ArticlePDF AvailableLiterature Review

Factors, fiction and endothelium-derived hyperpolarizing factor

Authors:

Abstract

1. The principal mediators of vascular tone are neural, endothelial and physical stimuli that result in the initiation of dilator and constrictor responses to facilitate the control of blood pressure. Two primary vasodilatory stimuli produced by the endothelium are nitric oxide (NO) and prostaglandins. An additional endothelium-dependent vasodilatory mechanism is characterized as the hyperpolarization-mediated relaxation that remains after the inhibition of the synthesis of NO and prostaglandins. This mechanism is due to the action of a so-called endothelium-derived hyperpolarizing factor (EDHF) and is dependent on either the release of diffusible factor(s) and/or to a direct contact-mediated mechanism. 2. Most evidence supports the concept that ‘EDHF’ activity is dependent on contact-mediated mechanisms. This involves the transfer of an endothelium-derived electrical current, as an endothelium-derived hyperpolarization (EDH), through direct heterocellular coupling of endothelial cells and smooth muscle cells via myoendothelial gap junctions (MEGJ). However, there is a lack of consensus with regard to the nature and mechanism of action of EDHF/EDH (EDH(F)), which has been shown to vary within and between vascular beds, as well as among species, strains, sex and during development, ageing and disease. 3. In addition to actual heterogeneity in EDH(F), further heterogeneity has resulted from the less-than-optimal design, analysis and interpretation of data in some key papers in the EDHF literature; with such views being perpetuated in the subsequent literature. 4. The focus of the present brief review is to examine what factors are proposed as EDH(F) and highlight the correlative structural and functional studies from our laboratory that demonstrate an integral role for MEGJ in the conduction of EDH, which account for the heterogeneity in EDH(F), while incorporating the reported diffusible mechanisms in the regulation of this activity. Furthermore, in addition to the reported heterogeneity in the nature and mechanism of action of EDH(F), the contribution of experimental design and technique to this heterogeneity will be examined.
Clinical and Experimental Pharmacology and Physiology
(2004)
31
,
563–570
BRIEF REVIEW
FACTORS, FICTION AND ENDOTHELIUM-DERIVED
HYPERPOLARIZING FACTOR
Shaun L Sandow
Division of Neuroscience, John Curtin School of Medical Research, Australian National University, Canberra,
Australian Capital Territory, Australia and Department of Pharmacy and Pharmacology, University of Bath,
Claverton Down, Bath, UK
SUMMARY
1. The principal mediators of vascular tone are neural,
endothelial and physical stimuli that result in the initiation of
dilator and constrictor responses to facilitate the control of
blood pressure. Two primary vasodilatory stimuli produced by
the endothelium are nitric oxide (NO) and prostaglandins. An
additional endothelium-dependent vasodilatory mechanism is
characterized as the hyperpolarization-mediated relaxation
that remains after the inhibition of the synthesis of NO and
prostaglandins. This mechanism is due to the action of a
so-called endothelium-derived hyperpolarizing factor (EDHF)
and is dependent on either the release of diffusible factor(s)
and/or to a direct contact-mediated mechanism.
2. Most evidence supports the concept that ‘EDHF’ activity
is dependent on contact-mediated mechanisms. This involves
the transfer of an endothelium-derived electrical current, as an
endothelium-derived hyperpolarization (EDH), through direct
heterocellular coupling of endothelial cells and smooth muscle
cells via myoendothelial gap junctions (MEGJ). However, there
is a lack of consensus with regard to the nature and mechanism
of action of EDHF/EDH (EDH(F)), which has been shown to
vary within and between vascular beds, as well as among
species, strains, sex and during development, ageing and
disease.
3. In addition to actual heterogeneity in EDH(F), further
heterogeneity has resulted from the less-than-optimal design,
analysis and interpretation of data in some key papers in the
EDHF literature; with such views being perpetuated in the
subsequent literature.
4. The focus of the present brief review is to examine what
factors are proposed as EDH(F) and highlight the correlative
structural and functional studies from our laboratory that
demonstrate an integral role for MEGJ in the conduction of
EDH, which account for the heterogeneity in EDH(F), while
incorporating the reported diffusible mechanisms in the
regulation of this activity. Furthermore, in addition to the
reported heterogeneity in the nature and mechanism of action
of EDH(F), the contribution of experimental design and
technique to this heterogeneity will be examined.
Key words: artery, connexin, endothelium, experimental
design, gap junction, smooth muscle, vascular tone, vaso-
constrictor, vasodilator.
WHAT IS EDH(F)?
The aim of the present brief review is to provide a critical overview
of the endothelium-derived hyperpolarizing factor/endothelium-
derived hyperpolarization (EDH(F)) field, with a focus on the role
of gap junctions in the EDH(F) phenomenon. More extensive
reviews on endothelium-derived hyperpolarizing factor (EDHF)
are provided by McGuire
et al.
,
1
Campbell and Gauthier,
2
Ding and
Triggle
3
and Griffith.
4
Briefly, the arterial endothelium produces three vasodilatory
factors: nitric oxide (NO), prostaglandins and EDH(F). Classically,
EDH(F) is the hyperpolarization and associated relaxation
remaining after the inhibition of the synthesis of NO synthase
(and, thus, NO) and prostaglandins
.
The two primary mechanisms
that can account for EDH(F) activity rely on either diffusible- and/
or contact-mediated mechanisms. Those that are dependent on the
release of a diffusible substance, for which there is yet to be
unequivocal evidence, are due to EDHF. Those that are dependent
on the direct contact of endothelial cells (EC) and smooth muscle
cells (SMC) via myoendothelial gap junctions (MEGJ) are due to
the transfer of an electrical current, as an endothelium-derived
hyperpolarization (EDH).
4–9
In both cases, the net result is the
hyperpolarization of the adjacent smooth muscle with subsequent
vessel dilation. For clarity, the term EDH(F) will be used here to
refer to both a diffusible- or contact-mediated mechanism.
Regardless of whether a diffusible- or contact-mediated mech-
anism is involved in EDH(F) activity, it is accepted that its action
is dependent on the release of intracellular calcium and the
activation of a specific pattern of potassium channels. The activ-
ation of receptors and/or application of physical stimuli, such as
shear stress, results in a rise in intracellular EC calcium.
1,4,10
Correspondence: Dr Shaun L Sandow, Division of Neuroscience, John
Curtin School of Medical Research, Australian National University,
Canberra, ACT 0200, Australia. Email: Shaun.Sandow@anu.edu.au
Presented at the Australian Physiological and Pharmacological Society
Symposium Potassium Channels and Endothelium-Derived Hyperpolariz-
ing Factor: Physiological and Clinical Roles, September 2003. Published
with the permission of the APPS and initially published on the APPS
website (http://www.apps.org.au).
Received 9 January 2004; revision 30 April 2004; accepted 7 June 2004.
564
SL Sandow
Subsequently, this results in the activation of small (S) and inter-
mediate (I) conductance calcium-activated potassium channels
(K
Ca
) located on EC and, in some cases, the activation of EC or
SMC large (B) K
Ca
.
1
This channel activation results in the gener-
ation of an EDH or the release of an EDHF, which is subsequently
transmitted to the adjacent SMC layer either via MEGJ or
diffusion.
1,2
Indeed, it is agreed that EDH(F) activity is blocked
by the application of K
Ca
antagonists, such as apamin (SK
Ca
antagonist) and charybdotoxin (non-selective IK
Ca
and BK
Ca
antagonist, with additional effects at voltage-dependent potassium
channels
3
) in combination
1,2
or apamin and 1-[(2-chloro-
phenyl)diphenylmethyl]-1H-pyrazole (TRAM)-34 (IK
Ca
antago-
nist) in combination
4,11,12
in the case of SK
Ca
- and IK
Ca
-dependent
responses, or by iberiotoxin in the case of BK
Ca
-dependent
responses.
2
The nature and mechanism of EDH(F) apparently varies within
and between vascular beds and among species, strains, sex and
during development, ageing and disease,
1–3
as well as with variable
experimental conditions and between laboratories.
4
A proposal for
unifying the role of EDH(F) and heterocellular coupling has
recently been put forward by Griffith.
4
This scheme incorporates
many of the proposed EDH(F) and questions others, for which
there is debatable evidence.
DIFFUSIBLE FACTORS
Contact-mediated mechanisms represent the simplest explanation
of EDH(F) activity, as a purely electrical event. However, the
release of diffusible factor/s from the endothelium, at a concen-
tration sufficient to change that of the internal elastic lamina and
the local environment surrounding the innermost layer of SMC, has
also been proposed to account for EDH(F) activity. This substance
then putatively effects the activation of SMC receptors and ion
channels to initiate smooth muscle hyperpolarization and
relaxation.
1–4
Diffusible factors proposed as EDHF include K
+
ions,
epoxyeicosatrienoic acids (EET), H
2
O
2
,
1,2
and C-type natriuretic
peptide (CNP).
13
N
G
-Nitro-
L
-arginine methyl ester (
L
-NAME)-
insensitive NO has also been suggested to account for EDHF
activity.
14,15
In addition,
S
-nitrosothiols have been suggested to
contribute to EDHF activity,
16
although the evidence for the
endothelial dependence of this response requires further
investigation.
Potassium ions
Several studies have supported the proposal that K
+
ions are an
EDHF in some vessels.
1,3,4,17
Indeed, since the original proposition
that K
+
ions were an EDHF, this hypothesis has received much
attention. Basically, this scheme involves the activation of EC K
Ca
and the subsequent EC efflux of K
+
from these channels. The
resultant potassium ‘cloud’
17
then reportedly diffuses across the
internal elastic lamina to act as an EDHF by evoking smooth
muscle hyperpolarization and relaxation via the activation of
smooth muscle Na
+
/K
+
-ATPase and inwardly rectifying potassium
channels,
17
key channels for the modulation of ionic mechanisms
that are reportedly sensitive to the application of ouabain and
barium, respectively. Antagonism of the EDHF response by these
blockers is used as defining evidence for K
+
as an EDHF. In its
current form, this mechanism is referred to as the ‘potassium cloud
hypothesis’.
