Europe PMC

This website requires cookies, and the limited processing of your personal data in order to function. By using the site you are agreeing to this as outlined in our privacy notice and cookie policy.

Abstract 


An endothelium-derived hyperpolarizing factor (EDHF) that is distinct from nitric oxide (NO) and prostanoids has been widely hypothesized to hyperpolarize and relax vascular smooth muscle following stimulation of the endothelium by agonists. Candidates as diverse as K(+) ions, eicosanoids, hydrogen peroxide and C-type natriuretic peptide have been implicated as the putative mediator, but none has emerged as a 'universal EDHF'. An alternative explanation for the EDHF phenomenon is that direct intercellular communication via gap junctions allows passive spread of agonist-induced endothelial hyperpolarization through the vessel wall. In some arteries, eicosanoids and K(+) ions may themselves initiate a conducted endothelial hyperpolarization, thus suggesting that electrotonic signalling may represent a general mechanism through which the endothelium participates in the regulation of vascular tone.

Free full text 


Logo of brjpharmLink to Publisher's site
Br J Pharmacol. 2004 Mar; 141(6): 881–903.
Published online 2004 Mar 18. https://doi.org/10.1038/sj.bjp.0705698
PMCID: PMC1574270
PMID: 15028638

Endothelium-dependent smooth muscle hyperpolarization: do gap junctions provide a unifying hypothesis?

Abstract

An endothelium-derived hyperpolarizing factor (EDHF) that is distinct from nitric oxide (NO) and prostanoids has been widely hypothesized to hyperpolarize and relax vascular smooth muscle following stimulation of the endothelium by agonists. Candidates as diverse as K+ ions, eicosanoids, hydrogen peroxide and C-type natriuretic peptide have been implicated as the putative mediator, but none has emerged as a ‘universal EDHF'. An alternative explanation for the EDHF phenomenon is that direct intercellular communication via gap junctions allows passive spread of agonist-induced endothelial hyperpolarization through the vessel wall. In some arteries, eicosanoids and K+ ions may themselves initiate a conducted endothelial hyperpolarization, thus suggesting that electrotonic signalling may represent a general mechanism through which the endothelium participates in the regulation of vascular tone.

Keywords: EDHF, connexin, cAMP, potassium, epoxyeicosatrienoic acid, hydrogen peroxide, C-type natriuretic peptide

Introduction

Endothelial cells contribute to vascular control by releasing vasodilator prostanoids and NO when stimulated by agonists or fluid shear stress. Such stimuli also evoke an endothelium-dependent hyperpolarization of smooth muscle cells that is independent of NO and prostanoids, a phenomenon first described in guinea-pig, porcine and canine arteries exposed to acetylcholine (ACh) or bradykinin (Bolton et al., 1984; Bény & Brunet, 1988; Chen et al., 1988; Feletou & Vanhoutte, 1988). These observations led to the hypothesis that a distinct endothelium-derived hyperpolarizing factor, or EDHF, transfers across the extracellular space to modulate smooth muscle membrane potential (Chen et al., 1991; Mombouli et al., 1996; Popp et al., 1996; Gebremedhin et al., 1998). Despite over a decade of research, however, no single agent has emerged unambiguously as a ‘universal EDHF' that mediates relaxation following release from the endothelium. Indeed, bioassay experiments with sandwich preparations constructed from closely apposed endothelium-intact and -denuded arterial strips, in which the donor endothelium and detector smooth muscle are electrically uncoupled, often fail to demonstrate relaxation in response to agonists such as ACh in the presence of an inhibitor of the constitutive endothelial NO synthase (eNOS) (Hecker et al., 1994; Plane et al., 1995; Chaytor et al., 1998; 2002; 2003; Hutcheson et al., 1999; Kagota et al., 1999). This raises the possibility that the EDHF phenomenon may involve passive electrotonic spread of hyperpolarization from the endothelium to smooth muscle cells via gap junctions, with relaxation being secondary to the closing of voltage-operated Ca2+ channels and associated reductions in the influx of extracellular Ca2+ ions that sustains contraction. Although experiments with agents that uncouple direct cell–cell communication nonspecifically, such as the long-chain alcohol heptanol and the anaesthetic halothane, initially failed to provide consistent evidence that gap junctions play a role in EDHF-type relaxations (Bény & Pacicca, 1994; Kühberger et al., 1994; Zygmunt & Hogestatt, 1996), there is growing evidence that more targeted inhibitors of direct cell–cell coupling do indeed attenuate endothelium-dependent smooth muscle hyperpolarization. A clear understanding of the participating mechanisms has nevertheless been clouded by the wide variety of signalling pathways that have been implicated in the EDHF phenomenon, raising questions as to which of the mechanisms involved might be central and which might be interactive consequences, and by the existence of species and vessel differences. This review will attempt to summarize the large matrix of data now available, and develop an integrative hypothesis that may reconcile some of the apparently conflicting observations reported in the literature.

Endothelial hyperpolarization: K+ channel activation by intracellular Ca2+

Agonists that evoke EDHF-type relaxations cause a rapid initial shift in the membrane potential of endothelial cells towards the reversal potential for K+ (~−80 mV), followed, variably, by stabilization some 10–20 mV below baseline or a slow return towards control that may sometimes be accompanied by an overshoot depolarization (Mehrke & Daut, 1990; Marchenko & Sage, 1993; Ohashi et al., 1999). The hyperpolarizing response is driven by the opening of Ca2+-activated K+ channels (KCa), as is evidenced by: (i) an associated efflux of Rb+ ions from suitably loaded endothelial cells (Gordon & Martin, 1983), (ii) an inverse relationship between the amplitude of agonist-induced changes in endothelial membrane potential and the prevailing concentration of extracellular K+ ions, such that hyperpolarization is converted to depolarization at [K+]0 >25–50 mM (Mehrke & Daut, 1990) and (iii) attenuated hyperpolarization in the presence of peptide toxins such as apamin (a selective inhibitor of small-conductance channels, SKCa), charybdotoxin (an inhibitor of intermediate and large-conductance channels, IKCa and BKCa, and certain voltage-dependent K+ channel subtypes, Kv) and iberiotoxin (a selective inhibitor of BKCa channels) (Edwards et al., 1998; Frieden et al., 1999; Ohashi et al., 1999). Activation of these different KCa subtypes may be agonist-specific and vary between native and cultured endothelial cells, in which KCa expression can alter during passage. Substance P and bradykinin, for example, hyperpolarize the native endothelium of porcine coronary arteries by opening SKCa and IKCa channels (Edwards et al., 2000; Bychkov et al., 2002). By contrast, cultured porcine coronary endothelial cells additionally express an iberiotoxin-sensitive BKCa channel that is activated by bradykinin, but not by substance P (Frieden et al., 1999). The participation of separate KCa subtypes explains observations that more than one toxin may be required to inhibit endothelial hyperpolarization completely. ACh-induced hyperpolarization of the endothelium of the rabbit aortic valve, for example, consists of a transient attenuated by charybdotoxin, but not apamin, and a sustained component that is partially attenuated by either toxin alone, with hyperpolarization being converted into depolarization by their combination (Ohashi et al., 1999).

The crucial physiological stimulus for the agonist-induced KCa channel activation that underpins endothelial hyperpolarization is an elevation in free cytosolic calcium, [Ca2+]i. Since the endothelium is devoid of voltage-gated Ca2+ channels and electrically nonexcitable, hyperpolarization will itself tend to promote elevations in [Ca2+]i by enhancing the electrochemical gradient that drives transmembrane Ca2+ influx (Luckhoff & Busse, 1990; Kamouchi et al., 1999). However, it is now established that the principal mechanism that sustains the opening of endothelial KCa channels that follows agonist stimulation is capacitative Ca2+ entry (CCE) via membrane channels whose functionality is closely linked to depletion of the endoplasmic reticulum (ER) Ca2+ store (Marchenko & Sage, 1993; Sedova et al., 2000; Nilius & Droogmans, 2001). Agonist-evoked endothelial hyperpolarization may consequently be suppressed by lowering extracellular [Ca2+], by buffering elevations in [Ca2+]i with a chelator, and by blocking CCE with 2-aminoethoxydiphenyl borate (2-APB), which reversibly inhibits store-operated Ca2+ channels (Chen & Suzuki, 1990; Marchenko & Sage, 1993; Frieden et al., 1999; Ohashi et al., 1999; Iwasaki et al., 2001; Bishara et al., 2002; Xie et al., 2002). In the converse sense, depletion of the ER by agents that prevent Ca2+ uptake by the SERCA pump, such as cyclopiazonic acid and thapsigargin, promote receptor-independent activation of endothelial KCa channels (Pasyk et al., 1995; Davis & Sharma, 1997; Fukao et al., 1997a). Agonist-induced depletion of the ER store is classically attributed to the activation of phospholipase C (PLC) via tyrosine phosphorylation, followed by the formation of inositol 1,4,5-trisphosphate (InsP3), which releases Ca2+ from the store (Fleming et al., 1996). However, studies with triple InsP3 receptor knockout B-lymphocytes suggest that additional pathways could also be involved, since CCE is not impaired in such cells (Ma et al., 2001). The nature of the store-operated Ca2+ influx mechanism thus remains controversial, but in endothelial cells may involve channels constructed from TRP proteins (Nilius et al., 2003). Whether the opening of store-operated channels is effected by direct mechanical coupling with the InsP3 receptor or a diffusible cytosolic ‘calcium influx factor' also remains the subject of debate (Kiselyov et al., 1998; Trepakova et al., 2000). Indeed, in endothelial cells there is evidence to support both hypotheses, since disruption of the cytoskeleton inhibits CCE, and store depletion may induce the formation of eicosanoid metabolites of arachidonic acid that promote an 2-APB-sensitive entry of Ca2+ ions via store-operated channels (Graier et al., 1995; Bishara et al., 2002; Xie et al., 2002; see below).

It has also become evident that the open-state probability of endothelial KCa channels can be modulated by secondary signalling events originating in the vascular media. Direct intercellular coupling via myoendothelial gap junctions may thus allow diffusion of InsP3 and/or Ca2+ ions from activated smooth muscle cells into the endothelium, with the resulting elevation in [Ca2+]i then promoting KCa channel-mediated endothelial hyperpolarization and depressing contraction through negative feedback (Dora et al., 1997; 2000; Yashiro & Duling, 2000; Budel et al., 2001). In theory, the ability of smooth muscle cells to modulate endothelial Ca2+ homeostasis could also contribute to the apparent paradox that KCa channel inhibitors reduce agonist-induced elevations in endothelial [Ca2+]i in isolated cells (Kamouchi et al., 1999), but not in intact arteries (Yamanaka et al., 1998; Bolz et al., 1999; Ghisdal & Morel, 2001; Ungvari et al., 2002).

Relationship between endothelial hyperpolarization and smooth muscle relaxation

The electrical smooth muscle response that occurs during the EDHF phenomenon closely parallels that in the endothelium, exhibiting an initial shift towards the reversal potential for K+ followed by a slow return towards baseline (e.g. Figure 1). Evidence that activation of endothelial KCa channels by capacitative Ca2+ entry provides the driving force for relaxation is provided by observations that: (i) EDHF-type relaxations to ACh can be abolished by selective intimal application of KCa channel blockers, whereas adventitial application is without effect (Doughty et al., 1999), and (ii) that EDHF-type relaxations evoked by SERCA inhibitors such as cyclopiazonic acid are attenuated by KCa channel blockers (Dora et al., 2001). Formation of InsP3, followed by depletion of the ER Ca2+ store, is likely to underpin the elevation in endothelial [Ca2+]i that sustains agonist-induced smooth muscle hyperpolarization, since the PLC inhibitor U73122 attenuates subintimal hyperpolarizations evoked by ACh in the rat mesenteric artery (Fukao et al., 1997a), and attenuates EDHF-type relaxations/dilations in porcine coronary, rabbit mesenteric, guinea-pig carotid and rat middle cerebral arteries (Weintraub et al., 1995; Hutcheson et al., 1999; Quignard et al., 2002; You et al., 2002) and the perfused rat heart (Fulton et al., 1996). The ability of KCa channel inhibitors to attenuate EDHF-type responses without affecting agonist-induced increases in endothelial [Ca2+]i in intact arterial preparations (see above) suggests that [Ca2+]i does not serve as a primary activator of a putative Ca2+-dependent ‘EDHF synthase'. Indeed, in rat middle cerebral arteries, the directly acting IKCa agonist 1-EBIO, whose action is independent of PLC, evokes EDHF-type relaxations without elevating endothelial [Ca2+]i above resting levels (Quignard et al., 2002; Marrelli et al., 2003).

