in PAH has not been previously studied, we investigated whether Kv1.7 channel expression changes in the lung tissue of a monocrotaline (MCT)-induced PAH rat model and whether this change is influenced by the endothelin (ET)-1 and reactive oxygen species (ROS) pathways. Methods: Rats were separated into 2 groups: the control (C) group and the MCT (M) group (60 mg/kg MCT). A hemodynamic study was performed by catheterization into the external jugular vein to estimate the right ventricular pressure (RVP), and pathological changes in the lung tissue were investigated. Changes in protein and mRNA levels were confirmed by western blot and polymerase chain reaction analysis, respectively. Results: MCT caused increased RVP, medial wall thickening of the pulmonary arterioles, and increased expression level of ET-1, ET receptor A, and NADPH oxidase (NOX) 4 proteins. Decreased Kv1.7 channel expression was detected in the lung tissue. Inward-rectifier channel 6.1 expression in the lung tissue also increased. We confirmed that ET-1 increased NOX4 level and decreased glutathione peroxidase-1 level in pulmonary artery smooth muscle cells (PASMCs). ET-1 increased ROS level in PASMCs. Conclusion: Decreased Kv1.7 channel expression might be caused by the ET-1 and ROS pathways and contributes to MCT-induced PAH.
Article
Full-text available
The present study aimed to investigate the role of propofol in mediating the vasomotor activity of the mesenteric arteriole (MA) of Sprague Dawley (SD) rats, and to elucidate the underlying mechanisms. The pressure myograph technique was used to examine the effect of different concentrations of propofol on the relaxation of blood vessels in the 2-3 mm MA segments freshly separated from the SD rats. The whole-cell patch-clamp technique was applied to observe the outward current of single vascular smooth muscle cells (VSMCs) obtained from the MAs of the SD rats. Furthermore, immunofluorescence was utilized to assess the expression of connexin (Cx) in the MAs of SD rats. The results indicated the following: i) Propofol relaxed the MA of SD rats in a concentration-dependent manner from 1x10⁻⁷ to 3x10⁻⁴ mol/l; ii) in the acutely dissociated VSMCs, propofol (1x10⁻⁷ to 3x10⁻⁴ mol/l) enhanced the outward current of VSMCs in a concentration-dependent manner; iii) the enhanced outward currents induced by propofol (1x10⁻⁵ mol/l) may be reversed by tetraethylammonium (TEA; 1 mmol/l), a calcium-activated K⁺ channel inhibitor; iv) the effect of propofol on the relaxation of the vasculature wAS reduced after perfusion with 1 mmol/l TEA; v) Cx40, Cx43 and Cx45 were expressed on the MA; 6) 18β-glycyrrhetintic acid and 2-aminoethoxydiphenyl borate, two types of gap junction blocker, inhibited the propofol-induced relaxation. The present study provides evidence that propofol relaxes the MA, which may be associated with its effect of enhancing the channel current of large-conductance calcium voltage-activated potassium channels, contributing to the K⁺ outflow and leading to VSMC hyperpolarization; the gap junction may facilitate the hyperpolarization, which may lead to vascular synchronized relaxation and thereby reduce the blood pressure.
Article
Full-text available
We investigated the alterations of ATP-sensitive K⁺ (KATP) channels in human umbilical arterial smooth muscle cells during gestational diabetes mellitus (GDM). The amplitude of the KATP current induced by application of the KATP channel opener pinacidil (10 μM) was reduced in the GDM group than in the control group. Pinacidil-induced vasorelaxation was also predominant in the normal group compared with the GDM group. Reverse transcription polymerase chain reaction and Western blot analysis suggested that the expression of KATP channel subunits such as Kir6.1, Kir6.2, and SUR2B were decreased in the GDM group relative to the normal group. The application of forskolin and adenosine, which activates protein kinase A (PKA) and thereby KATP channels, elicited KATP current in both the normal and GDM groups. However, the current amplitudes were not different between the normal and GDM groups. In addition, the expression levels of PKA subunits were not altered between the two groups. These results suggest that the reduction of KATP current and KATP channel-induced vasorelaxation are due to the decreased expression of KATP channels, not to the impairment of KATP-related signaling pathways.
Article
Objectives: The aim of this study was to investigate the vascular effects and mechanisms of propofol in the human internal mammary artery (IMA). Design: In vitro experimental study. Setting: The study was conducted in the research laboratory of a pharmacology department. Participants: IMA segments were obtained from 52 patients undergoing coronary artery bypass surgery. Interventions: The IMA rings were suspended in isolated organ baths, and the changes in the tension were isometrically recorded. The antagonistic effect of propofol (1 μM, 10 μM, and 100 μM) on contractions induced by potassium chloride (45 mM), phenylephrine (1 μM), 5-hydroxytryptamine (30 μM), and calcium chloride (10 μM-10 mM) was investigated. The relaxations induced by propofol also were tested in the presence of the nitric oxide synthase inhibitor, nitro-L-arginine methyl ester (100 mM); the cyclooxygenase inhibitor, indomethacin (10 mM); and the potassium ion channel inhibitors, tetraethylammonium (1 mM), iberiotoxin (20 nM), glibenclamide (10 µM), 4-aminopyridine (1 mM), and barium chloride (30 µM). Measurements and main results: Propofol caused a significant concentration-dependent vasorelaxation, which was endothelium independent. It inhibited the contractions induced by potassium chloride, phenylephrine, 5-hydroxytryptamine, and calcium chloride (p < 0.001), but it did not affect the basal tension. Propofol-induced relaxation was significantly inhibited by iberiotoxin and tetraethylammonium (p < 0.001); however, it was not affected by 4-aminopyridine, glibenclamide, and barium chloride. Conclusion: This study clearly reveals that propofol relaxes the IMA, and propofol-induced vasodilation may be related to large conductance calcium ion-activated potassium ion channel activation. Propofol use in coronary artery bypass surgery can be valuable via its favorable vasodilator effect to overcome perioperative vasospasm of IMA.