17
A complication to this hypothesis is the efflux of K
+
from SMC
that arises as a result of depolarization, which would contribute to
the basal level of K
+
surrounding vascular cells and thus suppress
the K
+
/EDHF effect. At a simplistic level, the term ‘potassium
cloud’ is misleading, in that it implies the presence of a global
cloud of potassium surrounding the vascular cells when, in fact, any
physiologically relevant change in the K
+
concentration will be
transient and localized. Indeed, a more plausible scenario is that the
K
+
flux acts at restricted localized sites (microdomains), as has
been described in SMC and other cell types.
18
Interestingly, the most recent version of the ‘potassium cloud
hypothesis’ includes a role for MEGJ in the action of K
+
as
EDHF.
17
However, once a role for MEGJ is included in this
mechanism, a role for K
+
as a diffusible EDHF may be redundant,
because the EDHF phenomenon can be explained simply through
the action of EDH. As alluded to above, a potential scenario where
the diffusion of K
+
may play a role in the EDHF activity could arise
if there is a close spatial relationship between MEGJ and K
Ca
distribution (as well as, perhaps, with sites of calcium extrusion),
in the form of microdomains, where highly localized changes in K
+
concentrations could play a role in the coordination and modulation
of heterocellular EDH(F) signalling (CJ Garland and SL Sandow,
unpubl. obs., 2004). Although evidence for similar functional
microdomains in SMC and other cell types is well documented,
18
it is interesting to speculate that this scenario may be the case in
EC of resistance vessels, such as the mesenteric bed of the rat,
where functional studies have suggested this to occur.
19
Further
anatomical support for the existence of microdomains in EC is not
currently available in resistance vessels and, thus, a role for K
+
in
this scenario is speculative.
Epoxyeicosatrienoic acids
There is evidence of a role for EET in EDH(F) activity in some
vascular beds.
1,2
Epoxyeicosatrienoic acids are cytochrome P450
expoxygenase metabolites of phospholipase-dependent arachi-
donic acid production, which putatively activate smooth muscle
BK
Ca20
to result in hyperpolarization and arterial relaxation in
cerebral, coronary and renal arteries of several species.
1,2
Indeed,
although there is evidence that EET play an integral role in EDH(F)
activity in some vascular beds, EET are not a universal EDH(F), in
that in many vascular beds, EDH(F) activity is not sensitive to the
application of iberiotoxin, a BK
Ca
antagonist.
4
Furthermore, it is
not clear whether EET activity is related to the participation of EET
in the facilitation of autocrine pathways that generate hyper-
polarization via mechanisms that are indistinct from alternative
agonist-induced pathways that result in an analogous activation of
an EDH(F)-type response.
4
Hydrogen peroxide
In human and mouse mesenteric and porcine coronary arteries,
H
2
O
2
has been proposed to act as an EDHF.
21–24
However, a
primary problem with these studies is that the appropriate time and
concentration controls for catalase, as an H
2
O
2
antagonist, were not
undertaken and, indeed, the proposal that H
2
O
2
is an EDHF in these
vascular beds is not consistent with several other studies under-
Factors, fiction and EDH(F)
565
taken in the same vascular beds (see below). Beny and von der
Weid,
25
for example, have shown that EDHF and H
2
O
2
are distinct
factors in porcine coronary arteries, where Pomposiello
et al
.
26
have demonstrated that catalase, an enzyme inhibitor of H
2
O
2
activity, has no effect in porcine coronary vessels, although, at
300 U/mL, catalase did abolish the endothelium-independent
relaxation to exogenously generated H
2
O
2
after 45 min incubation.
Catalase has been shown to have no effect on EDHF in the bovine
ciliary, rat saphenous and mesenteric and human radial and sub-
cutaneous arteries.
9,27–30
In this light, several studies have shown
that H
2
O
2
can cause a vasoconstriction
31,32
that can be attenuated
by a 20 min incubation in 100 U/mL catalase.
33
Furthermore, in a
membrane potential-independent manner, reactive oxygen species,
such as H
2
O
2
, have been reported to variably activate SMC K
Ca
,
ATP-sensitive potassium channels and Na
+
/K
+
-ATPase and to
modulate the sensitivity of the contractile apparatus to
calcium,
4,10
thus playing additional roles unrelated to EDHF, but
complicating any speculative role for H
2
O
2
in EDHF activity.
Indeed, in contrast with the original proposition that H
2
O
2
was an
EDHF in mouse mesenteric vessels, Ellis
et al
.
34
provide evidence
that H
2
O
2
is not an EDHF in these vessels. Indeed, Ellis
et al
.
34
found that an inhibitory effect of catalase does not provide
definitive evidence that H
2
O
2
is critical to a given vascular
response.
10
In any event, the physiological relevance of H
2
O
2
as an EDHF
is simply questioned based on the observation that the concen-
tration of H
2
O
2
produced in response to endothelial stimulation
(10–60 nmol/L;
35
see also Griffith
4
) is substantially less than the
3–100
mol/L of H
2
O
2
required to elicit a 30–90% relaxation in
human mesenteric vessels
22
or the 0.1 and 1 mmol/L of H
2
O
2
required to elicit a 60 and 100% relaxation, respectively, in porcine
coronary arteries.
25
In addition, concentration-dependent effects of
H
2
O
2
are critical to the question of whether physiological or
pathophysiological effects are observed, because H
2
O
2
can mediate
vascular cell proliferation, apoptosis, hyperplasia, cell adhesion
and migration, as well as having effects on arterial tone.
10
Indeed,
predominant evidence supports the proposition that H
2
O
2 is not
involved in the hyperpolarization-dependent EDHF response and
that it is not an EDHF.10,36
C-Type natriuretic peptide
C-Type natriuretic peptide (CNP) has been proposed to act as an
EDHF13 and, indeed, the data presented by Chauhan et al.13 are
consistent with the activation of the CNP receptor C subtype
playing a role in the EDH(F) phenomenon. However, in the same
mesenteric vessels as examined in Sprague-Dawley rats by
Chauhan et al.13 but in the mature Wistar rat, Sandow et al.37
demonstrate that heterocellular coupling of EC and SMC accounts
for EDH activity in this bed. Although the difference between the
two studies could be related to strain variation, such a fundamental
difference is unlikely and the specific reason for the discrepancy is
unknown. Interestingly, in this light, the use of the non-selective
gap junction antagonist glycyrrhetinic acid (GA) and its derivatives
have implicated a primary role for gap junctional coupling in
EDH(F) activity in this vascular bed.38–41 Indeed, Chauhan et al.13
implicate a role for MEGJ in the proposal that CNP is EDH(F) via
the use of -GA, although at present this role is currently unknown
but is being investigated (A Ahluwalia, pers. comm.). In any case,
a role for MEGJ in the activity of CNP as EDH(F) is based on the
assumption that GA is a specific antagonist for MEGJ and because
no control studies for the effects of GA were undertaken in the
study of Chauhan et al.,13 this claim is open to question. Indeed,
GA and its derivatives have been shown to block homocellular and
MEGJ in this vessel,39,41 as well as having direct effects on the EC
hyperpolarization to acetylcholine (ACh) via effects on phospho-
lipase activity and EC SKCa, IKCa and Na+/K+-ATPase, irrespective
of their putative effects at gap junctions.6,42 A limitation of future
studies examining a potential role for CNP as an EDH(F) is the lack
of availability of selective antagonists for the CNP receptor C
subtype that is reported to mediate this response. Furthermore,
specific limitations of the study of Chauhan et al.13 include the lack
of a demonstration that the CNP-mediated relaxation can occur
independently of the endothelium (which would thus demonstrate
CNP action at the smooth muscle) and a lack of explanation of the
observations that CNP evokes approximately 60–70% relaxation,
whereas EDHF evokes approximately 100% relaxation. In addi-
tion, there is also a lack of explanation as to why the (non-specific)
blockade of gap junctions with GA suppresses CNP activity or
what effects barium alone has on the CNP- and EDHF-mediated
relaxations, or the inclusion of appropriate control data to deter-
mine whether there was a basal release of CNP in these mesenteric
vessels. Thus, a definitive role for CNP in EDH(F) activity remains
to be elucidated.
L-NAME-insensitive NO
Endogenous or basal NO activity, which is insensitive to the
application of NO synthase antagonists used in the routine study of
EDH(F), has been suggested to account for EDH(F) activity.14,15
Current evidence suggests that, in some vascular beds under
specific experimental conditions, this L-NAME-insensitive NO
may account for a minor degree of EDH(F) activity and one not
consistently observed in studies of the same vascular bed. For
example, in the study of Chauhan et al.15 purporting to show that
L-NAME-insensitive NO accounts for a significant portion of
EDH(F) activity, 63% of hyperpolarization and 70% of
relaxation to ACh remain after the addition of the NO scavenger
oxyhaemoglobin (in the presence of L-NAME and indomethacin).
Furthermore, in the caudal and saphenous arteries of the rat and
mesenteric artery of the mouse, the NO scavengers hydroxo-
cobalamin and carboxy–2-(4-carboxyphenyl)-4,5-dihydro-4,4,5,5-
tetramethyl-1H-imidazol-1-yloxy-3-oxide (PTIO) have no effect
on EDH(F),9,24,43 thus demonstrating a lack of an L-NAME-
insensitive NO component in these vascular beds. Therefore, the
contribution of endogenous NO to EDH(F) activity appears
variable and, in many cases, non-existent. Further studies are
required to determine the physiological relevance of this
phenomenon.