An external file that holds a picture, illustration, etc.
Object name is 141-0705698f1.jpg

(a) Traces showing changes in subintimal smooth muscle cell potential in endothelium-intact strips of rabbit iliac artery impaled via the intima. ACh induced an initial shift towards the reversal potential for K+, followed by a sustained hyperpolarization that was attenuated by 37,43Gap27 (600 μM). (b) Confocal imaging of dye transfer from the endothelium into the media of rabbit femoral arteries following intraluminal perfusion and intimal loading with the cell-permeant tracer calcein AM. Subsequent cleavage of the acetoxymethyl moieties that permit endothelial uptake of this tracer results in a reduction in molecular mass from ~1000 to ~600 Da and allows diffusion of calcein (which is polar) through myoendothelial gap junctions. 37,43Gap27 (600 μM) caused retention of calcein within the intima, thereby attenuating diffusion of the dye into subjacent smooth muscle cells (from Griffith et al., 2002).

Smooth muscle hyperpolarizations of the magnitude observed during the EDHF phenomenon are capable of causing marked reductions in vascular tone, because the voltage-dependent Ca2+ channels that maintain contraction are highly sensitive to the membrane potential (Nelson et al., 1990). Measurements during EDHF-type relaxations of pharmacologically constricted hamster skeletal muscle resistance arteries, rat renal arterioles and porcine coronary arteries thus reveal large and rapid falls in smooth muscle [Ca2+]i to baseline levels, although, as with the associated electrical response, these may not always be sustained (Bolz et al., 1999; Marchetti et al., 2001; Ohnishi et al., 2001). While administration of apamin may sometimes be sufficient to abolish EDHF-type relaxations (Yamakawa et al., 1997; Ayajiki et al., 2000), in most vessels apamin and charybdotoxin are individually each only partially effective, and their co-administration necessary to attenuate relaxation completely (Edwards et al., 1998; Doughty et al., 1999). This observation parallels findings in isolated endothelial cells (see above), and has become widely regarded as a hallmark of the EDHF phenomenon. In many arteries, it is likely to reflect the dual participation of endothelial SKCa and IKCa channels, because the selective BKCa channel inhibitor iberiotoxin may be completely ineffective, even in combination with apamin, for example, in rat mesenteric, hepatic and renal arteries and guinea-pig coronary and basilar arteries (Rapacon et al., 1996; Petersson et al., 1997; Plane et al., 1997; Chataigneau et al., 1998a; Eckman et al., 1998; Edwards et al., 1998; Yamanaka et al., 1998). Furthermore, in rat carotid and mesenteric arteries, the sole involvement of SKCa and IKCa channels has been confirmed by using apamin in combination with the selective IKCa inhibitors TRAM-34 and TRAM-39 (Wulff et al., 2000; 2001; Eichler et al., 2003; Hinton & Langton, 2003). There may nevertheless be exceptions to the ‘general' rule that SKCa and IKCa channels are the only KCa subtypes capable of participating in the EDHF phenomenon. BKCa channels can be detected immunohistochemically in the endothelium of rat skeletal muscle arterioles (Ungvari et al., 2002) and by patch-clamp techniques in the endothelium of the porcine aorta and renal artery (Papassotiriou et al., 2000; Brakemeier et al., 2003), and the ability of iberiotoxin to inhibit EDHF-type relaxations in rabbit renal and femoral arteries could reflect the expression of BKCa channels, which has been documented electrophysiologically in freshly isolated endothelial cells from this species (Rusko et al., 1992; Kagota et al., 1999; Kwon et al., 1999; Yousif et al., 2002).

Despite the close relationship between endothelial and smooth muscle hyperpolarization, the nature of the pathways that allow changes in endothelial membrane potential to be translated into smooth muscle hyperpolarization, reductions in smooth muscle [Ca2+]i and relaxation remain controversial. As outlined above, there are two principal hypotheses, namely, electrotonic signalling via gap junctions and diffusion of freely transferable mediators across the extracellular space.

Gap junctional communication

Gap junctions are formed by the docking of two hemichannels (called connexons) at points of cell–cell contact, with each hemichannel being constructed from six connexin (Cx) protein subunits that traverse the cell membrane four times to expose two extracellular peptide loops (Kumar & Gilula, 1996; Perkins et al., 1998). Interdigitation of the extracellular loops of apposing connexons, followed by a 30° rotation, creates an aqueous central pore that allows intercellular diffusion of ions and signalling molecules <1 kDa in size, thereby conferring electrical continuity between coupled cells (see the schematic representation in Figure 2). Individual gap junctions may subsequently aggregate in plaques that consist of focal clusters of many hundreds of units in the cell membrane and whose characteristic pentalaminar appearance can be visualized by electron microscopy at points of hetero- and homocellular contact between endothelial and smooth muscle cells (Spagnoli et al., 1982; Sandow & Hill, 2000). The formation of such structures is thought to facilitate cooperative interactions between their component gap junctions and thereby strongly enhance intercellular signalling (Bukauskas et al., 2000). Indeed, in communication-incompetent Neuro-2a, HeLa and RIN cells, stably transfected to express a fluorescent connexin protein, the extent of dye transfer and electrical communication between cell pairs correlates closely with plaque size, and coupling is absent in the absence of detectable plaques, even when connexin protein is diffusely present in the cell membrane (Bukauskas et al., 2000). Three main connexin subtypes, classified as Cxs 37, 40 and 43 according to molecular mass in kDa, are widely distributed in the vasculature, and some vessels may also express Cx45. Immunostaining of the vessel wall with specific antibodies reveals the distinctive punctate appearance of plaques containing these connexins, with protein expression generally being more abundant in the endothelium than the media, and different connexin subtypes often co-localizing in the same plaque (Haas & Duling, 1997; Hong & Hill, 1998; Yeh et al., 1998; Chaytor et al., 2001; Li & Simard, 2001; Berman et al., 2002; Rummery et al., 2002; Ujiie et al., 2003). In addition to homotypic gap junctions, in which each connexon contains the same connexin subtype, heterotypic channels, in which each connexon is constructed from a different connexin subtype, and heteromeric channels, in which each connexon contains mixtures of subtypes, may therefore be present in the vascular wall. The existence of such hybrid gap junctions has been confirmed electrophysiologically in cultured smooth muscle cells and isolated arteries by patch-clamp identification of channels with complex conductances consistent with the formation from more than one connexin protein (He et al., 1999; Li & Simard, 1999; 2001; Wang et al., 2001; Yamazaki & Kitamura, 2003).

An external file that holds a picture, illustration, etc.
Object name is 141-0705698f2.jpg

Schematic representation showing the structure of a connexin protein and how six such elements form a connexon. Docking of connexons from apposing cells results in the formation of an aqueous pore that allows the transfer of ions and small signalling molecules between coupled cells. Each connexin possesses four transmembrane segments (M1–4). Also highlighted are the Gap 26 and Gap 27 domains of the first and second extracellular connexin loops which are conserved in man, rat and mouse. Differences in the amino-acid sequences of the Gap 26 and Gap 27 domains of the three major vascular connexins allow designation as 37,40Gap26, 43Gap26, 37,43Gap27 and 40Gap27. Synthetic peptides possessing these sequences inhibit direct intercellular communication in a connexin-specific fashion.

Attempts to demonstrate direct communication between endothelial and smooth muscle cells by microinjection of fluorescent tracer dyes into individual endothelial cells are often unsuccessful (Segal & Bény, 1992; Little et al., 1995; Jiang et al., 2001; Yamamoto et al., 2001). This observation may, at least in part, reflect the preferential diffusion of dye within the endothelial monolayer via large inter-endothelial plaques, rather than radial diffusion into the media via myoendothelial gap junction plaques which are much smaller and less numerous (Haas & Duling, 1997; Sandow & Hill, 2000). Loading the entire endothelial layer with a lipophilic tracer such as calcein AM, which is cleaved intracellularly after uptake, can overcome this ‘sink–source' problem by allowing diffusion of the polar fluorescent product calcein from the endothelium into the media (Figure 1). It should be appreciated, however, that failure to demonstrate dye coupling between adjacent cells does not necessarily imply the absence of electrical continuity, since the efficacy of dye transfer is influenced by the molecular mass and charge of the specific tracer employed, as well as the nature of the connexin subtypes expressed in the cells under study (Little et al., 1995; Kruger et al., 2002). Indeed, in hamster cheek pouch arterioles, hamster skeletal muscle feed arteries and guinea-pig mesenteric arterioles, electrophysiological studies have confirmed that endothelial hyperpolarizations evoked by ACh or direct current injection can be detected synchronously in smooth muscle cells, and conversely, that action potentials originating in smooth muscle cells are conducted to the endothelium (Segal & Bény, 1992; Emerson & Segal, 2000; Coleman et al., 2001a; Yamamoto et al., 2001). In porcine coronary artery and rat aorta oscillations in smooth muscle membrane potential also drive synchronous electrical fluctuations in the endothelium, generalizing such observations to conduit vessels (von der Weid & Bény, 1993; Marchenko & Sage, 1994). In guinea-pig mesenteric arterioles, gap junctions coupling endothelial and smooth muscle cells behave as simple ohmic resistors without rectification, since electrical responses transmitted in either direction are reduced by 10–20% in amplitude without alteration in their dynamic temporal form (Yamamoto et al., 2001). In rat pial arterioles, however, dual-electrode macroscopic current recordings from adjacent endothelial and smooth muscle cells demonstrate partial rectification, as evidenced by an asymmetric transjunctional conductance–voltage relationship that is likely to reflect the presence of heterotypic and/or heteromeric myoendothelial gap junctions (Yamazaki & Kitamura, 2003).

Despite compelling experimental evidence for direct electrical coupling between the endothelium and the media, it has nevertheless been argued that nonregenerative spread of endothelial hyperpolarization into subjacent smooth muscle cells will fail to contribute to relaxation in conduit arteries, on the basis that the large relative mass of the media will dissipate passive electrical signals originating from the endothelium by acting as a current sink (Bény, 1999). Evidence to support the contrary view is summarized in the following sections.

Connexin-mimetic peptides

Minor variations in the amino-acid sequences of the first and second extracellular loops of Cxs 37, 40 and 43 permit the synthesis of short synthetic peptides that are homologous to the conserved Gap 26 and 27 domains of these proteins and are capable of interrupting gap junctional communication following relatively short incubations (15–40 min) at concentrations in the range 300 μM–1 mM (Chaytor et al., 1997; 1998; 1999; 2001; Dora et al., 1999; Berman et al., 2002; Griffith et al., 2002; Figure 2). Experiments with cultured cells suggest that the inhibitory effects of such peptides against cell–cell coupling are connexin specific. In confluent COS fibroblasts, for example, in which the only connexin protein expressed is Cx43, intercellular transfer of the dye Lucifer yellow is attenuated by 37,43Gap 27, a peptide possessing homology with the Gap 27 domain of the second extracellular loop of Cx43 (and also Cx37), but not by 40Gap 27, which is homologous to the corresponding domain of Cx40 and differs by just three amino acids (Chaytor et al., 1999). Since similar concentrations of 37,43Gap 27 and 43Gap 26 are equally effective in attenuating dye transfer between HeLa cells transfected to express Cx43, it is apparent that first and second loop peptides may be used interchangeably to inhibit gap junctional communication (Berman et al., 2002).

Evidence that the role of direct endothelial–smooth muscle coupling is central to the EDHF phenomenon has been provided by observations that connexin-mimetic peptides attenuate subintimal smooth muscle hyperpolarizations and associated relaxations evoked by ACh, ATP, substance P and bradykinin in large arteries and veins from the rabbit, rat, pig and guinea-pig (Chaytor et al., 1998; 1999; 2001; 2003; Dora et al., 1999; Edwards et al., 1999; 2000; Griffith & Taylor, 1999; Hutcheson et al., 1999; Doughty et al., 2000; Berman et al., 2002; Griffith et al., 2002; Sandow et al., 2002; Xu et al., 2002; Ujiie et al., 2003). Although their molecular mechanism of action remains to be established, 43Gap 26 and 37,43Gap 27 do not perturb the physical stability of plaques constructed from Cx43, despite attenuating dye transfer (Berman et al., 2002). This suggests that connexin-mimetic peptides modulate channel gating rather than connexon docking, and is consistent with an ability to attenuate EDHF-type relaxations in a reversible fashion, with coupling being restored on peptide washout (Chaytor et al., 1998; 2001). An action downstream of specific membrane receptors has been confirmed by observations that 37,43Gap 27 attenuates receptor-independent EDHF-type relaxations evoked by the SERCA inhibitor cyclopiazonic acid, and does not impair ACh-induced endothelial NO synthesis in sandwich bioassay experiments (Chaytor et al., 1998). Importantly, connexin-mimetic peptides do not suppress endothelial hyperpolarization directly, and they do not influence mechanical smooth muscle responses to exogeneous nitrovasodilators, KATP channel openers or constrictor agonists (Chaytor et al., 1997; 1998; 2001; Dora et al., 1999; Richards et al., 2001; Sandow et al., 2002; Ujiie et al., 2003). In addition to attenuating electrotonic signalling via myoendothelial gap junctions, 37,43Gap 27 impedes the transfer of fluorescent tracer dye from the endothelium into the media of rabbit ilio-femoral arteries (Figure 2), thus providing evidence that connexin-mimetic peptides are capable of interrupting direct chemical signalling via myoendothelial gap junctions (Griffith et al., 2002).