Article
Background: In pulmonary arterial hypertension (PAH), pathological changes in pulmonary arterioles progressively raise pulmonary artery pressure and increase pulmonary vascular resistance, leading to right heart failure and high mortality rates. Recently, the first potassium channelopathy in PAH, because of mutations in KCNK3, was identified as a genetic cause and pharmacological target. Methods: Exome sequencing was performed to identify novel genes in a cohort of 99 pediatric and 134 adult-onset group I PAH patients. Novel rare variants in the gene identified were independently identified in a cohort of 680 adult-onset patients. Variants were expressed in COS cells and function assessed by patch-clamp and rubidium flux analysis. Results: We identified a de novo novel heterozygous predicted deleterious missense variant c.G2873A (p.R958H) in ABCC8 in a child with idiopathic PAH. We then evaluated all individuals in the original and a second cohort for rare or novel variants in ABCC8 and identified 11 additional heterozygous predicted damaging ABCC8 variants. ABCC8 encodes SUR1 (sulfonylurea receptor 1)-a regulatory subunit of the ATP-sensitive potassium channel. We observed loss of ATP-sensitive potassium channel function for all ABCC8 variants evaluated and pharmacological rescue of all channel currents in vitro by the SUR1 activator, diazoxide. Conclusions: Novel and rare missense variants in ABCC8 are associated with PAH. Identified ABCC8 mutations decreased ATP-sensitive potassium channel function, which was pharmacologically recovered.
Article
Nicorandil and long-acting nitrates are vasodilatory drugs used commonly in the management of chronic stable angina pectoris. Both nicorandil and long-acting nitrates exert anti-angina properties via activation of nitric oxide (NO) signalling pathways, triggering vascular smooth muscle cell relaxation. Nicorandil has additional actions as an arterial K+ ATP channel agonist, resulting in more “balanced” arterial and venous vasodilatation than nitrates. Ultimately, these drugs prevent angina symptoms through reductions in preload and diastolic wall tension and, to a lesser extent, epicardial coronary artery dilatation and lowering of systemic blood pressure. While there is some evidence to suggest a modest reduction in cardiovascular events among patients with stable angina treated with nicorandil compared to placebo, this prognostic benefit has yet to be proven conclusively. In contrast, there is emerging evidence to suggest that chronic use of long-acting nitrates might cause endothelial dysfunction and increased cardiovascular risk in some patients.
Article
Hypertension is a risk factor for cerebrovascular diseases, including stroke and dementia. During hypertension, arteries become constricted and are less responsive to vasodilators, including nitric oxide (NO). The regulation of arterial contractility by smooth muscle cell (myocyte) large-conductance calcium (Ca2+)-activated potassium (BK) channels is altered during hypertension, although mechanisms involved are unclear. We tested the hypothesis that dysfunctional trafficking of pore-forming BK channel (BKα) and auxiliary β1 subunits contributes to changes in cerebral artery contractility of stroke-prone spontaneously hypertensive rats (SP-SHRs). Our data indicate that the amounts of total and surface BKα and β1 proteins are similar in unstimulated arteries of age-matched SP-SHRs and normotensive Wistar-Kyoto rats. In contrast, stimulated surface-trafficking of β1 subunits by NO or membrane depolarization is inhibited in SP-SHR myocytes. PKCα (protein kinase C α) and PKCβII total protein and activity were both higher in SP-SHR than in Wistar-Kyoto rat arteries. NO or depolarization robustly activated Rab11, a small trafficking GTPase, in Wistar-Kyoto rat arteries but weakly activated Rab11 in SP-SHRs. Bisindolylmaleimide, a PKC inhibitor, and overexpression of a PKC phosphorylation-deficient Rab11A mutant (Rab11A S177A) restored stimulated β1 subunit surface-trafficking in SP-SHR myocytes. BK channel activation by NO was inhibited in SP-SHR myocytes and restored by Rab11A S177A expression. Vasodilation to NO and lithocholate, a BKα/β1 channel activator, was inhibited in pressurized SP-SHR arteries and reestablished by bisindolylmaleimide. In summary, data indicate that spontaneously active PKC inhibits Rab11A-mediated β1 subunit trafficking in arterial myocytes of SP-SHRs, leading to dysfunctional NO-induced BK channel activation and vasodilation.