CONTACT-MEDIATED MECHANISMS
Evidence supporting the critical role of MEGJ in EDH(F) activity
comes primarily from structural and functional studies from our
laboratory in Canberra and Tudor Griffith’s4,36,44,45 laboratory in
Cardiff. These studies, which illustrate the simplest explanation of
EDH(F) activity, use electron microscopic identification of MEGJ,
electrophysiological recordings from dye-identified EC and SMC
566 SL Sandow
and myography with pharmacological interventions, as well as
immunohistochemical methods for identifying the connexins
and ion channels involved in the EDH(F) phenomenon. These
studies are consistent with the hypothesis that EDH(F) is an
electrical phenomenon involving the gap junctional transfer of an
EDH from the EC to the innermost layer of intimal SMC in
the arterial wall for the subsequent generation of an arterial
relaxation.
Studies from our laboratory, which are the focus of this section
of the present review, have examined the role of MEGJ in EDH
activity. We have found that the distribution and activity of MEGJ
is correlated with the presence of EDH within and between vas-
cular beds, during development and in disease. In the proximal and
distal mesenteric arteries of the rat, for example, gap junctions play
a critical role in EDH activity39,41 where MEGJ are prevalent.46 In
this vascular bed, in collaborative studies with Marianne Tare in
Helena Parkington’s laboratory in Melbourne, we showed that the
presence of EDH is correlated with the presence of MEGJ, whereas
in the femoral artery a lack of MEGJ is correlated with the absence
of EDH.37 A similar situation is present in the lateral saphenous
artery of the juvenile rat, where MEGJ are prevalent and EDH-
mediated relaxation present.9 This is in contrast with the saphenous
artery of the adult, where MEGJ were rare and EDH absent.9 The
relationship between EDH and MEGJ is somewhat more
complicated in disease states, such as in hypertension. In a com-
parative study of the caudal artery of the spontaneously hyper-
tensive and normotensive Wistar-Kyoto rats, EDH activity was
maintained in spite of an increase in the number of SMC layers
in vessels from the hypertensive rat. This maintenance was found
to be correlated with a concomitant increase in the incidence of
MEGJ in the caudal artery of the hypertensive rat.43
The above studies demonstrate that there is a direct relationship
between the degree of EDH and the incidence of MEGJ. Indeed,
EDH increases with an increase in the number of MEGJ per EC,
whereas, conversely, it generally decreases with an increase in the
number of SMC layers and vessel diameter (Fig. 1). Interestingly,
although EDH is the predominant vasodilator in smaller vessels, it
is present in some larger vessels (Fig. 1), such as the rabbit iliac,
rat caudal and superior mesenteric arteries.41,43,45 In the rabbit iliac
artery, cAMP has been proposed to enhance the spread of EDH via
modulating gap junctional coupling within the multiple SMC
layers, as well as at MEGJ.47 Although conclusive biophysical
evidence for this mechanism being relevant in larger vessels is
lacking,48 this mechanism may be of some importance for EDH
activity.
These studies demonstrate that there is a consistent positive
correlation between MEGJ and EDH activity within and between
vascular beds and during development and disease. Although this
correlation is not definitive evidence that contact-mediated mech-
anisms account for EDH(F) activity, to date these data provide the
most conclusive and plausible explanation for this activity.
Fig. 1 Summary data demonstrating the relationship between acetyl-
choline (ACh)-induced endothelium-derived hyperpolarizing factor/
endothelium-derived hyperpolarization (EDH(F)) activity and arterial
morphology as the number of myoendothelial gap junctions (MEGJ) per
endothelial cell (EC), per number of medial smooth muscle cell (SMC)
layers and per vessel diameter. Individual data points are presented as the
meanSEM, with data being derived from earlier studies.9,37,43,46 Data were
fitted with a one-phase exponential curve using Graphpad Prism (Graphpad
Software, San Diego, CA, USA). PE, phenylephrine. (), tertiary mesen-
teric (juvenile); (), primary mesenteric (juvenile); (), primary/secondary
mesenteric (juvenile); (), caudal (adult); (), saphenous (adult); (),
femoral (adult).
Factors, fiction and EDH(F) 567
ROLE OF DIFFUSIBLE FACTORS IN
CONTACT-MEDIATED MECHANISMS
Direct electrical coupling is the most plausible mechanism to fully
account for EDH activity. Indeed, there is increasing evidence that
the diffusible factors that act as credible EDHF may, in fact, be
associated with the modulation of gap junction activity and,
specifically, of MEGJ4 for the transfer of EDH as the most plausible
mechanism of their activity. These mechanisms are outlined
below.
Potassium ions
The original hypothesis regarding the mechanism of action of K+
as EDHF has been modified to include a role for MEGJ.17 However,
although K+ is involved in mediating EDH(F) activity, once a role
for such MEGJ is included, no direct role for K+ as a diffusible
EDHF is necessary for the transfer of an EDH. Indeed, in a series
of experiments that repeated those in the original proposal that K+
was EDHF, the data in the original study could not be repeated.49
In addition, several studies have questioned the nature of K+ as
EDHF because barium and ouabain, which are used to define the
role of K+ as an EDHF, do not universally block EDHF-mediated
responses.1,3,4,17,50 Indeed, the efficacy of ouabain as a selective
Na+/K+-ATPase antagonist has been questioned,4,51,52 whereby it
has inhibitory effects on cell coupling via modulating gap junction
function.53 Indeed, ouabain may directly attenuate the transfer of
EDH by its action at gap junctions.4,51,52 This action includes direct
effects on gap junctional coupling, such as reducing connexin (Cx)
expression through reduced Cx trafficking to the cell membrane, as
well as modulating gap junction conductance.52 The implication of
these observations is that the attenuation of an EDH(F) response
by ouabain, as with high concentrations of potassium, does not
necessarily provide evidence of the EDH(F) nature of the
response.4,6,52 The demonstration that ouabain has direct effects on
gap junctions and, thus, on EDH(F) are essentially control studies
for the earlier work that relied on the use of ouabain to show that
K+ was EDHF. Thus, based on these ‘control’ data,4,51,52 K+ ions are
not an EDH(F) but, rather, may simply be involved in the modu-
lation of the signal transduction pathways associated with gap
junction function54,55 and, thus, with EDH activity.4 Further investi-
gation is required to elucidate any potentially specific effects of
ouabain on vasomotor responses and those at gap junctions. Indeed,
this point is critical for the accurate interpretation of future EDH(F)
data.
Epoxyeicosatrienoic acids
In studies of cultured EC, EET have been shown to modulate
homocellular gap junctions,56 thus providing a potential mech-
anism for a modulatory role for EET in EDH action.4 Griffith4
suggests that EET activity may be related to a complex interaction
of calcium and potassium homeostasis, cAMP and arachidonic acid
activity and electrotonic signalling (see Fig. 3 in Griffith4 and also
Dhein50). Indeed, EET have been suggested to modulate EC KCa
activity,57 thus providing a further mechanism for their potential
role in modulating EDH, independent of acting directly as an
EDHF. Further studies of the role of EET in EDH activity in intact
vessels are required to clarify these proposals.
Hydrogen peroxide
There is some evidence that H2O2 can affect gap junction activity
and calcium homeostasis, two factors that are integral for EDH
activity. Depending on the experimental conditions, studies have
shown that H2O2 can both increase58 and decrease59 gap junctional
coupling and effect changes in intracellular calcium homeostasis,
both in cultured cells and in intact arteries.59–61 Although no
specific evidence is currently available to support this proposal,
these observations provide potential support for a mechanism to
link the putative role of H2O2 as an EDH(F) with the MEGJ
dependence of the EDH phenomenon.
C-Type natriuretic peptide
The putative action of CNP as an EDH(F)13 may be via acting as
yet another factor that facilitates electrical coupling through gap
junctions, although any putative mechanism for this is unknown.
Indeed, any putative action for CNP as EDH(F) cannot be associ-
ated directly with the gap junctional transfer of CNP from EC to
SMC because gap junctions are limited to passing substances of
1 kDa and CNP has a molecular weight of approximately
2.2 kDa (A Ahluwalia, pers. comm.). Interestingly, in the study of
Chauhan et al.13 proposing that CNP is an EDH(F), the response is
sensitive to the combination of barium and ouabain, an observation
that this is not a universal characteristic of EDH(F) in this, the rat
mesenteric vascular bed.49 Indeed, because ouabain is recognized
as a non-specific gap junction antagonist, this result may, in fact,
reflect an MEGJ dependence of EDH(F) in the mesenteric bed of
the rat, as demonstrated by Sandow et al.37
Myoendothelial gap junctions, EDH and gap junction
inhibitors
The demonstration of the dependence of EDH activity on gap
junctions relies, in part, on the specific pharmacological inhibition
of gap junctions. Unfortunately, there are a number of limitations
regarding this methodology. The primary one of these relates to the
dependence on the use of gap junction inhibitors that have not been
characterized adequately in terms of their specificity and mech-
anism of action. Currently, there is no unequivocal evidence that
the available gap junction inhibitors are specific,62 let alone selec-
tive, for gap junctions, be they heterocellular or homocellular.
Indeed, unfortunately, to date few studies have examined this
problem in detail and few have performed the defining experiment
of examining the effect of these agents on cell input resistance,
whereby an increase in input resistance would provide key data on
the gap junction antagonist effects of these agents. Of the studies
that have undertaken such technically demanding experiments, the
data are not consistent and are incomplete, although this may
reflect, in part, the heterogeneity in the Cx composition of vascular
gap junctions.63
Much of the current evidence for the gap junction and, specific-
ally, MEGJ dependence of EDH relies on the use of the licorice
derivatives (the GA and carbenoxolone; see above for an outline of
non-specific actions), the Cx-mimetic peptides (Gap26,43 Gap2740
and Gap27,37,43 which, based on putative selectivity, are the current
gap junction inhibitors of choice4,9,44) and, decreasingly, with the
long-chain alcohols, such as heptanol.64,65 However, there is little
568 SL Sandow
equivocal evidence that these agents are gap junction specific and
that they do not induce other non-gap junctional effects. Although
there is well-documented (and often ignored) evidence for the non-
specific effects of the licorice derivatives (see above) and hepta-
nol,64,65 the Cx-mimetic peptides have not yet been equivocally
tested for specificity and nor is their mechanism of action known.