Connexin-mimetic peptides also uncouple vascular smooth muscle cells (Chaytor et al., 1997), so that in some arteries their inhibitory effects against the EDHF phenomenon may reflect an ability to attenuate electrotonic signalling within the media. In porcine coronary arteries stimulated by substance P, for example, comparative measurements of subintimal and adventitial smooth muscle membrane potential suggest that 37,43Gap 27 preferentially depresses the relay of agonist-induced hyperpolarizing signals originating in the endothelium through successive layers of smooth muscle cells (Edwards et al., 2000). Whole-cell patch clamping of smooth muscle cells in the media of rat cerebral pial arterioles has also demonstrated that 37,43Gap 27 markedly impairs current flow to neighbouring cells, and thereby increases smooth muscle membrane input resistance (Yamazaki & Kitamura, 2003). Whether direct electrical coupling within the endothelium contributes to the EDHF phenomenon remains to be established. However, it may be speculated that the presence of large inter-endothelial gap junction plaques will allow this monolayer to function as a low-resistance current source, thereby facilitating transmission of hyperpolarization into the media via high-resistance myoendothelial gap junctions (Haas & Duling, 1997; Sandow & Hill, 2000; Chaytor et al., 2001; Yamamoto et al., 2001). In support of this concept, pharmacological uncoupling of gap junctions increases the input electrical resistance of the endothelium of guinea-pig mesenteric arterioles by ~150-fold (Yamamoto et al., 1998).

Although connexin-mimetic peptides targeted against a single connexin subtype are capable of causing substantial reductions in EDHF-type responses in specific arteries, there is emerging evidence that peptide combinations directed against more than one subtype are generally more effective (Table 1). This is likely to reflect variations in the size, frequency, location and connexin composition of the gap junction plaques present in different vessels, and an ability of channels constructed from different connexin proteins to compensate for each other functionally. In confluent rat aortic smooth muscle A7r5 cells, which are highly coupled by numerous plaques constructed from Cxs 40 and 43, dye transfer is unaffected by 40Gap 27 and 43Gap 26 administered individually at concentrations of 600 μM, but is inhibited by dual administration of the peptides at the same net overall concentration, that is, 300 μM each (Chaytor et al., 2001). Analogously, in the rabbit iliac artery, in which immunostaining reveals plaques containing Cxs 37 and 40 in the endothelium and Cxs 40 and 43 in the media, the triple peptide combination 37,43Gap 27+43Gap 26+40Gap 27 (at 300 μM each) effectively abolishes subintimal EDHF-type smooth muscle hyperpolarizations evoked by the Ca2+ ionophore A23187, whereas 37,43Gap 27 alone attenuates by ~65% at an equivalent total peptide concentration of 900 μM (Chaytor et al., 2003). These observations suggest that more than one connexin subtype contributes to myoendothelial coupling in this vessel. The inhibitory effects of single peptides and peptide combinations against ACh-evoked relaxations in rabbit ear, rabbit middle cerebral and rat hepatic arteries are also complex, and may additionally reflect heterogeneity in the patterns of connexin protein expressed in the media. In these vessels, endothelial gap junction plaques contain Cxs 37, 40 and 43, albeit in slightly different ratios, whereas the connexin composition of smooth muscle plaques is more vessel specific. In rabbit ear arteries, Cx43 is the sole subtype detectable in medial plaques on immunostaining and 37,43Gap 27 and 43Gap 26 both abolish EDHF-type relaxations, consistent with a dominant role for gap junctions containing Cx43 (Berman et al., 2002). In rabbit middle cerebral arteries, Cxs 40 and 43 are both present in the media, and 37,43Gap 27 and 40Gap 27 partially attenuate relaxation, with responses being almost eliminated by the two peptides in combination (Ujiie et al., 2003). In the rat hepatic artery, Cxs 37 and 43 are both highly expressed in medial plaques and EDHF-type relaxations are unaffected by individual peptides, whereas a combination simultaneously targeting Cxs 40 and 43 (43Gap 26+40Gap 27) attenuates by ~50% and the triple combination 37,43Gap 27+43Gap 26+40Gap 27 inhibits relaxation almost completely (Chaytor et al., 2001). The participation of different connexin subtypes in the EDHF phenomenon is also evident in vivo, as intrarenal infusion of 40Gap 27 and 37,43Gap 27 each attenuate the increase in renal blood flow induced by ACh in anaesthetized rats (De Vriese et al., 2002).

Table 1

Semiquantitative analysis of connexin expression and the effects of connexin-mimetic peptides on Lucifer yellow dye in rat smooth muscle cells and EDHF-type hyperpolarizations and relaxations in rabbit and rat arteries

Connexins identified/peptide inhibitors Cx37Cx40Cx4337,43Gap 2743Gap 2640Gap 2737,43Gap27+40Gap 2743Gap 26+40Gap 2737,43Gap 26+43Gap 26+40Gap 27
A7r5 rat aortic myocytes dye transfer ++++ −(600 μM)−(600 μM) **(600 μM) 
Rabbit iliac artery A23187 hyperpolarizationE++*(900 μM)  **(900 μM) ***(900 μM)
 SM++      
Rabbit ear artery ACh relaxationE+++***(300 μM)***(300 μM)    
 SM+      
Rabbit middle cerebral artery ACh relaxationE+++*(300 μM) *(300 μM)***(600 μM)  
 SM++**(600 μM) **(600 μM)   
Rat hepatic artery ACh relaxationE+++−(600 μM)−(600 μM)−(600 μM)*(600 μM)*(600 μM)***(900 μM)
 SM++++      
+/− denote the presence/absence of Cxs 37, 40 and 43 in endothelial (E) and smooth muscle (SM) gap junction plaques as visualized by antibody staining. Functional inhibition by connexin-mimetic peptides (denoted as */−) was estimated at the total peptide concentrations indicated in brackets. The data provide evidence that more than one connexin subtype may contribute to electrotonic signalling in the vessel wall and that the efficacy of connexin-mimetic peptides as inhibitors of the EDHF phenomenon in different vessels is likely to reflect heterogeneity in the size, frequency, location and connexin composition of gap junction plaques. Data compiled from Chaytor et al. (2001, 2003); Berman et al. (2002); Ujiie et al. (2003).

Heterogeneous connexin expression could also explain more major differences in electrotonic signalling. In the rat femoral artery, for example, some workers have documented large EDHF-type responses to ACh (Savage et al., 2003), whereas others have reported an almost complete absence of relaxation (Zygmunt et al., 1995; Sandow et al., 2002). Notably, the absence of a conducted subintimal smooth muscle response following hyperpolarization of the endothelium by ACh has been correlated with a lack of myoendothelial gap junctions, as assessed by serial electron microscopy in this vessel (Sandow et al., 2002). Such variability could conceivably reflect differences in strain or age, since the frequency of endothelial–smooth muscle contacts made via fenestrations in the internal elastic lamina decreases during maturation (Kristek & Gerova, 1997). Major regional variations are also evident in porcine vessels: in the ciliary artery, agonist-induced endothelial hyperpolarization is only occasionally transmitted to immediately subjacent smooth muscle cells, and is consequently undetectable deep in the media, in marked contrast to the porcine coronary artery (Pacicca et al., 1996; Bény et al., 1997; Edwards et al., 2000). Differences in connexin expression could also influence the space constant for propagation of hyperpolarization through the media (i.e., the distance over which a change in smooth muscle membrane potential decrements by a factor of 1/e to ~37% of its original value). This parameter can be estimated to be of the order of 1–2 mm in porcine coronary and rabbit iliac arteries, but just 50–100 μm in guinea-pig mesenteric arterioles denuded of their endothelium (Pacicca et al., 1996; Yamamoto et al., 2001; Griffith et al., 2002).

Glycyrrhetinic acid derivatives

Gap junctional communication can also be inhibited by the 18α- and 18β-isoforms of glycyrrhetinic acid (GA), a lipophilic steroidal aglycone derived from glycyrrhizic acid that is found in the liquorice root glycyrrhizia glabra, and by carbenoxolone, a water-soluble hemisuccinate derivative of 18β-GA (Davidson et al., 1986; Davidson & Baumgarten, 1988). EDHF-type relaxations of rabbit superior mesenteric arteries and jugular veins evoked by ACh are attenuated by these compounds, with rank inhibitory potencies being 18β-GA>18α-GA[dbl greater-than sign]carbenoxolone in the superior mesenteric artery, suggesting that the decreased lipophilicity of carbenoxolone reduces its ability to inhibit gap junctional communication in the vessel wall (Taylor et al., 1998; Griffith & Taylor, 1999; Chaytor et al., 2000). Consistent with an action distal to the occupation of specific membrane receptors, 18α-GA also attenuates EDHF-type relaxations evoked by the SERCA inhibitor cyclopiazonic acid (Taylor et al., 1998). Evidence from a spectrum of rat and guinea-pig arteries suggests that GA derivatives, like connexin-mimetic peptides, inhibit EDHF-type relaxations by impairing the electrotonic spread of endothelial hyperpolarization into and through the vascular media via gap junctions (Edwards et al., 1999; Yamamoto et al., 1999; Jiang et al., 2001; Yamazaki & Kitamura, 2003). Of the three commonly employed derivatives, 18α-GA has emerged as the most suitable pharmacological probe for assessing the contribution of gap junctions to endothelium-dependent relaxation. In rabbit arteries, 18α-GA inhibits EDHF-type responses at concentrations that do not affect smooth muscle tone, whereas 18β-GA and carbenoxolone both relax smooth muscle directly, and carbenoxolone functionally enhances NO activity (Dembinska-Kiec et al., 1991; Chaytor et al., 2000). There is also evidence that 18β-GA and carbenoxolone nonspecifically depress endothelial hyperpolarization (Tare et al., 2002), although this has not been a universal finding, and does not appear to be a problem with 18α-GA (Edwards et al., 1999; Murai et al., 1999; Yamamoto et al., 1999; Jiang et al., 2001). Nonspecific ‘toxic' effects of GA derivatives are also more pronounced with the β than the α configuration in nonvascular cells (Davidson et al., 1986; Davidson & Baumgarten, 1988; Goldberg et al., 1996).

In contrast to the reversible action of connexin-mimetic peptides, inhibition of EDHF-mediated relaxations of rabbit arteries by 18α-GA becomes irreversible following 1 h incubation (Chaytor et al., 1998; 2000), an observation that may reflect the ability of GA derivatives to disrupt gap junction plaques. In liver and alveolar epithelial cells expressing Cx43, GA derivatives cause time- and concentration-dependent dephosphorylation of this connexin subtype, plaque disassembly and internalization, with progressive reductions in the expression of Cx43 becoming evident as exposure times are extended beyond 30 min (Guan et al., 1996; Guo et al., 1999). By contrast, the initial interruption of intercellular communication is rapid (within 15–30 min), reversible, and not associated with changes in the integrity of gap junction plaques or the phosphorylation status of connexin proteins (Guan et al., 1996; Guo et al., 1999). The time course of these observations correlates closely with the ability of 18α-GA to decrease the macroscopic junctional current that flows between pairs of coupled rat pial arteriolar smooth muscle cells following depolarizing voltage steps and the number of unitary channel events that can be detected as the cells become progressively uncoupled (Yamazaki & Kitamura, 2003). The molecular mechanisms that underlie the action of GA derivatives remain unknown, and a definitive link between connexin dephosphorylation and the stability of gap junction plaques remains to be established. Indeed, in C6 glioma cells, the water-soluble hemisuccinate form of 18α-GA causes plaque disaggregation without affecting the phosphorylation status of Cx43 (Goldberg et al., 1996), and 18α-GA itself attenuates dye transfer between HeLa cells transfected to express functional gap junctions constructed from Cx26, a connexin subtype that is not regulated by phosphorylation (George et al., 1998). Another lipophilic compound, palmitoleic acid, is also known to interrupt gap junctional communication and inhibit EDHF-type hyperpolarizations and relaxations, although again its mechanism of action remains to be delineated (Harris et al., 2000; Kenny et al., 2002; Ungvari et al., 2002).