In this regard, a primary issue with the use of the Cx-mimetic
peptides relates to the apparent requirement to use very high
concentrations and long incubation times to attenuate gap junction
activity.4 Interestingly, others report significant effects with lesser
concentrations of the peptide/s and reduced incubation times.62,66,67
Clearly, there is a pressing need for these issues to be addressed.
WHY IS THERE SUCH A DISPARITY OF VIEWS
AS TO THE NATURE AND MECHANISM OF
ACTION OF EDH(F)?
The conventional reason given for the disparity of views as to the
nature and mechanism of action of EDH(F) is that there is hetero-
geneity within and between arteries, species, sex, strain and disease
states.1–4,10,17 However, a further cause of the heterogeneity relates
to the less-than-optimal design, analysis and interpretation of data
present in some key papers in the EDH(F) literature. Although
some earlier studies can be seen as flawed with hindsight, this is
not necessarily the case, because they may, in fact, represent
significant contributions to the EDHF literature through their role
in advancing the evolution of the field. Unfortunately, this is not
always the case and the perpetuation of now potentially misleading
data is problematic. In any case, it is recognized that there is
variation in the nature and mechanism of EDH(F) between labor-
atories,4 thus questioning the relevance of the data and conclusions
of some studies.
The problems of experimental technique, with regard to the
design, analysis and interpretation of data that contribute to the
reported heterogeneity in the nature and mechanism of action of
EDH(F) in the literature include the following:
1. The use of selected agonists, antagonists and/or modulators
of the investigators’ choice and interest, but not those that may
indicate an alternative nature or mechanism of EDH(F).1,3,4,17 That
is, for example, an investigator may be interested in EET or gap
junctions to account for EDH(F) activity, but may limit the
investigation to the use of antagonists of the mechanism of their
interest, rather than of alternative pathways. This results in a
potential for a bias in favour of a particular putative EDH(F).1,3,4,17
2. The lack of control data for the effects of agonists, antago-
nists and other modulators. For instance, in the studies of Matoba
et al.21–23 examining the role of H2O2 as an EDH(F), justification
should be provided as to the incubation time with catalase (2 h or
longer), as well as the high (and variable) concentration of catalase
that was used.
3. The clear need for greater transparency with regard to
variability in cell, vessel and species specific responses, as a result
of a specific receptor and channel population and associated signal
transduction pathways.63
4. Making inappropriate comparative analyses between
studies, including a lack of consideration of strain,68,69 age,9,70–72
sex,73–76 the use of intact versus isolated tissue and tension versus
pressurized myography,63 as well as variation in the classification
of arterial branching patterns.39,41,46 Indeed, such characteristics are
often not stated in the methods section of papers and, thus, result
in an inability to make comparative analyses between studies.
5. Lack of clarity and relevance as to the experimental pro-
tocol. For instance, under conditions of little or no vascular tone,
the use of buffers (such as HEPES)77 that have non-physiological
effects, the use of preconstrictor agents that adversely effect
channel activity4 (such as the effect of U46619 on SKCa78 and the
effect of the GA and related compounds on a variety of cell
processes), as outlined above.
6. Extrapolation of data to other vascular beds. For example,
Chauhan et al.13,15 examined EDH(F) in mesenteric vessels of the
mature male Sprague-Dawley rat, but extrapolate the data to be
applicable to the vasculature as a whole. Although several studies
have made such claims,1,3,4 this contention merely confuses the
field because there is no evidence to justify this point of view.
CONCLUSIONS
The nature and mechanism of action of EDH(F) can apparently
differ along and between vascular beds, between species, strains,
sex and during development, ageing and disease. This hetero-
geneity can be explained through the action of heterocellular
coupling. Indeed, contact-mediated mechanisms represent the
simplest explanation of EDH(F) activity and involve the transfer of
an endothelium-derived electrical signal to the smooth muscle via
MEGJ, as EDH. Of the putative diffusion-mediated mechanisms,
K+ ions have received much attention in the literature and although
they may not be an EDHF, they are involved in the signal trans-
duction pathways associated with the generation of the EDH and
they may be involved in the modulation of gap junction activity. In
a similar manner, there is good evidence for a role of EET in
EDH(F) activity in some vascular beds, although this role may be
confined to a modulatory role of homo- and heterocellular
coupling, as well as modulating the KCa component of the EDH
mechanism. The role of CNP as an EDH(F) is yet to be clarified,
but may also be related to the modulation of EDH activity. Pre-
dominant evidence supports the proposition that H2O2 is not an
EDH(F), although, again, its activity may be related to the
modulation of gap junction function and, thus, of EDH. NG-Nitro-
L-arginine methyl ester-insensitive NO may account for a degree of
EDH(F) activity in some vascular beds, but the extent of this is
limited to only a minor part of such activity. Although the nature
and mechanism of action of EDH(F) is due, in part, to actual
heterogeneity, it is also unfortunately due to a lack of consistent and
sound scientific methodology.
ACKNOWLEDGEMENTS
The author’s work was supported by the National Heart Foundation
and National Health and Medical Research Council of Australia
(NHMRC) and the Wellcome Trust. SLS was supported by a
NHMRC Peter Doherty Fellowship. The author thanks Dr Alister
McNeish (Department of Pharmacy and Pharmacology, University
of Bath, Bath, UK) for critical comments on the manuscript.
REFERENCES
1. McGuire JJ, Ding H, Triggle CR. Endothelium-derived relaxing
factors. A focus on endothelium-derived hyperpolarizing factor(s).
Can. J. Physiol. Pharmacol. 2001; 79: 443–70.
Factors, fiction and EDH(F) 569
2. Campbell WB, Gauthier KM. What is new in endothelium-derived
hyperpolarizing factors? Curr. Opin. Nephrol. Hypertens. Res. 2002;
11: 177–83.
3. Ding H, Triggle CR. Contribution of EDHF and the role of potassium
channels in the regulation of vascular tone. Drug Dev. Res. 2003; 58:
81–9.
4. Griffith TM. Endothelium-dependent smooth muscle hyperpolariz-
ation: Do gap junctions provide a unifying hypothesis? Br. J.
Pharmacol. 2004; 141: 881–903.
5. Yamamoto Y, Imaeda K, Suzuki H. Endothelium-dependent hyper-
polarization and intercellular electrical coupling in guinea-pig
mesenteric arterioles. J. Physiol. 1999; 514: 505–13.
6. Coleman HA, Tare M, Parkington HC. K+ currents underlying the
action of endothelium-derived hyperpolarizing factor in guinea-pig
and human blood vessels. J. Physiol. 2001; 531: 359–73.
7. Coleman HA, Tare M, Parkington HC. EDHF is not K+ but may be due
to spread of current from the endothelium in guinea pig arterioles. Am.
J. Physiol. Heart Circ. Physiol. 2001; 280: H2478–83.
8. Coleman HA, Tare M, Parkington HC. Myoendothelial electrical
coupling in arteries and arterioles and its implications for endo-
thelium-derived hyperpolarizing factor. Clin. Exp. Pharmacol.
Physiol. 2002; 29: 630–7.
9. Sandow SL, Goto K, Rummery N, Hill CE. Developmental depen-
dence of EDHF on myoendothelial gap junctions in the saphenous
artery of the rat. J. Physiol. 2004; 556: 875–86.
10. Ellis A, Triggle CR. Endothelium-dependent reactive oxygen species:
Their relationship to endothelium-dependent hyperpolarization and
the regulation of vascular tone. Can. J. Physiol. Pharmacol. 2003; 81:
1013–28.
11. Crane GJ, Walker SD, Dora KA, Garland CJ. Evidence for a differ-
ential cellular distribution of inward rectifier K channels in the rat
isolated mesenteric artery. J. Vasc. Res. 2003; 40: 159–68.
12. Eichler I, Wibawa J, Grgic I et al. Selective blockade of endothelial
Ca(2+)-activated small- and intermediate-conductance K(+)-channels
suppresses EDHF-mediated vasodilation. Br. J. Pharmacol. 2003;
138: 594–601.
13. Chauhan SD, Nilsson H, Ahluwalia A, Hobbs AJ. Release of C-type
natriuretic peptide accounts for the biological activity of endothelium-
derived hyperpolarizing factor. Proc. Natl Acad. Sci. USA 2003; 100:
1426–31.
14. Cohen RA, Plane F, Najibi S, Huk I, Malinski T, Garland CJ. Nitric
oxide is the mediator of both endothelium-dependent relaxation and
hyperpolarization of the rabbit carotid artery. Proc. Natl Acad. Sci.
USA 1997; 94: 4193–8.
15. Chauhan S, Rahman A, Nilsson H, Clapp L, MacAllister R, Ahluwalia
A. NO contributes to EDHF-like responses in rat small arteries: A role
for NO stores. Cardiovasc. Res. 2003; 57: 207–16.
16. Batenburg WW, Popp R, Fleming I et al. Bradykinin-induced relax-
ation of coronary microarteries: S-Nitrosothiols as EDHF? Br. J.
Pharmacol. 2004; 142: 125–35.
17. Busse R, Edwards G, Feletou M, Fleming I, Vanhoutte PM, Weston A.
EDHF: Bringing the concepts together. Trends Pharmacol. Sci. 2002;
23: 374–80.