Permissive role of cyclic AMP

In many nonvascular cell types, elevations in cAMP levels enhance cell–cell coupling via gap junctions constructed from Cxs 40 and 43 through poorly understood mechanisms that involve connexin phosphorylation by protein kinase A (PKA) and/or rapid recruitment of connexin protein to the cell membrane (Burghardt et al., 1995; Chanson et al., 1996; Abudara et al., 2000; Paulson et al., 2000; van Rijen et al., 2000; Gladwell & Jefferys, 2001; Grazul-Bilska et al., 2001). The ability of this cyclic nucleotide to modulate intercellular communication is also evident in the vascular wall as 8-bromo-cAMP (a cell-permeant analogue) and the cAMP phosphodiesterase inhibitor isobutylmethylxanthine (IBMX) enhance the diffusion of fluorescent tracer dye from the endothelium into the media via gap junctions in rabbit ilio-femoral arteries (Griffith et al., 2002). Endogenous formation of cAMP may therefore play an important role in the EDHF phenomenon, since agonists such as ACh are capable of promoting endothelial synthesis of the nucleotide through a mechanism that is independent of the formation of prostanoids (Kamata et al., 1996; Taylor et al., 2001). Indeed, in rabbit ilio-femoral arteries, the adenylyl cyclase inhibitor 2′,5′-dideoxyadenosine (2′,5′-ddA) markedly attenuates subintimal smooth muscle hyperpolarizations evoked by ACh, thus suggesting that cAMP generated within the endothelium plays an important role in facilitating electrical coupling via myoendothelial gap junctions during agonist-induced responses (Griffith et al., 2002). This permissive role of cAMP is likely to explain the ability of 2′,5′-ddA and the PKA inhibitor Rp-cAMPS to attenuate EDHF-type relaxations in rabbit arteries (Taylor et al., 2001; Chaytor et al., 2002; Griffith et al., 2002).

The pathways that underpin prostanoid-independent endothelial cAMP synthesis nevertheless remain unclear. In theory, they could involve stimulation of specific Ca2+-stimulated adenylyl cyclase isoforms by elevations in [Ca2+]i, since there is evidence that in nonexcitable cells such isoforms co-localize with store-operated Ca2+ channels in the plasma membrane, where they are preferentially regulated by CCE (Burnay et al., 1998; Watson et al., 1998; Smith et al., 2002). This scenario would explain the ability of the SERCA inhibitor cyclopiazonic acid to promote cAMP efflux from rat endothelial cells (Kamata et al., 1996) and to evoke EDHF-type relaxations of rabbit arteries that, like those induced by ACh, are attenuated by inhibition of adenylyl cyclase by 2′,5′-ddA (Taylor et al., 1998; 2001). However, there is also evidence that gap junction-dependent elevations in endothelial Ca2+ secondary to smooth muscle activation may also stimulate cAMP formation, since phenylephrine, an α1-adrenoceptor agonist that does not stimulate the endothelium directly, induces an endothelium-dependent efflux of cAMP from perfused rabbit ear preparations that can be abolished by 18α-GA (Taylor et al., 2001). In this situation, however, the magnitude of the pressor response to phenylephrine is unaffected by blockade of gap junctions with 18α-GA, suggesting that endothelium-derived cAMP is unable to modulate constrictor tone in the absence of a co-existent endothelial hyperpolarization that can be transmitted into the media (Taylor et al., 2001). An additional pathway that may participate in agonist-induced synthesis of cAMP by the endothelium involves the formation of eicosanoid metabolites of arachidonic acid that stimulate adenylyl cyclase via a G protein-dependent mechanism (Node et al., 2001; Popp et al., 2002; see below).

In rabbit iliac and rat mesenteric arteries, EDHF-type responses are also accompanied by an endothelium-dependent increase in smooth muscle cAMP content that peaks at ~30 s before declining towards control after ~1 min (Taylor et al., 2001; Chaytor et al., 2002; Griffith et al., 2002; Matsumoto et al., 2003). This dynamic elevation in smooth muscle cAMP content can be attenuated by blockade of gap junctions with 37,43Gap 27 or 18α-GA, and is therefore a secondary phenomenon, although it is unknown if it is mediated by diffusion of endothelium-derived cAMP through myoendothelial gap junctions, modulation of smooth muscle adenylyl cyclase activity by changes in membrane potential, or the formation of a diffusible intermediate activator of adenylyl cyclase/inhibitor of cAMP phosphodiesterase (Taylor et al., 2001; Chaytor et al., 2002; Griffith et al., 2002). Elevations in smooth muscle cAMP levels have been shown to facilitate electrotonic signalling within the vascular media, and thereby amplify and prolong the transmission of ACh-induced hyperpolarizations to smooth muscle cells remote from the endothelium (Griffith et al., 2002). This mechanism is likely to contribute to the marked potentiation of EDHF-type relaxations reported in the presence of IBMX or 8-bromo-cAMP in rabbit iliac arteries (Taylor et al., 2001; Chaytor et al., 2002; Griffith et al., 2002), IBMX in rat mesenteric arteries (Matsumoto et al., 2003), and the cell-permeant cAMP analogue Sp-5,6-DCI-cBIMPS in rat renal arteries (Büssemaker et al., 2003).

Freely transferable mediators of smooth muscle hyperpolarization

Vasoactive species that are released from the endothelium and have been postulated to contribute to the EDHF phenomenon following diffusion across the extracellular space include K+ ions, arachidonic acid derivatives (eicosanoids and the endocannabinoid anandamide), H2O2 and C-type natriuretic peptide. Controversy nevertheless exists in respect of the nature and physiological relevance of the hyperpolarizing mechanisms activated by such agents and, specifically, whether their ability to increase the open-state probability of smooth muscle K+ channels accounts for EDHF-type relaxant activity. Although vascular smooth muscle cells express a range of K+ channels (KCa, Kir, KATP and Kv), in the case of the KCa family, histochemical, patch-clamp and Western blot analysis suggests that the subtype involved in direct smooth muscle hyperpolarization would most likely be BKCa, because expression of this channel predominates in smooth muscle cells with a contractile phenotype (Neylon et al., 1999; Burnham et al., 2002). Indeed, there is evidence that functional SKCa channels are not present in the media of freshly isolated arteries, and that IKCa channels are preferentially expressed by immature and de-differentiated cultured vascular smooth muscle cells (Neylon et al., 1999; Burnham et al., 2002). Only a small number of reports have claimed a role for ATP-sensitive K+ channels (KATP) channels in the EDHF phenomenon, and studies investigating the role of voltage-gated K+ channels (Kv) have been inconsistent. The Kv channel inhibitor 4-aminopyridine attenuates EDHF-type responses in rabbit femoral and guinea-pig carotid and coronary arteries, but not in rabbit mesenteric, rat cerebral and human resistance arteries (Murphy & Brayden, 1995; Ohlmann et al., 1997; Petersson et al., 1997; Eckman et al., 1998; Kwon et al., 1999; Quignard et al., 2000). Notably, 4-AP can also inhibit KCa channels, and more selective Kv inhibitors, such as dendrotoxin, have not been reported to attenuate the EDHF phenomenon (Adeagbo & Triggle, 1993; Petersson et al., 1997; Zygmunt et al., 1997a). In rat basilar arteries, 4-AP may also initiate a smooth muscle depolarization that may be conducted electrotonically via myoendothelial gap junctions to depress endothelial function, presumably by diminishing the electrochemical gradient for endothelial Ca2+ influx (Kamouchi et al., 1999; Allen et al., 2002).

K+ ions

K+ as an EDHF

Exogenous K+ ions hyperpolarize cell membranes by opening inwardly rectifying, Ba2+-sensitive K+ channels (Kir) and stimulating a family of Na+/K+-ATPase isoenzymes that are constructed from heterogeneous α ion-transporting and β-regulatory subunits, and are inhibited when ouabain binds to the α component of the pump (Blanco & Mercer, 1998; Zaritsky et al., 2000). Both hyperpolarizing mechanisms have been suggested to be activated by the efflux of K+ ions that follow the opening of endothelial KCa channels by agonists, and thereby contribute to smooth muscle relaxation (Edwards et al., 1998). This hypothesis (i.e. that endothelium-derived K+ is an EDHF) is based on observations that in rat hepatic and mesenteric arteries: (i) EDHF-type responses evoked by ACh can be attenuated by co-administration of Ba2+ ions and ouabain, without suppression of the initiating endothelial hyperpolarization, (ii) elevations in extracellular [K+] hyperpolarize/relax arterial preparations lacking an intact endothelium through mechanisms that can be blocked by ouabain and Ba2+, and (iii) measurements with a K+-sensitive electrode demonstrate an increase in [K+] in a putative ‘myoendothelial space' following stimulation with ACh (Edwards et al., 1998). Functional studies with vessels from Kir channel knockout mice suggest that the Ba2+-sensitive component of the smooth muscle relaxant response to K+ could involve a Kir 2.1 subtype (Zaritsky et al., 2000), and functional studies in concert with specific antibody staining suggest that the ouabain-sensitive component of K+-induced hyperpolarizations of rat mesenteric arteries involves Na+/K+-ATPase isoenzymes possessing α2 and α3 subunits that can be inhibited by the glycoside at nanomolar (~500 nM) concentrations (Blanco & Mercer, 1998; Weston et al., 2002). These subunits are stimulated by small elevations in extracellular [K+] above the concentrations normally employed in organ chamber experiments, that is, 5–6 mM, whereas a more ubiquitously expressed α1 subunit-containing isoenzyme may be almost fully activated at such levels of [K+]0 (Blanco & Mercer, 1998; Weston et al., 2002).

Reports that EDHF-type responses are often resistant to Ba2+ and ouabain in arteries in which exogenous K+ causes relaxation, even when these inhibitors are administered in combination, nevertheless suggest that endothelium-derived K+ does not contribute to the EDHF phenomenon in a consistent fashion (Suzuki, 1988; Quignard et al., 1999; Doughty et al., 2000; 2001; Lacy et al., 2000; Coats et al., 2001; Coleman et al., 2001a,2001b; McIntyre et al., 2001). One potential source of variability is the influence of smooth muscle activation on the ability of endothelium-derived K+ to serve as an EDHF. Constriction of rat arteries by high concentrations of agonist results in a depolarization that will diminish outward K+ currents carried by Kir channels, and also promotes an efflux of K+ via smooth muscle KCa channels, thereby resulting in the creation of an extracellular ‘cloud' of K+ ions that mask the activation of the Na+/K+-ATPase by additional sources of K+ (Dora & Garland, 2001; Richards et al., 2001). Indeed, in agonist-constricted arteries, charybdotoxin and iberiotoxin enhance hyperpolarizations and relaxations induced by exogenous K+ ions by blocking smooth muscle K+ efflux, in marked contrast to their inhibitory effects against authentic EDHF-type responses to agonists such as ACh (Dora et al., 2002; Weston et al., 2002). Electrophysiological characterization of the ionic currents activated by agonists and exogenous K+ in guinea-pig mesenteric arterioles has also cast doubt on the role of K+ as an EDHF. In these vessels, endothelial and smooth muscle cells behave as an electrical syncytium and exogenous K+ activates an inwardly rectifying Ba2+-sensitive current, whereas currents evoked by ACh or substance P are outwardly rectifying (Coleman et al., 2001a,2001b). Furthermore, inhibition of basal Kir channel activity with Ba2+ causes constriction and depolarization, with subsequent administration of ACh repolarizing the cell membrane by activating channels that are sensitive to charybdotoxin and apamin, and the combination of Ba2+ and ouabain failing to modulate the currents evoked by ACh and associated EDHF-type relaxations (Imaeda et al., 2000; Coleman et al., 2001b).

K+, ouabain and gap junctions

In some vessels, there is evidence that a component of the relaxation evoked by exogenous K+ is secondary to activation of endothelial Kir channels, which may be a significant determinant of endothelial membrane potential (Nilius & Droogmans, 2001). In the perfused rat mesenteric bed and rat mesenteric and human subcutaneous arteries, for example, K+-induced relaxations may be either abolished or attenuated by endothelial denudation, with residual responses remaining sensitive to ouabain, but insensitive to Ba2+ (Harris et al., 2000; Lacy et al., 2000; Dora & Garland, 2001; McIntyre et al., 2001). As in the case of agonists, K+-induced endothelial hyperpolarization can be transmitted to subjacent smooth muscle cells via gap junctions, because the endothelium-dependent component of the hyperpolarizing and relaxant response to K+ is attenuated by 37,43Gap 27 and 18α-GA (Doughty et al., 2000; 2001; Richards et al., 2001).

Interpretation of the effects of exogenous K+ and ouabain on vascular tone is also complicated by the ability of the glycoside to impair the functionality of gap junctions and the expression of connexin proteins in a sequential fashion (Schirrmacher et al., 1996; Harris et al., 2000; Martin et al., 2003). This biphasic action correlates with the affinity of ouabain to bind to the dominant Na+/K+-ATPase α subunits expressed in different target cells, although it is unknown if this reflects altered Na+/K+ homeostasis or conversion of the Na+/K+-ATPase into a general signal transducer that modulates downstream signal transduction pathways (Martin et al., 2003). In COS fibroblasts and HeLa epithelial cells, whose endogenous α1 subunits possess high ouabain affinity (Ki~0.3 μM), low concentrations (0.1–10 μM) attenuate dye transfer within ~1 h, whereas high concentrations (100 μM–1 mM) are required to achieve an equivalent effect in cultured rat aortic A7r5 myocytes or COS cells selected to express the ouabain-resistant rodent α1 Na+/K+-ATPase subunit (Ki~100 μM) (Martin et al., 2003). More prolonged exposure of A7r5 cells to ouabain, at concentrations that inhibit the resistant rodent α1 subunit, further results in a time-dependent loss of endogenous Cx40 and Cx43 protein, which is first evident after 90 min, almost complete after 4 h, but reversed on drug washout (Martin et al., 2003). Observations that high concentrations of ouabain (100 μM–1 mM) are required to attenuate EDHF-type relaxations in rat mesenteric, gastric, renal and femoral arteries, whereas nanomolar concentrations inhibit the α2 and α3 subunits that mediate hyperpolarization (Doughty et al., 2000; Van de Voorde & Vanheel, 2000; Jiang et al., 2001; Weston et al., 2002; Savage et al., 2003), thus suggest that the mechanisms mediating agonist- and K+-induced smooth muscle hyperpolarizations are distinct, and that effects of ouabain against gap junctional communication might contribute to its ability to inhibit the EDHF phenomenon in the rat vasculature.

Time-dependent suppression of connexin expression by ouabain might also explain observations by Nelli et al. (2003) that the glycoside fails to block bradykinin-induced EDHF-type relaxations of bovine coronary arteries after 30 min, whereas relaxation is abolished after 90 min. Indeed, these authors have suggested that inappropriately short exposure times might account for the highly variable effects of ouabain against EDHF-type relaxations of bovine and porcine arteries reported in the literature (Drummond et al., 2000; Pratt et al., 2001; Büssemaker et al., 2002; Nelli et al., 2003). A case can therefore be made for systematic examination of the concentration- and time-dependent effects of ouabain in vessels in which the glycoside has been reported to be ineffective as an inhibitor of the EDHF phenomenon (Suzuki, 1988; Quignard et al., 1999; Doughty et al., 2000; 2001; Lacy et al., 2000; Coats et al., 2001; Jiang & Dusting, 2001; McIntyre et al., 2001). It also remains to be determined if the ability of glycyrrhetinic acid derivatives to inhibit the Na+/K+-ATPase, which could in theory reflect the similarity of their steroidal structure to that of ouabain, contributes to their action against gap junctional communication, although such compounds are reportedly ~100-fold less potent as inhibitors of the ionic activity of the pump than ouabain (Terasawa et al., 1992).

Arachidonate metabolites

Evidence that mobilization of arachidonic acid from membrane phospholipids by phospholipase A2 (PLA2) may be an initiating step in the EDHF phenomenon has been obtained in a range of arteries and vascular beds in which inhibitors of this enzyme attenuate NO/prostanoid-independent relaxations (Table 2). Observations that inhibitors directed against either the cytosolic Ca2+-dependent (cPLA2) or secretory (sPLA2) forms of the enzyme are capable of depressing EDHF-type relaxations could reflect physiological cross-talk between these isoenzymes (Balsinde et al., 2002), as well as a potential lack of pharmacological selectivity (Table 2). The participation of arachidonate metabolites in the EDHF phenomenon is also suggested by similarities in the endothelium-dependent effects of agonists and mellitin, a polypeptide that activates PLA2 through a receptor-independent mechanism and evokes EDHF-type responses that are attenuated by inhibition of cPLA2 or blockade of gap junctional communication by 37,43Gap 27 in the rabbit superior mesenteric artery (Hutcheson et al., 1999; Hutcheson & Griffith, 2000). In theory, the ability of PLA2 inhibitors to depress the EDHF phenomenon could reflect impaired synthesis of arachidonate products that induce smooth muscle hyperpolarization through a paracrine action or, alternatively, impaired synthesis of products that promote endothelial hyperpolarization by stimulating Ca2+ influx via store-operated Ca2+ channels (Graier et al., 1995; Fleming, 2001; see below). Observations that mobilization of arachidonate by agonists or SERCA inhibitors is itself crucially dependent on sustained endothelial Ca2+ influx via store-operated channels, and therefore suppressed by inhibitors of the Ca2+-dependent cPLA2 or buffering Ca2+ influx with an intracellular Ca2+ chelator (Millanvoye-Van Brussel et al., 1999), may explain why a reportedly selective inhibitor (HELSS) of the Ca2+-independent cytosolic isoenzyme (iPLA2) is inactive against EDHF-type relaxations (Table 2).

Table 2

Reported effects of three classes of PLA2 inhibitor against EDHF-type relaxations

PLA2 isoformcPLA2iPLA2sPLA2Inhibits EDHF/species and artery typeReference
ONO-RS-082+ ++Rabbit mesentericHutcheson et al. (1999)
    +Rat coronaryFulton et al. (1996)
    −Rat mesentericTanaka et al. (1999)
AACOCF3+++ +Rabbit mesentericHutcheson et al. (1999); Hutcheson & Griffith (2000)
    +Rat coronary, mesenteric, cerebralFulton et al. (1996); Adeagbo & Henzel (1998); You et al. (2002)
    −Guinea-pig coronary, carotid −bovine coronaryYamanaka et al. (1998); Quignard et al. (2002); Drummond et al. (2000)
PACOCF3+++++Rat cerebralYou et al. (2002)
OOPC  ++Human subcutaneousCoats et al. (2001)
    +Rat coronary, mesentericFulton et al. (1996); Adeagbo & Henzel (1998)
HELSS + −Rabbit mesentericHutcheson et al. (1999)
    −Rat mesenteric, cerebralAdeagbo & Henzel (1998); You et al. (2002)
+/− signs reflect the ability of each compound to inhibit different PLA2 isoenzymes and relaxation. cPLA2, cytosolic Ca2+-dependent phospholipase A2; iPLA2, cytosolic Ca2+-independent phospholipase A2; sPLA2, secretory phospholipase A2. ONO-RS-082, 2-(p-amylcinnamoyl) amino-4-chlorobenzoic acid; AACOCF3, arachidonyl trifluoromethyl ketone; PACOCF3, palmitoyl trifluoromethyl ketone; OOPC, oleyloxyethyl phosphorylcholine; HELSS, haloenol lactone suicide substrate.

Eicosanoids

The endothelium is capable of synthesizing four epoxyeicosatrienoic acid regioisomers (5,6-, 8,9-, 11,12- and 14,15-EET) from arachidonic acid via cytochrome P450 (CYP450) epoxygenases, variously reported as belonging to the CYP 1A, 2C and 2J subfamilies, and complementary experimental approaches have suggested that these eicosanoids can contribute to the EDHF phenomenon (Campbell & Harder, 1999; Fleming, 2001; Node et al., 2001). In some vessels, CYP450 inhibitors, such as clotrimazole, sulfaphenazole and ODYA, and an EET antagonist (14,15-EEZE) impair EDHF-type relaxations and hyperpolarizations, and in the converse sense, compounds that upregulate the expression of CYP450 epoxygenases, such as nifedipine and β-napthoflavone, can enhance EET synthesis and potentiate EDHF-type responses (Campbell & Harder, 1999; Fisslthaler et al., 1999; 2000; Fleming, 2001; Gauthier et al., 2002). Although some of these pharmacological probes exert nonspecific actions, antisense oligonucleotides targeted to the coding region of CYP 2C attenuate EDHF-type responses in porcine coronary arteries and hamster skeletal muscle arterioles, consistent with the involvement of this isoenzyme in events that lead to relaxation, at least in specific vessels (Fisslthaler et al., 1999; Bolz et al., 2000).

Smooth muscle action

One mechanism that may explain these observations is that EETs act as freely transferable EDHFs, since in some arteries they activate smooth muscle BKCa channels, possibly via the occupation of specific membrane receptors (Hecker et al., 1994; Campbell et al., 1996; Fisslthaler et al., 1999; Gauthier et al., 2002; Snyder et al., 2002; Archer et al., 2003). In canine coronary microvessels, exogenous EETs are reportedly capable of evoking relaxation at subnanomolar concentrations, and cascade bioassay studies using bovine coronary arteries or cultured porcine coronary endothelium as the donor tissue have detected release of an EDHF whose effects on BKCa channel activity in downstream vascular myocytes can be mimicked by exogenous 5,6-EET or 14,15-EET (Popp et al., 1996; Gebremedhin et al., 1998; Oltman et al., 1998). However, in porcine coronary and human internal mammary arteries, in which 11,12-EET is the dominant regioisomer produced by the endothelium, concentrations of exogenous 11,12-EET in the range 0.1–3 μM are required to evoke iberiotoxin-sensitive smooth muscle relaxations that mimic the ‘authentic' EDHF-type response to agonists, and it remains unproven whether the endothelium of these arteries can generate this eicosanoid at equivalent levels (Fisslthaler et al., 1999; 2000; Archer et al., 2003).

Endothelial actions

EETs may also exert autocrine effects that promote endothelial hyperpolarization through mechanisms that are functionally indistinguishable from those initiated by agonists. In the rabbit superior mesenteric artery, for example, exogenous 5,6-EET is devoid of direct smooth muscle relaxant activity, but evokes EDHF-type relaxations in preparations with intact endothelium (Hutcheson et al., 1999). Analogously, in the porcine coronary artery, exogenous 11,12-EET induces EDHF-type smooth muscle hyperpolarizations that are sensitive to the combination of apamin and charybdotoxin, with only a small additional direct BKCa-mediated change in smooth muscle membrane potential being evident (Edwards et al., 2000). Notably, in cultured human, porcine and bovine endothelial cells, nanomolar concentrations of 5,6-EET elevate endothelial Ca2+ levels by promoting Ca2+ entry via store-operated channels, leading to the suggestion that this regioisomer is an endothelial ‘Ca2+ influx factor' whose synthesis by CYP450 epoxygenase is activated by the store depletion associated with administration of agonists or SERCA inhibitors (Graier et al., 1995; Hoebel et al., 1997; Rzigalinski et al., 1999; Xie et al., 2002). EET-induced elevations in [Ca2+]i would be expected to increase the open-state probability of all endothelial KCa subtypes, and there is also evidence that each of the four regioisomers can open BKCa channels directly when applied to the cytosolic side of the plasmalemma in cultured endothelial cells (Baron et al., 1997).

Further similarities in the endothelial effects of EETs, agonists and SERCA inhibitors are evident in rabbit mesenteric and iliac arteries in which EDHF-type relaxations evoked by exogenous 5,6-EET mimic the response to ACh and cyclopiazonic acid as they are attenuated by 37,43Gap 27, 18α-GA and 2′,5′-ddA, and therefore involve gap junctional communication and formation of cAMP (Hutcheson et al., 1999; Taylor et al., 2001). Although the ability of 5,6-EET to elevate endothelial [Ca2+]i may promote the formation of cAMP by Ca2+-stimulated adenylyl cyclase isoforms (see above), all the four EET regioisomers may also enhance endothelial cAMP synthesis via activation of adenylyl cyclase by Gαs following ADP ribosylation of this component of the heterotrimeric G protein complex (Li et al., 1999; Node et al., 2001). Evidence for a functional role of this pathway has been obtained in cultured porcine coronary endothelial cells, in which bradykinin enhances inter-endothelial dye transfer through a cAMP-dependent mechanism that may be secondary to endogenous synthesis of 11,12-EET (Popp et al., 2002). The ability of bradykinin and 11,12-EET to modulate the functionality of gap junctions in these cells is biphasic, and consists of an initial rapid enhancement of inter-endothelial communication, followed by a delayed reduction in dye transfer after 5–10 min that results from phosphorylation of Cx43 by extracellular regulated kinases 1/2 (ERK1/2) (Brandes et al., 2002; Popp et al., 2002).