18. Poburko D, Kuo KH, Dai J, Lee CH, van Breemen C. Organellar
junctions promote targeted Ca2+ signalling in smooth muscle: Why two
mechanisms are better than one. Trends Pharmacol. Sci. 2004; 25:
8–15.
19. Crane GJ, Gallagher NT, Dora KA, Garland CJ. Small and inter-
mediate calcium-dependent K+ channels provide different facets of
endothelium-dependent hyperpolarization in rat mesenteric artery.
J. Physiol. 2003; 553: 183–9.
20. Archer SL, Gragasin FS, Wu X et al. Endothelium-derived hyper-
polarizing factor in human internal mammary artery is 11,12-
epoxyeicosatrienoic acid and causes relaxation by activating smooth
muscle BK(Ca) channels. Circulation 2003; 107: 769–76.
21. Matoba T, Shimokawa H, Nakashima M et al. Hydrogen peroxide is
an endothelium-derived hyperpolarizing factor in mice. J. Clin. Invest.
2000; 106: 1521–30.
22. Matoba T, Shimokawa H, Kubota H et al. Hydrogen peroxide is an
endothelium-derived hyperpolarizing factor in human mesenteric
arteries. Biochem. Biophys. Res. Commun. 2002; 290: 909–13.
23. Matoba T, Shimokawa H, Morikawa K et al. Electron spin resonance
detection of hydrogen peroxide as an endothelium-derived hyper-
polarizing factor in porcine coronary microvessels. Arterioscler.
Thromb. Vasc. Biol. 2003; 23: 1224–30.
24. Morikawa K, Shimokawa H, Matoba T et al. Pivotal role of Cu,Zn-
superoxide dismutase in endothelium-dependent hyperpolarization.
J. Clin. Invest. 2003; 112: 1871–9.
25. Beny JL, von der Weid PY. Hydrogen peroxide: An endogenous
smooth muscle cell hyperpolarizing factor. Biochem. Biophys. Res.
Commun. 1991; 176: 378–84.
26. Pomposiello S, Rhaleb NE, Alva M, Carretero OA. Reactive oxygen
species: Role in the relaxation induced by bradykinin or arachidonic
acid via EDHF in isolated porcine coronary arteries. J. Cardiovasc.
Pharmacol. 1999; 34: 567–74.
27. Hamilton CA, McPhaden AR, Berg G, Pathi V, Dominiczak AF. Is
hydrogen peroxide an EDHF in human radial arteries? Am. J. Physiol.
Heart Circ. Physiol. 2001; 280: H2451–5.
28. McNeish AJ, Wilson WS, Martin W. Ascorbate blocks endothelium-
derived hyperpolarizing factor (EDHF)-mediated vasodilatation in the
bovine ciliary vascular bed and rat mesentery. Br. J. Pharmacol. 2002;
135: 1801–9.
29. Kawabata A, Kubo S, Nakaya Y et al. Distinct roles for protease-
activated receptors 1 and 2 in vasomotor modulation in rat superior
mesenteric artery. Cardiovasc. Res. 2004; 61: 683–92.
30. Luksha L, Nisell H, Kublickiene K. The mechanism of EDHF-
mediated responses in subcutaneous small arteries from healthy
pregnant women. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2004;
286: R1102–9.
31. Jin N, Packer CS, Rhoades RA. Reactive oxygen-mediated contraction
in pulmonary arterial smooth muscle: Cellular mechanisms. Can. J.
Physiol. Pharmacol. 1998; 69: 383–8.
32. Gao YJ, Lee RM. Hydrogen peroxide induces a greater contraction in
mesenteric arteries of spontaneously hypertensive rats through throm-
boxane A(2) production. Br. J. Pharmacol. 2001; 134: 1639–46.
33. Ulker S, McMaster D, McKeown PP, Bayraktutan U. Impaired
activities of antioxidant enzymes elicit endothelial dysfunction in
spontaneous hypertensive rats despite enhanced vascular nitric oxide
generation. Cardiovasc. Res. 2003; 59: 488–500.
34. Ellis A, Pannirselvam M, Anderson TJ, Triggle CR. Catalase has
negligible inhibitory effects on endothelium-dependent relaxations in
mouse isolated aorta and small mesenteric artery. Br. J. Pharmacol.
2003; 140: 1193–200.
35. Cosentino F, Patton S, d’Uscio LV et al. Tetrahydrobiopterin alters
superoxide and nitric oxide release in prehypertensive rats. J. Clin.
Invest. 1998; 101: 1530–7.
36. Chaytor AT, Edwards DH, Bakker LM, Griffith TM. Distinct hyper-
polarizing and relaxant roles for gap junctions and endothelium-
derived H2O2 in NO-independent relaxations of rabbit arteries. Proc.
Natl Acad. Sci. USA 2003; 100: 15 212–17.
37. Sandow SL, Tare M, Coleman HA, Hill CE, Parkington HC.
Involvement of myoendothelial gap junctions in the actions of
endothelium-derived hyperpolarizing factor. Circ. Res. 2002; 90:
1108–13.
38. Edwards G, Feletou M, Gardener MJ, Thollon C, Vanhoutte PM,
Weston AH. Role of gap junctions in the responses to EDHF in rat and
guinea-pig small arteries. Br. J. Pharmacol. 1999; 128: 1788–94.
39. Hill CE, Hickey H, Sandow SL. Role of gap junctions in acetyl-
choline-induced vasodilation of proximal and distal arteries of the rat
mesentery. J. Auton. Nerv. Syst. 2000; 81: 122–7.
40. Doughty JM, Boyle JP, Langton PD. Blockade of chloride channels
reveals relaxations of rat small mesenteric arteries to raised potassium.
Br. J. Pharmacol. 2001; 132: 293–301.
41. Goto K, Fujii K, Kansui Y, Abe I, Iida M. Critical role of gap junctions
in endothelium-dependent hyperpolarization in rat mesenteric arteries.
Clin. Exp. Pharmacol. Physiol. 2002; 29: 595–602.
570 SL Sandow
42. Tare M, Coleman HA, Parkington HC. Glycyrrhetinic derivatives
inhibit hyperpolarization in endothelial cells of guinea pig and rat
arteries. Am. J. Physiol. Heart Circ. Physiol. 2002; 282: H335–41.
43. Sandow SL, Bramich NJ, Bandi HP, Rummery N, Hill CE. Structure,
function and EDHF in the caudal artery of the SHR and WKY rat.
Arterioscler. Thromb. Vasc. Biol. 2003; 23: 822–8.
44. Chaytor AT, Martin PE, Edwards DH, Griffith TM. Gap junctional
communication underpins EDHF-type relaxations evoked by ACh in
the rat hepatic artery. Am. J. Physiol. Heart Circ. Physiol. 2001; 280:
H2441–50.
45. Chaytor AT, Taylor HJ, Griffith TM. Gap junction-dependent and
-independent EDHF-type relaxations may involve smooth muscle
cAMP accumulation. Am. J. Physiol. Heart Circ. Physiol. 2002; 282:
H1548–55.
46. Sandow SL, Hill CE. The incidence of myoendothelial gap junctions
in the proximal and distal mesenteric arteries of the rat is suggestive
of a role in EDHF-mediated responses. Circ. Res. 2000; 86: 341–6.
47. Griffith TM, Chaytor AT, Taylor HJ, Giddings BD, Edwards DH.
cAMP facilitates EDHF-type relaxations in conduit arteries by
enhancing electrotonic conduction via gap junctions. Proc. Natl Acad.
Sci. USA 2002; 99: 6392–7.
48. Diep HK, Vigmond EK, Welsh DG. Modelling of electrical communi-
cation in resistance arteries. FASEB J. 2004; 18: A4236 (Abstract).
49. Vanheel B, Van de Voorde J. Effects of barium, ouabain and K+ on the
resting membrane potential and endothelium-dependent responses in
rat arteries. In: Vanhoutte PM (ed.). EDHF 2000. Taylor & Francis,
London. 2001; 146–55.
50. Dhein S. Pharmacology of gap junctions in the cardiovascular system.
Cardiovasc. Res. 2004; 62: 287–98.
51. Harris D, Martin PE, Evans WH, Kendall DA, Griffith TM, Randall
MD. Role of gap junctions in endothelium-derived hyperpolarizing
factor responses and mechanisms of K(+) -relaxation. Eur. J.
Pharmacol. 2000; 402: 119–28.
52. Martin PE, Hill NS, Kristensen B, Errington RJ, Griffith TM. Ouabain
exerts biphasic effects on connexin functionality and expression in
vascular smooth muscle cells. Br. J. Pharmacol. 2003; 140: 1261–71.
53. Ledbetter ML, Gatto CL. Concentrations of ouabain that prevent
intercellular communication do not affect free calcium levels in
cultured fibroblasts. Cell Biochem. Funct. 2003; 21: 363–70.
54. Giaume C, Venance L. Characterization and regulation of gap junction
channels in cultured astrocytes. In: Spray DC, Dermietzel R (eds). Gap
Junctions in the Nervous System. Landes: Austin. 1996; 136–57.
55. Pina-Benabou MH, Srinivas M, Spray DC, Scemes E. Calmodulin
kinase pathway mediates the K+-induced increase in gap junctional
communication between mouse spinal cord astrocytes. J. Neurosci.
2001; 21: 6635–43.
56. Popp R, Brandes RP, Ott G, Busse R, Fleming I. Dynamic modulation
of interendothelial gap junctional communication by 11,12-epoxyeico-
satrienoic acid. Circ. Res. 2002; 90: 800–6.
57. Baron A, Frieden M, Beny JL. Epoxyeicosatrienoic acids activate a
high-conductance, Ca(2+)-dependent K+ channel on pig coronary
artery endothelial cells. J. Physiol. 1997; 504: 537–43.
58. Rouach N, Calvo CF, Duquennoy H, Glowinski J, Giaume C.
Hydrogen peroxide increases gap junctional communication and
induces astrocyte toxicity: Regulation by brain macrophages. Glia
2004; 45: 28–38.
59. Todt I, Ngezahayo A, Ernst A, Kolb HA. Hydrogen peroxide inhibits
gap junctional coupling and modulates intracellular free calcium in
cochlear Hensen cells. J. Membr. Biol. 2001; 181: 107–14.
60. Blanc EM, Bruce-Keller AJ, Mattson MP. Astrocytic gap junctional
communication decreases neuronal vulnerability to oxidative stress-
induced disruption of Ca2+ homeostasis and cell death. J. Neurochem.
1998; 70: 958–70.
61. Touyz RM. Activated oxygen metabolites: Do they really play a role
in angiotensin II-regulated tone? J. Hypertens. 2003; 21: 2235–8.
62. Spray DC, Rozental R, Srinivas M. Prospects for rational development
of pharmacological gap junction channel blockers. Curr. Drug Targets
2002; 3: 455–64.
63. Hill CE, Phillips JK, Sandow SL. Heterogeneous control of blood flow
amongst different vascular beds. Med. Res. Rev. 2001; 21: 1–60.
64. Hashitani H, Suzuki H. K+ channels which contribute to the acetyl-
choline-induced hyperpolarization in smooth muscle of the guinea-pig
submucosal arteriole. J. Physiol. 1997; 501: 505–13.
65. Matchkov VV, Rahman A, Peng H, Nilsson H, Aalkjaer C. Junctional
and nonjunctional effects of heptanol and glycyrrhetinic acid derivates
in rat mesenteric small arteries. Br. J. Pharmacol. 2004; 142: 961–72.
66. Dahl G, Nonner W, Werner R. Attempts to define functional domains
of gap junction proteins with synthetic peptides. Biophys. J. 1994; 67:
1816–22.
67. Hill CE, Haddock RE, Brackenbury T. Role of the endothelium and
gap junctions in cerebral vasomotion. FASEB J. 2004; 18: A3631
(Abstract).
68. Kurtz TW, Morris Jr RC. Biological variability in Wistar-Kyoto rats:
Implications for research with the spontaneously hypertensive rat.
Hypertension 1987; 10: 127–31.
69. Louis WJ, Howes LG. Genealogy of the spontaneously hypertensive
rat and Wistar-Kyoto rat strains: Implications for studies of inherited
hypertension. J. Cardiovasc. Pharmacol. 1990; 16 (Suppl.): S1–5.
70. Fujii K, Ohmori S, Tominaga M et al. Age-related changes in
endothelium-dependent hyperpolarization in the rat mesenteric artery.
Am. J. Physiol. 1993; 265: H509–16.
71. Goto K, Fujii K, Onaka U, Abe I, Fujishima M. Angiotensin-
converting enzyme inhibitor prevents age-related endothelial
dysfunction. Hypertension 2000; 36: 581–7.
72. Goto K, Fujii K, Kansui Y, Iida M. Changes in EDHF in hypertension
and ageing: Response to chronic treatment with renin–angiotensin
system inhibitors. Clin. Exp. Pharmacol. Physiol. 2004; 31: 650–5.
73. Huang A, Sun D, Koller A, Kaley G. Gender difference in flow-
induced dilation and regulation of shear stress: Role of estrogen and
nitric oxide. Am. J. Physiol. Renal Physiol. 1998; 275: R1571–7.
74. Golding EM, Kepler TE. Role of estrogen in modulating EDHF-
mediated dilations in the female rat middle cerebral artery. Am. J.
Physiol. Heart Circ. Physiol. 2001; 280: H2417–23.
75. Xu HL, Santizo RA, Baughman VL, Pelligrino DA. ADP-induced pial
arteriolar dilation in ovariectomized rats involves gap junctional
communication. Am. J. Physiol. Heart Circ. Physiol. 2002; 283:
H1082–91.
76. Huang A, Sun D, Wu Z et al. Estrogen elicits cytochrome P450-
mediated flow-induced dilation of arterioles in NO deficiency. Role of
PI3K–Akt phosphorylation in genomic regulation. Circ. Res. 2004; 94:
245–52.
77. Edwards G, Feletou M, Gardener MJ et al. Further investigations into
the endothelium-dependent hyperpolarizing effects of bradykinin and
substance P in porcine coronary artery. Br. J. Pharmacol. 2001; 133:
1145–53.
78. Crane GJ, Garland CJ. Thromboxane receptor stimulation associated
with loss of SKCa activity and reduced EDHF responses in the rat
isolated mesenteric artery. Br. J. Pharmacol. 2004; 142: 43–50.
... The IEL represents a flexible barrier between the endothelium and inner smooth muscle cell layer and may have a role in atherogenesis via its modulation of diffusion across the artery wall (Hutchison & Sanders, 1990;Osborne-Pellegrin, 1986). Holes in the IEL are implicated in myoendothelial signal-ling; enabling the passage of endothelial and smooth muscle cell projections and subsequent contact between these cell layers in some vessels, which is integral for local vasodilator activity enabling the passage of endotheliumderived hyperpolarization (EDH), as well as for the conduction of vasomotor responses over distance (Sandow et al. 2002;Sandow, 2004;De Wit et al. 2006;Sandow & Tare, 2007). However, a significant proportion of such holes do not have cellular contents and the functional significance of these holes is unknown, although various possibilities have been suggested. ...
... The reason for increased IEL hole density in cerebral vessels is unknown. Of interest, EDH is correlated with MEGJ density in a number of arteries, including within the same vascular beds, such as in proximal and distal mesenteric arteries (Sandow & Hill, 2000;Sandow, 2004). Results from proximal and distal mesenteric arteries suggest that there was a similar increased density of IEL holes in smaller distal mesenteric vessels, thus in this case suggesting a relationship to function. ...
... Results from proximal and distal mesenteric arteries suggest that there was a similar increased density of IEL holes in smaller distal mesenteric vessels, thus in this case suggesting a relationship to function. Indeed, in the aorta (Sandow, 2004;Feletou & Vanhoutte, 2007;Sandow & Tare, 2007) the presence or absence of MEJGs shows an apparent correspondence with the presence of EDH, this mechanism being present in rat aorta of juvenile (Martinez-Org et al. 1999) and disease models (hypercholesterolemic, diabetic and hypertensive and with altered estrogen levels), but absent in healthy adult aorta (Wu et al. 1993;Endo et al. 1995;Kagota et al. 2000;Shimamura et al. 2000;Matsumoto et al. 2004;Woodman & Boujaoude, 2004;Malakul et al. 2008). In many vascular beds, the vasodilator EDH mechanism is dependent on MEGJs, with correlative and concomitant modification in development, ageing and disease (Griffith, 2004;Feletou & Vanhoutte, 2007;Sandow & Tare, 2007). ...
Article
Internal elastic lamina (IEL) hole (fenestration) characteristics and myoendothelial gap junction (MEGJ) density were examined in selected resistance and conduit arteries of normal and diseased rat and mouse models, using conventional, ultrastructural and confocal microscopy methods. Selected vessels were those commonly used in functional studies: thoracic aorta, proximal and distal mesenteric, caudal, saphenous, middle‐cerebral and caudal cerebellar artery. Rat and mouse strains and treatment groups examined were Dahl, Sprague Dawley, Wistar Kyoto, Wistar, spontaneously hypertensive (SHR), deoxycorticosterone (DOC) treated rat; and apolipoprotein E knockout, C57/BL6 and BALB/c mice. Vessel size (as IEL circumference), IEL hole and MEGJ density were quantified. In mesenteric arteries, the width of IEL holes and the percent of IEL occupied by holes were also determined. IEL hole density varied significantly within and between mesenteric artery beds, even among normotensive rat strains. Among the hypertensive rats (SHR and DOC), hole density in some vessels was higher in the normotensives than in the hypertensives within each strain, whereas in Dahl rats, hole density was similar between hypertensives and normotensives. Hole density was not correlated with the formation of intimal lesions in superior mesenteric artery. There was no positive general correlation between IEL hole and MEGJ density in resistance and conduit vessels. However, there was a positive correlation between the size of some resistance arteries and MEGJ density, although such a relationship did not hold for conduit vessels or during development, and there was no such relationship between vessel size and IEL hole density. Whilst IEL holes are obviously required for MEGJ communication, their presence is not an indication of contact‐mediated communication, but rather may be related to the presence of sites for the low resistance passage of diffusion‐mediated release of vasoactive endothelial and smooth muscle substances.
... In addition to the release of relaxing factors such as nitric oxide (NO) and prostaglandins, endothelial cells relax the vascular smooth muscle cells through the generation of smooth muscle hyperpolarization in an endothelium-dependent manner [2][3][4]. Although the mechanisms by which endothelial cells produce smooth muscle hyperpolarization may vary depending on the vascular beds and species, both diffusible factors and contact-mediated pathways contribute to the endothelium-dependent smooth muscle hyperpolarization [5][6][7]. ...
... Specifically, endothelium-dependent hyperpolarization (EDH) initiated in endothelial cells with a rise in intracellular calcium and the subsequent activation of small (SKCa) and intermediate conductance (IKCa) Ca 2+ -activated K + channels spreads to adjacent smooth muscle cells via myoendothelial gap junctions (MEGJs) in a number of vascular beds [17][18][19][20][21][22][23][24][25]. Although there is a consensus that the intracellular release of Ca 2+ from the endoplasmic reticulum (ER) and subsequent activation of the SKCa and IKCa channels in the endothelium is a prerequisite for the generation of EDH [5][6][7], several studies suggest that Ca 2+ influx through endothelial non-selective cation channels of the transient receptor potential (TRP) family also plays an important role in EDH via the downstream activation of SKCa and IKCa channels in some vascular beds [26][27][28][29][30]. In certain vascular beds in specific conditions, diffusible factors such as EETs and K + ions generate EDH through the activation of endothelial TRP channels [14] and endothelial inward rectifier K + (Kir) channels [31], respectively ( Figure 1). ...