Evidence against the involvement of EETs

There is nevertheless substantial evidence to suggest that endogenous synthesis of EETs cannot be regarded as a universal participant in the EDHF phenomenon. Inhibitors of PLA2 are inactive in arteries from some species (Table 2), and more specifically, in rat mesenteric and hepatic arteries, guinea-pig carotid arteries, bovine coronary arteries and the perfused mouse hindlimb EDHF-type responses are unaffected by blockade of CYP450 epoxygenases (Zygmunt et al., 1996; Fukao et al., 1997b; Vanheel & Van de Voorde, 1997; Chataigneau et al., 1998a; Brandes et al., 2000; Drummond et al., 2000; Quignard et al., 2002). Paracrine activation of smooth muscle BKCa channels by endogenously released EETs also seems improbable in the guinea-pig coronary artery, in which iberiotoxin attenuates smooth muscle hyperpolarizations evoked by exogenous 11,12-EET, whereas in this and other guinea-pig arteries authentic EDHF-type responses to ACh are unaffected by the toxin (Petersson et al., 1997; Eckman et al., 1998; Yamanaka et al., 1998). In guinea-pig carotid arteries, exogenous EETs also fail to promote hyperpolarization and mediate relaxation in preparations with intact endothelium, thus seemingly excluding autocrine effects (Chataigneau et al., 1998a). As noted above, EDHF-type relaxations may also be insensitive to iberiotoxin in rat arteries (Rapacon et al., 1996; Plane et al., 1997; Edwards et al., 1998).

Anandamide

Another endothelial derivative of arachidonic acid that has been proposed as an EDHF is the endocannabinoid N-arachidonylethanolamide (anandamide), which can evoke smooth muscle hyperpolarizations and relaxations that are susceptible to BKCa channel blockade by iberiotoxin (Randall et al., 1996; Deutsch et al., 1997; Plane et al., 1997). Observations that the CB1 cannabinoid receptor antagonist SR141716A attenuates agonist-evoked EDHF-type relaxations initially suggested a role for the activation of specific receptors following endogenous release of anandamide (Randall et al., 1996; White & Hiley, 1997). It subsequently became apparent, however, that anandamide is unlikely to be a universal participant in the EDHF phenomenon, as it relaxes endothelium-denuded rat hepatic arteries without altering smooth muscle membrane potential, and fails to evoke hyperpolarization and relaxation in endothelium-denuded porcine coronary arteries, which nevertheless exhibit prominent EDHF-type responses (Zygmunt et al., 1997b; Chataigneau et al., 1998b). Furthermore, in some arteries, low micromolar concentrations of SR141716A, which are thought to be selective for the CB1 receptor subtype, fail to inhibit EDHF-type relaxations evoked by muscarinic agonists, bradykinin, and even exogenous anandamide itself (Plane et al, 1997; White & Hiley, 1997; Zygmunt et al, 1997b; Chataigneau et al., 1998b; Fulton & Quilley, 1998; Pratt et al., 1998; Wagner et al., 1999). CB1 receptor agonists may also be without EDHF-type vasodilator activity in vivo, for example, in the rabbit under conditions of combined NO synthase and cyclooxygenase blockade (Niederhoffer & Szabo, 1999).

In addition to direct smooth muscle relaxant activity, in rat hepatic and rabbit mesenteric arteries, exogenous anandamide can itself stimulate endothelium-dependent relaxations that are independent of NO and prostanoids, and thus exhibit the characteristics of the EDHF phenomenon (Zygmunt et al., 1997b; Chaytor et al., 1999). One explanation might be that anandamide can occupy endothelial CB2 receptors and mobilize Ca2+ from intracellular stores via the formation of InsP3, thereby activating Ca2+ capacitative entry (Zoratti et al., 2003). However, in rabbit mesenteric arteries, EDHF-type relaxations evoked by anandamide, while being insensitive to the selective CB1 receptor antagonist LY320135, appear to involve facilitated endothelial uptake of the endocannabinoid via the high-affinity anandamide transporter (Chaytor et al., 1999). In rabbit mesenteric arteries, observations that 37,43Gap 27 and 18α-GA and high micromolar concentrations of SR141716A each block EDHF-type relaxations evoked by anandamide and ACh may reflect a common ability to attenuate electrotonic signalling, as dye-transfer studies have demonstrated that SR141716A can ‘nonspecifically' block gap junctional communication (Chaytor et al., 1999). High micromolar concentrations of SR141716A may also impair capacitative Ca2+ influx initiated by agonists and SERCA inhibitors (Mombouli et al., 1999).

Hydrogen peroxide

In vitro and in vivo studies have indicated that endogenous formation of H2O2 may contribute to aspects of circulatory control as diverse as the vascular myogenic response and coronary autoregulation (Nowicki et al., 2001; Yada et al., 2003). H2O2 has also been implicated as an EDHF in isolated human and mouse mesenteric arteries, on the basis that NO/prostanoid-independent hyperpolarizations and relaxations to ACh are attenuated by catalase and associated with endothelial production of H2O2, as assessed by imaging with the oxidant-sensitive dye dihydrodichlorofluorescein (Matoba et al., 2000; 2002; Rabelo et al., 2003). Catalase also attenuates EDHF-type relaxations evoked by bradykinin and a cytokine (leukaemia inhibitory factor) in rat mesenteric and porcine pial arteries (Kimura et al., 2002; Lacza et al., 2002). By contrast, EDHF-type relaxations of human radial arteries, which account for ~50% of the relaxant response to ACh in this vessel, are insensitive to catalase (Hamilton et al., 2001), and the enzyme also fails to impair EDHF-type responses in porcine coronary arteries and the perfused bovine ocular and rat and mouse mesenteric vascular beds (Bény & Von der Weid, 1991; Pomposiello et al., 1999; Brandes et al., 2000; McNeish et al., 2002).

The reasons for such marked vessel and species differences remain unclear, although it has become apparent that the cellular mechanisms that mediate the smooth muscle response to H2O2 exhibit considerable heterogeneity. Indeed, in rat mesenteric arteries, the response to exogenous H2O2 may be biphasic, with low concentrations promoting constriction (Gao et al., 2003). Furthermore, in human, rat, murine and porcine arteries, hyperpolarizations and relaxations evoked by exogenous H2O2 display widely different susceptibities to the pharmacological blockade of KCa, KATP and Kv channels and the Na+/K+-ATPase, and catalase-sensitive ‘EDHF-type' relaxations exhibit differential sensitivities to KCa and KATP channel blockers in human, murine and rat arteries compared to porcine arteries (Pomposiello et al., 1999; Barlow et al., 2000; Matoba et al., 2000; 2002; Kimura et al., 2002; Lacza et al., 2002; Gao et al., 2003; Miura et al., 2003). In the rabbit, endothelium-derived H2O2 may more correctly be regarded as a relaxing factor, rather than an EDHF, that depresses smooth muscle tone by modulating the sensitivity of the contractile apparatus to Ca2+, rather than causing changes in membrane potential (Iesaki et al., 1996; Chaytor et al., 2003; Itoh et al., 2003). In rabbit iliac arteries, for example, the Ca2+ ionophore A23187 evokes a pronounced release of H2O2 from the endothelium that causes large catalase-sensitive relaxations that are independent of changes in smooth muscle membrane potential, and can be clearly distinguished from a co-existent catalase-insensitive conducted hyperpolarization that is abolished by connexin-mimetic peptides (Chaytor et al., 2002; 2003). The action of A23187 in this vessel contrasts with ACh, which only weakly stimulates endothelial H2O2 production and evokes EDHF-type smooth muscle hyperpolarizations and relaxations that are essentially unaffected by catalase (Chaytor et al., 2003). There is also evidence that H2O2 may act synergistically with electronically conducted mechanisms in human mesenteric arteries, in which EDHF-type relaxations and hyperpolarizations evoked by bradykinin are incompletely attenuated by catalase, and a gap junction-dependent component is revealed by administration of 18α-GA (Matoba et al., 2002).

A further difficulty in classifying H2O2 unambiguously as an EDHF is that electrochemical measurements of endogenous H2O2 production by the endothelium reveal extracellular levels of just 10–100 nM following stimulation with A23187, for example, in the rat aorta (Cosentino et al., 1998), whereas in a variety of human, rat, murine, rabbit and porcine arteries, authentic H2O2 induces significant direct smooth muscle hyperpolarization/relaxation only at ‘supraphysiological' concentrations in the range 100 μM–1 mM (Bény & Von der Weid, 1991; Karasu, 1999; Matoba et al., 2000; Fujimoto et al., 2001; Chaytor et al., 2003; Gao et al., 2003; Hattori et al., 2003; Miura et al., 2003). Speculatively, this discrepancy between the functional effects of endogenous and authentic H2O2 might reflect higher biological activity of ‘nascent' H2O2 generated enzymatically in close proximity to its site of action (Chaytor et al., 2003; Miura et al., 2003). The enzymatic mechanisms that underpin production of H2O2 by the endothelium are also controversial. Matoba et al. (2000) have suggested that agonist-induced formation of H2O2 depends on dismutation of eNOS-generated superoxide anions, on the basis that ACh-evoked hyperpolarization, relaxation and endothelial dihydrodichlorofluorescein fluorescence are depressed in mesenteric arteries from eNOS knockout mice. This hypothesis would, however, appear to conflict with evidence that pharmacological inhibition of eNOS by L-arginine analogues causes a reduction in the formation of superoxide anions and H2O2 by this enzyme, and that release of H2O2 can still be detected in the presence of such inhibitors (Cosentino et al., 1998; Xia et al., 1998; Chaytor et al., 2003). Indeed, in human resistance arteries, catalase-sensitive endothelium-dependent dilatations induced by fluid shear stress have been attributed to H2O2 generated by Complex I of the mitochondrial electron transport chain (Liu et al., 2003), and in cultured bovine aortic endothelial cells flow-induced H2O2 formation appears to originate from xanthine oxidase (McNally et al., 2003). It also remains to be determined if autocrine effects of H2O2 within the endothelium contribute to the EDHF phenomenon, since this reactive oxygen species can activate endothelial PLA2 and elevate endothelial [Ca2+]i (Boyer et al., 1995; Saito et al., 2001).

C-type natriuretic peptide

Endothelial cells are capable of synthesizing and storing C-type natriuretic peptide, which is structurally related to atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP). CNP can subsequently be released by endothelium-dependent agonists, such as ACh and bradykinin, to activate specific natriuretic peptide receptors (NPR) on vascular smooth muscle cells and thereby modulate arterial tone (Wennberg et al., 1999). Following occupation of the NPR-B subtype, which is coupled to the particulate guanylyl cyclase enzyme, CNP promotes relaxation through an NO-independent elevation in cGMP levels (Tao et al., 1995; Mori et al., 1997; Barber et al., 1998). Since CNP-induced relaxation has variously been associated with an opening of KCa or KATP channels that may be secondary to cGMP-dependent channel phosphorylation, the peptide may thus be considered as an EDHF (Barber et al., 1998; Honig et al., 2001). It has also been suggested that CNP can activate a non-guanylyl cyclase-coupled NPR-C receptor that opens Kir channels through a pertussis toxin-sensitive G protein-linked mechanism that contributes to EDHF-type relaxations in rat mesenteric arteries (Chauhan et al., 2003a). The generality of this mechanism, nevertheless, remains to be established since pertussis toxin does not consistently attenuate EDHF-type relaxations (Graier et al., 1996), and such relaxations are not associated with the elevations in cGMP levels that typically follow administration of CNP. Indeed, Barton et al. (1998) concluded that CNP is not the principal mediator of the EDHF phenomenon in porcine coronary arteries, on the basis that the relaxation and the associated hyperpolarization evoked by exogenous CNP were much less pronounced than those evoked by bradykinin.

Interactions with nitric oxide

Agonist-induced responses

Some studies have suggested that NO may contribute to agonist-induced EDHF-type responses, either as a consequence of ‘residual' activity resulting from incomplete pharmacological blockade of eNOS or release of NO from preformed endothelial stores (Cohen et al., 1997; Chauhan et al., 2003b). While it is apparent that NO can hyperpolarize vascular smooth muscle cells by opening K+ channels, the ambient NO levels necessary to evoke changes in membrane potential are nevertheless generally much higher than those required to induce relaxation (Parkington et al., 1995; Tare et al., 2000). Furthermore, the existence of a distinct hyperpolarizing mechanism, activated either by agonists or fluid shear stress, is evident in eNOS knockout mice (Waldron et al., 1999; Brandes et al., 2000; Huang et al., 2000; 2001). There is also accumulating evidence to support a dynamic reciprocal relationship between EDHF-type and NO-mediated mechanisms of vasomotor control: EDHF-type responses are potentiated by pharmacological blockade or knockout of eNOS, and reduced by exogenous NO or the overproduction of NO that occurs in sepsis (Bauersachs et al., 1996; McCulloch et al., 1997; Waldron et al., 1999; Hutcheson et al., 1999; Kessler et al., 1999; Huang et al., 2000; 2001; Hutcheson & Griffith, 2000). Indeed, in the canine coronary microcirculation, the EDHF phenomenon can completely offset loss of NO-mediated vasodilatation in respect of the dilator response to bradykinin (Nishikawa et al., 2000), with the corollary that EDHF-type relaxations can serve as a ‘back-up' mechanism that in specific situations may fully compensate for diminished NO activity in vivo. Mechanisms contributing to the inhibitory action of NO against the EDHF phenomenon may involve direct effects of NO or its second messenger cGMP (Olmos et al., 1995; McCulloch et al., 1997). Specifically, these may include: (i) inhibition of CCE into endothelial cells following cGMP-dependent, protein kinase G (PKG)-mediated phosphorylation of the store-operated channels that mediate Ca2+ influx (Dora et al., 2001), (ii) attenuated CCE as a consequence of enhanced SERCA-mediated filling of Ca2+ stores following phosphorylation of phospholamban by PKG (Trepakova et al., 1999; Mundina-Weilenmann et al., 2000) or cGMP-independent, NO-mediated S-glutathiolation of the SERCA pump (Adachi et al., 2002), and (iii) inhibitory effects of NO on the activity of CYP450 epoxygenases (Fleming, 2001). Although there is evidence that NO can reduce the permeability of gap junctions in neurones and retinal cells, probably via PKG-mediated phosphorylation of connexin proteins (Lu & McMahon, 1997; Strata et al., 1998; Xin & Bloomfield, 2000), it remains to be established if a similar mechanism modulates electrotonic conduction in the vascular wall.