... Endothelial cells also produce smooth muscle hyperpolarization in a contact-dependent manner [5][6][7]. Specifically, endothelium-dependent hyperpolarization (EDH) initiated in endothelial cells with a rise in intracellular calcium and the subsequent activation of small (SK Ca ) and intermediate conductance (IK Ca ) Ca 2+ -activated K + channels spreads to adjacent smooth muscle cells via myoendothelial gap junctions (MEGJs) in a number of vascular beds [17][18][19][20][21][22][23][24][25]. Although there is a consensus that the intracellular release of Ca 2+ from the endoplasmic reticulum (ER) and subsequent activation of the SK Ca and IK Ca channels in the endothelium is a prerequisite for the generation of EDH [5][6][7], several studies suggest that Ca 2+ influx through endothelial non-selective cation channels of the transient receptor potential (TRP) family also plays an important role in EDH via the downstream activation of SK Ca and IK Ca channels in some vascular beds [26][27][28][29][30]. ...
Article
Full-text available
Upon stimulation with agonists and shear stress, the vascular endothelium of different vessels selectively releases several vasodilator factors such as nitric oxide and prostacyclin. In addition, vascular endothelial cells of many vessels regulate the contractility of the vascular smooth muscle cells through the generation of endothelium-dependent hyperpolarization (EDH). There is a general consensus that the opening of small- and intermediate-conductance Ca2+-activated K⁺ channels (SKCa and IKCa) is the initial mechanistic step for the generation of EDH. In animal models and humans, EDH and EDH-mediated relaxations are impaired during hypertension, and anti-hypertensive treatments restore such impairments. However, the underlying mechanisms of reduced EDH and its improvement by lowering blood pressure are poorly understood. Emerging evidence suggests that alterations of endothelial ion channels such as SKCa channels, inward rectifier K⁺ channels, Ca2+-activated Cl- channels, and transient receptor potential vanilloid type 4 channels contribute to the impaired EDH during hypertension. In this review, we attempt to summarize the accumulating evidence regarding the pathophysiological role of endothelial ion channels, focusing on their relationship with EDH during hypertension.
... The close spatial proximity reached by endothelial and SMCs at these microdomains provides a rapid pathway for paracrine signaling and, consistent with this notion, a pool of the NO-synthetizing enzyme in endothelial cells, the endothelial isoform of NO synthase, is located at myoendothelial junctions [65][66][67]. Likewise, the endotheliummediated, NO-independent smooth muscle hyperpolarization was first thought to rely on the release of a diffusible factor by ECs and, consequently, was termed as endothelium-derived hyperpolarizing factor (EDHF) [13,60,68,69]. Several candidates have been proposed to mediate the EDHF signaling, such as K + ions [70], epoxyeicosatrienoic acids [71,72], hydrogen peroxide [73], and C-type natriuretic peptide [74][75][76]. ...
Article
Vascular system is a complex network in which different cell types and vascular segments must work in concert to regulate blood flow distribution and arterial blood pressure. Although paracrine/autocrine signaling is involved in the regulation of vasomotor tone, direct intercellular communication via gap junctions plays a central role in the control and coordination of vascular function in the microvascular network. Gap junctions are made up by connexin (Cx) proteins, and among the four Cxs expressed in the cardiovascular system (Cx37, Cx40, Cx43, and Cx45), Cx40 has emerged as a critical signaling pathway in the vessel wall. This Cx is predominantly found in the endothelium, but it is involved in the development of the cardiovascular system and in the coordination of endothelial and smooth muscle cell function along the length of the vessels. In addition, Cx40 participates in the control of vasomotor tone through the transmission of electrical signals from the endothelium to the underlying smooth muscle and in the regulation of arterial blood pressure by renin-angiotensin system in afferent arterioles. In this review, we discuss the participation of Cx40-formed channels in the development of cardiovascular system, control and coordination of vascular function, and regulation of arterial blood pressure.
... localized K þ release are suggested. 45 Importantly, no difference in acetylcholine response 21 nor in the expression of total eNOS was found between a2 þ/G301R and WT arteries. We thus hypothesize that a larger [Ca 2þ ] i elevation occurs in a2 þ/G301R endothelial cells and, with a greater activity of endothelium-dependent relaxing factor(s), which remains to be identified. ...
Article
Full-text available
Aims: Acute migraine attack in familial hemiplegic migraine type 2 (FHM2) patients is characterized by sequential hypo- and hyperperfusion. FHM2 is associated with mutations in the Na,K-ATPase α2 isoform. Heterozygous mice bearing one of these mutations (α2+/G301R) were shown to have elevated cerebrovascular tone and, thus, hypoperfusion that might lead to elevated concentrations of local metabolites. We hypothesize that these α2+/G301R mice also have increased cerebrovascular hyperemic responses to these local metabolites leading to hyperperfusion in the affected part of the brain. Methods and results: Neurovascular coupling was compared in α2+/G301R and matching wild type (WT) mice using Laser Speckle Contrast Imaging. In brain slices, parenchymal arteriole diameter and intracellular calcium changes in neuronal tissue, astrocytic endfeet and smooth muscle cells in response to neuronal excitation were assessed. Wall tension and smooth muscle membrane potential were measured in isolated middle cerebral arteries. Quantitative PCR, Western blot and immunohistochemistry were used to assess the molecular background underlying the functional changes. Whisker stimulation induced larger increase in blood perfusion, i.e. hyperemic response, of the somatosensory cortex of α2+/G301R than WT mice. Neuronal excitation was associated with larger parenchymal arteriole dilation in brain slices from α2+/G301R than WT mice. These hyperemic responses in vivo and ex vivo were inhibited by BaCl2, suggesting involvement of inward-rectifying K+ channels (Kir). Relaxation to elevated bath K+ was larger in arteries from α2+/G301R compared to WT mice. This difference was endothelium-dependent. Endothelial Kir2.1 channel expression was higher in arteries from α2+/G301R mice. No sex difference in functional responses and Kir2.1 expression was found. Conclusions: This study suggests that an abnormally high cerebrovascular hyperemic response in α2+/G301R mice is a result of increased endothelial Kir2.1 channel expression. This may be initiated by vasospasm-induced accumulation of local metabolites and underlie the hyperperfusion seen in FHM2 patients during migraine attack. Translational perspective: Migraine, especially familial migraine with aura, is strongly associated with severe cerebrovascular events. Acute migraine attack in familial hemiplegic migraine type 2 (FHM2) patients is characterized by transient hypoperfusion, which is followed by hyperperfusion of the affected hemisphere. We have previously described the mechanism responsible for FHM2-associated vasospasm of cerebral arteries and brain hypoperfusion. In this study, we characterize the mechanism for an abnormally strong cerebral vasodilator response that follows vasospasm and causes delayed hyperperfusion in a migraine attack. These defects may contribute to the cerebrovascular damage seen in patients with migraine with aura and are suggestive as future therapeutic targets.
... Otherwise, EDHF induces vascular relaxation by the opening of the K + channels which hyperpolarize the vascular smooth muscle membrane [48,50,51]. This relaxation has the characteristic of persistence, even after inhibition of the synthesis of NO and prostanoid [51,52]. Several mechanisms have been proposed to link this crucial step for good muscle hyperpolarization. ...
Article
Objective: This study aims to evaluate the vasodilatory effect of Chenopodium ambrosioides on the isolated rat aorta, and to explore its mechanism of action. Methods: The vasorelaxant effect and the mode of action of various extracts from the leaves of C. ambrosioides were evaluated on thoracic aortic rings isolated from Wistar rats. In addition, ethyl acetate and methanol fractions were analyzed, using thin-layer chromatography and high-performance liquid chromatography techniques, for their polyphenolic content. Results: The various active extracts of C. ambrosioides at four concentrations (10-3, 10-2, 10-1 and 1 mg/mL) relaxed the contraction elicited by phenylephrine, in a concentration-dependent manner. This effect seems to be endothelium-dependent, since the vasodilatory effect was entirely absent in denuded aortic rings. The vasorelaxant effect of the methanol fraction (MF) of C. ambrosioides at 1 mg/mL was also inhibited by atropine and tetraethylammonium. This effect remained unchanged by Nω-nitro-l-arginine methyl ester hydrochloride and glibenclamide. The preliminary phytochemical analysis showed that the leaves of C. ambrosioides are rich in phenolic and flavonoid derivatives. Conclusion: These results suggest that the MF of C. ambrosioides produces an endothelium-dependent relaxation of the isolated rat aorta, which is thought to be mediated mainly through stimulation of the muscarinic receptors, and probably involving the opening of Ca2+-activated potassium channels.
... Nevertheless, caution should be exercised in extrapolating the present results to other peripheral arteries, because the nature of EDH may be heterogeneous depending on the animal species and vascular bed studied. 34 Although LCZ696 did not exert an additional benefit over valsartan alone in restoring EDH-mediated responses, despite the significant reduction in SBP, this does not imply that LCZ696 is less effective in improving endothelial function associated with hypertension. Our previous findings showed that there was a significant negative relationship between the amplitude of EDH and SBP in mesenteric arteries of SHRs; however, this negative relationship disappeared when the blood pressure was lowered within normotensive range. ...