As vessel size diminishes in successive branch generations of many microvascular beds, including the human myocardium, so the magnitude of EDHF-type relaxations/dilations increases relative to NO-mediated responses (Hwa et al., 1994; Shimokawa et al., 1996; Berman & Griffith, 1997; 1998; Miura et al., 1999; Tomioka et al., 1999). More specifically, there is an inverse correlation between the magnitude of NO- and gap junction-dependent relaxations evoked by ACh in rabbit resistance arteries of different sizes (Berman et al., 2002). While such observations could reflect the suppression of EDHF-type responses by NO, through the mechanisms noted above, other factors may also contribute to spatial heterogeneity in the contribution of gap junctions to relaxation. For example, the frequency of points of focal contact between endothelial and smooth muscle cells (Aydin et al., 1991; Kristek & Gerova, 1992), and more specifically myoendothelial gap junction plaques (Sandow & Hill, 2000), is most numerous in distal vessels, suggesting that there may be closer electrical coupling between the two cell layers as vessel size diminishes, with obvious implications for electrotonic signalling. The larger EDHF-type relaxations observed in distal vessels could also reflect regional variability in the cellular mechanisms that maintain contraction: in small arteries, force development is highly dependent on Ca2+ influx via voltage-dependent channels (Tomioka et al., 1999), and would therefore be expected to be particularly sensitive to hyperpolarizing signals conducted via gap junctions.

Shear stress-induced responses

A major physiological role of the endothelium is to optimize tissue perfusion through an adaptive dynamic flow-dependent mechanism that normalizes intimal shear stress following changes in blood flow and minimizes the hydraulic work expended in the maintenance of flow (see Griffith (2002) for a review). It is well-established that the underlying cellular mechanisms may involve endothelial NO production by shear forces, and there is emerging evidence also for a contribution from EDHF-type mechanisms, since flow-dependent dilations of porcine coronary and rat mesenteric arteries and mouse skeletal muscle arterioles are attenuated by high extracellular [K+] and/or blockade of KCa channels (Takamura et al., 1999; Dube & Canty, 2001; Huang et al., 2001). Evidence that increases in intimal shear forces induce endothelial hyperpolarization has been provided by observations that flow activates endothelial Kir channels and that KCa channel blockade with apamin/charybdotoxin depresses flow-induced NO release (Olesen et al., 1988; Hutcheson & Griffith, 1994), although it has yet to be demonstrated experimentally that shear-induced endothelial hyperpolarization is conducted electrotonically into the media via gap junctions. A further parallel between agonist and shear-induced responses is reflected by the existence of a reciprocal relationship between NO-mediated and EDHF-type mechanisms of vasodilation that is evident in respect of flow-dependent dilation: normally mediated by co-release of NO and prostanoids, an EDHF-type component is unmasked by inhibitors of eNOS and cyclooxygenase in arterioles from wild-type mice, and can also substitute for the loss of NO in vessels from eNOS knockouts (Huang et al., 2000; 2001). A role for EETs in flow-dependent dilation has also been proposed in such knockout animals (Huang et al., 2001).

Conclusions and future directions

Diverse and often conflicting reports in the literature suggest that pathways involved in the EDHF phenomenon can be vessel-, species- and, in some instances, even laboratory-specific. There is nevertheless growing evidence that the two central components of this dilatory mechanism are an initiating endothelial hyperpolarization that depends on the opening of KCa channels, and its subsequent electrotonic relay through the vascular wall via myoendothelial and homocellular smooth muscle gap junctions. This contrasts with the idea that EDHF-type relaxations are mediated by a freely transferable factor that activates smooth muscle KCa channels (such as EETs or H2O2). Indeed, it can be argued that such mechanisms are mutually antagonistic, because passive spread of endothelial hyperpolarization would be expected to promote the closure of KCa channels, whose open-state probability will be reduced by the decrease in smooth muscle [Ca2+]i known to accompany the EDHF phenomenon.

Capacitative Ca2+ entry (CCE) may not only sustain the KCa channel opening that underpins endothelial hyperpolarization, but also promote a prostanoid-independent synthesis of cAMP that facilitates electrotonic spread of this electrical response through the vessel wall via gap junctions. Further research is necessary to clarify the role of eicosanoids in the EDHF phenomenon, since EETs can exert autocrine effects within the endothelium, including the ability to stimulate CCE at nanomolar concentrations, to promote membrane hyperpolarization by opening endothelial KCa channels, and to stimulate cAMP synthesis. Interactions between these mechanisms may provide the basis for a unifying hypothesis that accounts for many characteristics of the EDHF phenomenon, at least in terms of the signalling events that occur in the endothelium of some arteries (Figure 3). It should nevertheless be noted that the role of arachidonate metabolites is unlikely to be universal, because in a significant number of reports inhibitors of PLA2 or CYP450 epoxygenase fail to attenuate NO/prostanoid-independent relaxations. Indeed, even in species in which exogenous EETs are capable of evoking EDHF-type relaxations that involve gap junctional communication and cAMP synthesis, such as the rabbit, the endothelium may normally be unable to synthesize these regioisomers at physiologically active concentrations (Pfister et al., 1991). K+ ions, which can promote smooth muscle relaxation by stimulating Na+/K+-ATPases and Kir channels, may be relevant to the EDHF phenomenon only when physiological arterial tone is low, since contraction is accompanied by an efflux of K+ ions from smooth muscle cells that can mask the ability of endothelium-derived K+ to promote smooth muscle hyperpolarization. The participation of K+ is also complicated by an ability to hyperpolarize the endothelium by activating Kir channels, thereby initiating an endothelial hyperpolarization that can be conducted into the media via gap junctions, although a physiological role for this mechanism has yet to be established. The involvement of endothelium-derived H2O2 in NO- and prostanoid-independent relaxations also requires further evaluation as this reactive oxygen species can variably activate smooth muscle KCa and KATP channels, the Na+/K+-ATPase, and also modulate the sensitivity of the smooth muscle contractile machinery to Ca2+ ions independently of changes in membrane potential.

An external file that holds a picture, illustration, etc.
Object name is 141-0705698f3.jpg

Interactions within the endothelium that may participate in the EDHF phenomenon and involve a complex matrix of pathways that link electrotonic signalling with Ca2+ homeostasis, cAMP accumulation and arachidonate metabolism. The initiating endothelial hyperpolarization is dependent on the opening of Ca2+-activated K+ channels (KCa) by elevations in cytosolic free Ca2+ that result principally from capacitative Ca2+ entry via store-operated channels (SOC). This influx is triggered by depletion of Ca2+ stores in the endoplasmic reticulum. Hyperpolarization is conducted to subjacent smooth muscle cells via myoendothelial gap junctions, and may subsequently spread through the vessel wall via homocellular gap junctions that couple smooth muscle cells (not shown). Elevations in cytosolic Ca2+ may also elevate cAMP levels, thereby enhancing the electrical conductance of myoendothelial gap junctions. Epoxyeicosatrienoic acid (EET) metabolites of arachidonic acid (AA) may contribute to endothelial hyperpolarization by opening SOCs and elevating [Ca2+]i as well as possible direct effects on KCa channels. EETs may also stimulate the synthesis of cAMP by adenylyl cyclase following ADP ribosylation by Gαs. The sites at which NO and its second messenger cGMP could theoretically inhibit endothelial and smooth muscle hyperpolarization are shown in red (see text). Note that redundancy in these interacting pathways will still allow the endothelium to mediate EDHF-type relaxations in arteries in which arachidonic acid metabolism does not contribute to Ca2+ homeostasis through autocrine mechanisms. cPLA2, cytosolic Ca2+-dependent phospholipase A2; sPLA2, secretory phospholipase A2; ER, endoplasmic reticulum; CYP P450, cytochrome P450 epoxygenase; SERCA, sarcoplasmic–endoplasmic reticulum Ca2+-ATPase.

From a physiological perspective, the role of the EDHF phenomenon in integrative vascular control may be of particular importance in the microcirculation, where there may be a general inverse relationship between the amplitude of gap junction-dependent and NO-mediated responses. Complementary contributions for these two dilatory mechanisms are implicit in their relative distribution from larger to smaller arteries and in their potential for compensatory adaptation, and may ultimately provide a novel focus for understanding circulatory regulation in disease states where NO bioavailability is decreased and ‘EDHF' upregulated. These range from atheroma and its risk markers such as hypertension and diabetes, to heart failure and ischaemia/reperfusion injury (Katz & Krum, 2001; Wigg et al., 2001; Marrelli, 2002; Sofola et al., 2002). In rats rendered hypertensive by a high-salt diet, for example, apparently ‘normal' endothelial-dependent responses to ACh may conceal major alterations in the relative contributions of NO-mediated and EDHF-type mechanisms to relaxation (Sofola et al., 2002), and in spontaneously hypertensive rats enhanced myoendothelial coupling can compensate for hyperplastic structural changes in the media, thereby maintaining a functional role for the EDHF phenomenon (Sandow et al., 2003). In diabetic rats, reductions in the amplitude of EDHF-type relaxations may reflect cAMP phosphodiesterase overactivity (specifically of the PDE3 isoenzyme), with enhanced cAMP hydrolysis presumably resulting in impaired gap junctional communication, so that relaxation can be normalized by pharmacological inhibition of PDE3 (Matsumoto et al., 2003). EDHF-type responses are also enhanced in arteries from hyperthyroid rats as a consequence of upregulated cAMP synthesis (Büssemaker et al., 2003). A more complete understanding of gender-dependent influences on gap junctional communication may ultimately provide new insights into the mechanisms that underlie patho-physiological changes in vascular reactivity. Oestrogens upregulate the expression of Cx43 and enhance EDHF-type responses in the rat mesenteric circulation (Liu et al., 2002), and in human pregnancy the relaxant response of myometrial arteries to ACh becomes entirely dependent on gap junctions, rather than NO (Kenny et al., 2002). By contrast, in cerebral arteries oestrogens inhibit EDHF-type relaxations, so that connexin-mimetic peptides attenuate the endothelium-dependent component of ADP-induced dilation in the pial circulation of ovariectomized rats, but not in controls (Golding & Kepler, 2001; Xu et al., 2002).

Finally, from a pharmacological perspective, evidence that electrotonic signalling may play a central role in vascular control should stimulate re-evaluation of the actions of many of the common pharmacological probes used to investigate the nature of the mechanisms regulating arterial tone. In addition to their more widely accepted actions, compounds as diverse as the Na+/K+-ATPase inhibitor ouabain, the CYP450 epoxygenase inhibitor clotrimazole, the cannabinoid receptor antagonist SR141716A and fenamate inhibitors of cyclooxygenase, which have all been employed experimentally to dissociate pathways contributing to the EDHF phenomenon, have now also been shown to exert major inhibitory effects on gap junctional communication (Chaytor et al., 1999; Harris et al., 2000; Harks et al., 2001; Brandes et al., 2002; Martin et al., 2003; Srinivas & Spray, 2003).