Article
Full-text available
Background We have previously demonstrated that antihypertensive treatment with renin‐angiotensin system inhibitors restores the impaired endothelium‐dependent hyperpolarization (EDH)–mediated responses in spontaneously hypertensive rats (SHRs). Herein, we investigated whether the angiotensin II receptor–neprilysin inhibitor sacubitril/valsartan (LCZ696) would improve reduced EDH‐mediated responses and whether LCZ696 would exert additional effects on endothelium‐dependent and endothelium‐independent vasorelaxation compared with an angiotensin II type 1 receptor blocker alone during hypertension. Methods and Results SHRs were treated for 3 months with either LCZ696 or valsartan, from the age of 8 to 11 months. Age‐matched, untreated SHRs and Wistar‐Kyoto rats served as controls. Membrane potentials and contractile responses were recorded from the isolated superior mesenteric arteries. Acetylcholine‐induced, EDH‐mediated responses were impaired in untreated SHRs compared with Wistar‐Kyoto rats. EDH‐mediated responses were similarly improved in the LCZ696‐ and valsartan‐treated SHRs. No difference was observed in acetylcholine‐induced, nitric oxide‐mediated relaxations among the 4 groups. Endothelium‐independent relaxations in response to a nitric oxide donor, sodium nitroprusside, and those to levcromakalim, an ATP‐sensitive K⁺‐channel opener, were similar among the 4 groups; however, the sensitivities to levcromakalim were significantly higher in both LCZ696‐ and valsartan‐treated SHRs. Conclusions LCZ696 appears to be as effective as valsartan in improving the impaired EDH‐mediated responses during hypertension. LCZ696 and valsartan exert similar beneficial effects on endothelium‐independent relaxation via enhanced sensitivity of the ATP‐sensitive K⁺ channel. However, the dual blockade of renin‐angiotensin system and neutral endopeptidase with LCZ696 does not appear to provide additional benefit over valsartan alone on vasomotor function in mesenteric arteries of SHRs.
Article
Objective This study investigated the actions of advanced glycated end‐products (AGE), their receptors (RAGE), and NAD(P)H oxidase (Nox) subtypes 1, 2, and 4 on mechanisms of endothelium‐dependent dilation of the rat cremaster muscle artery (CMA). Methods Immunofluorescence studies were used to examine expression of RAGE in rat arteries. ROS accumulation was measured using luminescence and fluorescence assays. Functional studies were performed using pressure myography. Results High levels of RAGE expression were shown in the endothelial cells of the CMA, compared with low endothelial expression in middle cerebral and mesenteric arteries and the aorta. Exogenous AGE (in vitro glycated bovine serum albumin) stimulated H2O2 accumulation in CMA, which was prevented by the RAGE antagonist FPS‐ZM1, the NAD(P)H oxidase (Nox) inhibitor apocynin and inhibited by the Nox1/4 inhibitor setanaxib, but not the Nox2 inhibitor GSK2795039. In functional studies, AGE inhibited vasodilation of CMA stimulated by acetylcholine, sodium nitroprusside, and the BKCa activator NS1619, but not adenosine‐induced dilation. FPS‐ZM1, apocynin, and setanaxib prevented the inhibitory effects of AGE on responses to acetylcholine and NS‐1619. Conclusion These observations suggest RAGE are constitutively expressed in the endothelium of the rat CMA and may be activated by AGE to stimulate Nox1/4 and ROS formation with resulting inhibition of NO and BKCa‐mediated endothelium‐dependent dilation.
Article
The transient receptor potential vallinoid type 4 (TRPV4) channel, a Ca2+‐permeable nonselective cation channel, is widely distributed in the circulatory system, particularly in vascular endothelial cells (ECs) and smooth muscle cells (SMCs). The TRPV4 channel is activated by various endogenous and exogenous stimuli, including shear stress, low intravascular pressure, and arachidonic acid. TRPV4 has a role in mediating vascular tone and arterial blood pressure. The activation of the TRPV4 channel induces Ca2+ influx, thereby resulting in endothelium‐dependent hyperpolarization and SMC relaxation through SKCa and IKCa activation on ECs or through BKCa activation on SMCs. Ca2+ binds to calmodulin, which leads to the production of nitric oxide, causing vasodilation. Furthermore, the TRPV4 channel plays an important role in angiogenesis and arteriogenesis, and is critical for tumor angiogenesis and growth, since it promotes or inhibits the development of various types of cancer. The TRPV4 channel is involved in the active growth of collateral arteries induced by flow shear stress, which makes it a promising therapeutic target in the occlusion or stenosis of the main arteries. In this review, we explore the role and the potential mechanism of action of the TRPV4 channel in the regulation of vascular tone and in the induction of neovascularization to provide a reference for future research.
Article
Obesity induces inflammation and oxidative stress, and ultimately leads to vasodilatory dysfunction in which Transient receptor potential vanilloid type 4 (TRPV4) and Nicotinamide Adenine Dinucleotide Phosphate Oxidase (Nox2) have been reported to be involved. However, little attention has been paid to the role of the TRPV4-Nox2 complex in these problems. The purpose of this study was to figure out the role of the TRPV4-Nox2 complex in obesity-induced inflammation, oxidative stress, and vasodilatory dysfunction. Using fluorescence resonance energy transfer and immunoprecipitation assays, we found enhanced TRPV4 and Nox2 interactions in obese mice. Using q-PCR, fluorescent dye dihydroethidium staining, and myotonic techniques, we found that obesity caused inflammation, oxidative stress, and vasodilatory dysfunction. Using adeno-associated viruses, we found that enhancement or attenuation of TRPV4-Nox2 interaction altered the vaso-function. Based on these findings, we found a small-molecule drug, M12, that interrupted the TRPV4-Nox2 interaction, thereby reducing inflammatory factors and reactive oxygen species production and helping to restore the vasodilatory function. In summary, our results revealed a new mechanism by which obesity-induced inflammation, oxidative stress, and vasodilatory dysfunction is caused by enhanced TRPV4-Nox2 interactions. Using M12 to interrupt the TRPV4-Nox2 interaction may have anti-inflammatory and anti-oxidative stress effects and help restore vasodilatory function and thus provide a new therapeutic approach to obesity.
Article
Full-text available
Purpose Hypertension is associated with impaired haemodynamic control mechanisms and autonomic dysfunction. Isometric exercise (IE) interventions have been shown to improve autonomic modulation and reduce blood pressure (BP) predominantly in male participants. The physiological responses to IE are unexplored in female populations; therefore, this study investigated the continous cardiac autonomic and haemodynamic response to a single bout of IE in a large female population. Methods Forty physically inactive females performed a single, individually prescribed isometric wall squat training session. Total power spectral density of heart rate variability (HRV) and associated low-frequency (LF) and high-frequency (HF) power spectral components were recorded in absolute (ms²) and normalised units (nu) pre, during and post an IE session. Heart rate (HR) was recorded via electrocardiography and baroreceptor reflex sensitivity (BRS) via the sequence method. Continuous blood pressure was recorded via the vascular unloading technique and stroke volume via impedance cardiography. Total peripheral resistance (TPR) was calculated according to Ohm’s law. Results During IE, there were significant reductions in HRV (p < 0.001) and BRS (p < 0.001), and significant increases in heart rate (p < 0.001), systolic, mean and diastolic BP (p < 0.001 for all). In recovery following the IE session, cardiac autonomic parameters returned to baseline (p = 0.974); however, total peripheral vascular resistance significantly reduced below baseline (p < 0.001). This peripheral vascular response was associated with significant reductions in systolic (−17.3 ± 16.5 mmHg, p < 0.001), mean (−18.8 ± 17.4 mmHg, p < 0.001) and diastolic BP (−17.3 ± 16.2 mmHg, p < 0.001), below baseline. Conclusion A single IE session is associated with improved haemodynamic cardiovascular responses in females. Cardiac autonomic responses return to baseline values, which suggests that alternative mechanisms are responsible for the post-exercise haemodynamic improvements in females. Future mechanistic research is required to investigate the acute and chronic effects of IE in female populations with different resting BP profiles.
Chapter
For decades neurons have been considered as the unique active constituents of the brain while glial cells were viewed solely as bystand-ers. However, now there is increasing evidence to support that glial cells are more than simple supporting cells since they appear to nourish, protect and interact with neurons.1–3 In particular the star-shaped cells called astrocytes constitute a major class of brain glial cells which possesses all the tools to receive, integrate and transmit signals to neurons. In the last few years, several reviews have focused on ionic channels in glia,4–7 but none of these have taken into consideration the channels forming gap junctions. This omission is rather surprising since astrocytes, in culture systems as well as in vivo, are presumed to be the most widely coupled cell population in the central nervous system.
Article
Acetylcholine hyperpolarizes the membrane of vascular smooth muscles by increasing potassium permeability, the response being mediated by an endothelium-derived hyperpolarizing factor (EDHF). The membrane hyperpolarization produced by EDHF is therefore one of the components contributing to endothelium-dependent relaxation of vascular smooth muscles.
Article
Aging per se is associated with abnormalities of the vascular wall linked to both structural and functional changes that can take place at the level of the extracellular matrix, the vascular smooth muscle and the endothelium of blood vessels. Endothelial dysfunction is generally defined as a decrease in the capacity of the endothelium to dilate blood vessels in response to physical and chemical stimuli. It is one of the characteristic changes that occur with age, independently of other known cardiovascular risk factors. This may account in part for the increased incidence of cardiovascular events in elderly people that can be reversed by restoring endothelial function. A better understanding of the mechanisms involved and the aetiopathogenesis of this process will help in the search for new therapeutic agents. Age-dependent alteration of endothelium-dependent relaxation seems to be a widespread phenomenon both in conductance and resistance arteries from several species. In the course of aging, there is an alteration in the equilibrium between relaxing and contracting factors released by the endothelium. Hence, there is a progressive reduction in the participation of nitric oxide and endothelium-derived hyperpolarising factor associated with increased participation of oxygen-derived free radicals and cyclo-oxygenase-derived prostanoids. Also, the endothelin-1 and angiotensin II pathways may play a role in age-related endothelial dysfunction. The use of drugs acting at different levels of these signalling cascades, including antioxidant therapy, lipid-lowering drugs and estrogens, seems to be promising.