Abbreviations

AChacetylcholine
2-APB2-aminoethoxydiphenyl borate
calcein-AMcalcein acetoxymethyl ester
cAMPcyclic adenosine 3′,5′-monophosphate
CCEcapacitative Ca2+ entry
cGMPcyclic guanosine 3′,5′-monophosphate
Cxconnexin
CYP450cytochrome P450 epoxygenase
2′,5′-ddA2′,5′-dideoxyadenosine
1-EBIO1-ethyl-2-benzimidazolinone
EETepoxyeicosatrienoic acid
14,15-EEZE14,15-epoxyeicosa-5(Z)-enoic acid
ERendoplasmic reticulum
GAglycyrrhetinic acid
IBMX3-isobutyl-1-methylxanthine
InsP3inositol 1,4,5-trisphosphate
KATPATP-sensitive K+ channel
KCaCa2+-activated K+ channel
Kirinwardly-rectifying K+ channel
Kvvoltage-dependent K+ channel
LY3201356-methoxy-2-(4-methoxyphenyl) benzo[b]thien-3-yl)(4-cyanophenyl) methanone
NOnitric oxide
ODYA17-octadecynoic acid
PLA2phospholipase A2
Rp-cAMPSRp-8-bromoadenosine-3′,5′-cyclic monophosphorothioate
SERCAsarcoplasmic–endoplasmic reticulum Ca2+-ATPase
Sp-5,6-DCI-cBIMPSSp-5,6-dichlorobenzimidazole-1-beta-D-ribofuranoside 3′,5′-cyclic phosphorothioate
SR141716AN-(piperidin-1-yl)-5-(4-chlorophenyl)-1-(2,4-dichlorophenyl)-4-methyl-1H-pyrazole-3-carboxamide hydrochloride
TRAM-34(1-[(2-chlorophenyl)diphenylmethyl]-1H-pyrazole)
TRAM-392-(2-chlorophenyl)-2,2-diphenylacetonitrile
TRPtransient receptor potential
U731221-(6-{[17β-3-methoxyoestra-1,3,5(10)-trien-17-yl]amino}hexyl)-1H-pyrrole-2,5-dione

References

  • ABUDARA V., EYZAGUIRRE C., SAEZ J.C. Short- and long-term regulation of rat carotid body gap junctions by cAMP. Identification of connexin43, a gap junction subunit. Adv. Exp. Med. Biol. 2000;75:359–369. [Abstract] [Google Scholar]
  • ADACHI T., SHAROV V.S., SCHÖNEICH C., COHEN R.A. Reactive nitrogen species S-glutathiolate the sarco/endoplasmic reticulum Ca2+ ATPase and increase its activity which may account for cyclic GMP-independent response to nitric oxide. Circulation. 2002;106 Suppl:II-271. [Google Scholar]
  • ADEAGBO A.S., HENZEL M.K. Calcium-dependent phospholipase A2 mediates the production of endothelium-derived hyperpolarizing factor in perfused rat mesenteric prearteriolar bed. J. Vasc. Res. 1998;35:27–35. [Abstract] [Google Scholar]
  • ADEAGBO A.S., TRIGGLE C.R. Varying extracellular [K+]: a functional approach to separating EDHF- and EDNO-related mechanisms in perfused rat mesenteric arterial bed. J. Cardiovasc. Pharmacol. 1993;1:423–429. [Abstract] [Google Scholar]
  • ALLEN T., IFTINCA M., COLE W.C., PLANE F. Smooth muscle membrane potential modulates endothelium-dependent relaxation of rat basilar artery via myo-endothelial gap junctions. J. Physiol. 2002;545:975–986. [Abstract] [Google Scholar]
  • ARCHER S.L., GRAGASIN F.S., WU X., WANG S., MCMURTRY S., KIM D.H., PLATONOV M., KOSHAL A., HASHIMOTO K., CAMPBELL W.B., FALCK J.R., MICHELAKIS E.D. Endothelium-derived hyperpolarizing factor in human internal mammary artery is 11,12-epoxyeicosatrienoic acid and causes relaxation by activating smooth muscle BKCa channels. Circulation. 2003;107:769–776. [Abstract] [Google Scholar]
  • AYAJIKI K., FUJIOKA H., OKAMURA T. Mechanisms underlying endothelium-dependent, nitric oxide/prostacyclin-independent, acetylcholine-induced relaxation in canine corpus cavernosum. Naunyn-Schmiedeberg's Arch. Pharmacol. 2000;362:448–451. [Abstract] [Google Scholar]
  • AYDIN F., ROSENBLUM W.I., POVLISHOCK J.T. Myoendothelial junctions in human brain arterioles. Stroke. 1991;22:1592–1597. [Abstract] [Google Scholar]
  • BALSINDE J., WINSTEAD M.V., DENNIS E.A. Phospholipase A2 regulation of arachidonic acid mobilization. FEBS Lett. 2002;531:2–6. [Abstract] [Google Scholar]
  • BARBER D.A., BURNETT J.C., FITZPATRICK L.A., SIECK G.C., MILLER V.M. Gender and relaxation to C-type natriuretic peptide in porcine coronary arteries. J. Cardiovasc. Pharmacol. 1998;32:5–11. [Abstract] [Google Scholar]
  • BARLOW R.S., EL-MOWAFY A.M., WHITE R.E. H2O2 opens BKCa channels via the PLA2-arachidonic acid signaling cascade in coronary artery smooth muscle. Am. J. Physiol. 2000;279:H475–H483. [Abstract] [Google Scholar]
  • BARON A., FRIEDEN M., BÉNY J.L. Epoxyeicosatrienoic acids activate a high-conductance, Ca2+-dependent K+ channel on pig coronary artery endothelial cells. J. Physiol. 1997;504:537–543. [Abstract] [Google Scholar]
  • BARTON M., BÉNY J.L., D'USCIO L.V., WYSS T., NOLL G., LÜSCHER T.F. Endothelium-independent relaxation and hyperpolarization to C-type natriuretic peptide in porcine coronary arteries. J. Cardiovasc. Pharmacol. 1998;31:377–383. [Abstract] [Google Scholar]
  • BAUERSACHS J., POPP R., HECKER M., SAUER E., FLEMING I., BUSSE R. Nitric oxide attenuates the release of endothelium-derived hyperpolarizing factor. Circulation. 1996;94:3341–3347. [Abstract] [Google Scholar]
  • BÉNY J.L. Information networks in the arterial wall. NIPS. 1999;14:68–73. [Abstract] [Google Scholar]
  • BÉNY J.L., BRUNET P.C. Neither nitric oxide nor nitroglycerin accounts for all the characteristics of endothelially mediated vasodilatation of pig coronary arteries. Blood Vessels. 1988;25:308–311. [Abstract] [Google Scholar]
  • BÉNY J.L., PACICCA C. Bidirectional electrical communication between smooth muscle and endothelial cells in the pig coronary artery. Am. J. Physiol. 1994;266:H1465–H1472. [Abstract] [Google Scholar]
  • BÉNY J.L., VON DER WEID P.Y. Hydrogen peroxide: an endogenous smooth muscle cell hyperpolarizing factor. Biochem. Biophys. Res. Commun. 1991;176:378–384. [Abstract] [Google Scholar]
  • BÉNY J.L., ZHU P., HAEFLIGER I.O. Lack of bradykinin-induced smooth muscle cell hyperpolarization despite heterocellular dye coupling and endothelial cell hyperpolarization in porcine ciliary artery. J. Vasc. Res. 1997;34:344–350. [Abstract] [Google Scholar]
  • BERMAN R.S., GRIFFITH T.M. Differential actions of charybdotoxin on central and daughter branch arteries of the rabbit isolated ear. Br. J. Pharmacol. 1997;120:639–646. [Europe PMC free article] [Abstract] [Google Scholar]
  • BERMAN R.S., GRIFFITH T.M. Spatial heterogeneity in the mechanisms contributing to acetylcholine-induced dilatation in the rabbit isolated ear. Br. J. Pharmacol. 1998;124:1245–1253. [Europe PMC free article] [Abstract] [Google Scholar]
  • BERMAN R.S., MARTIN P.E.M., EVANS W.H., GRIFFITH T.M. Relative contributions of NO and gap junctional communication to endothelium-dependent relaxations of rabbit resistance arteries vary with vessel size. Microvasc. Res. 2002;63:115–128. [Abstract] [Google Scholar]
  • BISHARA N.B., MURPHY T.V., HILL M.A. Capacitative Ca2+ entry in vascular endothelial cells is mediated via pathways sensitive to 2 aminoethoxydiphenyl borate and xestospongin C. Br. J. Pharmacol. 2002;135:119–128. [Europe PMC free article] [Abstract] [Google Scholar]
  • BLANCO G., MERCER R.W. Isozymes of the Na–K–ATPase: heterogeneity in structure, diversity in function. Am. J. Physiol. 1998;275:F633–F650. [Abstract] [Google Scholar]
  • BOLTON T.B., LANG R.J., TAKEWAKI T. Mechanisms of action of noradrenaline and carbachol on smooth muscle of guinea-pig anterior mesenteric artery. J. Physiol. 1984;351:549–572. [Abstract] [Google Scholar]
  • BOLZ S.S., DE WIT C., POHL U. Endothelium-derived hyperpolarizing factor but not NO reduces smooth muscle Ca2+ during acetylcholine-induced dilation of microvessels. Br. J. Pharmacol. 1999;128:124–134. [Europe PMC free article] [Abstract] [Google Scholar]
  • BOLZ S.S., FISSLTHALER B., PIEPERHOFF S., DE WIT C., FLEMING I., BUSSE R., POHL U. Antisense oligonucleotides against cytochrome P450 2C8 attenuate EDHF-mediated Ca2+ changes and dilation in isolated resistance arteries. FASEB J. 2000;14:255–260. [Abstract] [Google Scholar]
  • BOYER C.S., BANNENBERG G.L., NEVE E.P., RYRFELDT A., MOLDEUS P. Evidence for the activation of the signal-responsive phospholinase A2 by exogenous hydrogen peroxide. Biochem. Pharmacol. 1995;50:753–761. [Abstract] [Google Scholar]
  • BRAKEMEIER S., EICHLER I., KNORR A., FASSHEBER T., KOHLER R., HOYER J. Modulation of Ca2+-activated K+ channel in renal artery endothelium in situ by nitric oxide and reactive oxygen species. Kidney Int. 2003;64:199–207. [Abstract] [Google Scholar]
  • BRANDES R.P., POPP R., OTT G., BREDENKOTTER D., WALLNER C., BUSSE R., FLEMING I. The extracellular regulated kinases (ERK) 1/2 mediate cannabinoid-induced inhibition of gap junctional communication in endothelial cells. Br. J. Pharmacol. 2002;136:709–716. [Europe PMC free article] [Abstract] [Google Scholar]
  • BRANDES R.P., SCHMITZ-WINNENTHAL F.H., FELETOU M., GODECKE A., HUANG P.L., VANHOUTTE P.M., FLEMING I., BUSSE R. An endothelium-derived hyperpolarizing factor distinct from NO and prostacyclin is a major endothelium-dependent vasodilator in resistance vessels of wild-type and endothelial NO synthase knockout mice. Proc. Natl. Acad. Sci. U.S.A. 2000;97:9747–9752. [Europe PMC free article] [Abstract] [Google Scholar]
  • BUDEL S., SCHUSTER A., STERGIOPOULOS N., MEISTER J.-J., BÉNY J.-L. Role of smooth muscle cells on endothelial cell cytosolic free calcium in procine coronary arteries. Am. J. Physiol. 2001;281:H1156–H1162. [Abstract] [Google Scholar]
  • BUKAUSKAS F.F., JORDAN K., BUKAUSKIENE A., BENNETT M.V., LAMPE P.D., LAIRD D.W., VERSELIS V.K. Clustering of connexin 43-enhanced green fluorescent protein gap junction channels and functional coupling in living cells. Proc. Natl. Acad. Sci. U.S.A. 2000;97:2556–2561. [Europe PMC free article] [Abstract] [Google Scholar]
  • BURGHARDT R.C., BARHOUMI R., SEWALL T.C., BOWEN J.A. Cyclic AMP induces rapid increases in gap junction permeability and changes in the cellular distribution of connexin43. J. Membr. Biol. 1995;148:243–253. [Abstract] [Google Scholar]
  • BURNAY M.M., VALLOTTON M.B., CAPPONI GA.M., ROSSIER M.F. Angiotensin II potentiates adrenocorticotrophic hormone-induced cAMP formation in bovine adrenal glomerulosa cells through a capacitative calcium influx. Biochem. J. 1998;330:21–27. [Europe PMC free article] [Abstract] [Google Scholar]
  • BURNHAM M.P., BYCHKOV R., FELETOU M., RICHARDS G.R., VANHOUTTE P.M., WESTON A.H., EDWARDS G. Characterization of an apamin-sensitive small-conductance Ca2+-activated K+ channel in porcine coronary artery endothelium: relevance to EDHF. Br. J. Pharmacol. 2002;135:1133–1143. [Europe PMC free article] [Abstract] [Google Scholar]
  • BÜSSEMAKER E., POPP R., FISSLTHALER B., LARSON CM., FLEMING I., BUSSE R., BRANDES RP. Hyperthyroidism enhances endothelium-dependent relaxation in the rat renal artery. Cardiovasc. Res. 2003;59:181–188. [Abstract] [Google Scholar]
  • BÜSSEMAKER E., WALLNER C., FISSLTHALER B., FLEMING I. The Na–K–ATPase is a target for an EDHF displaying characteristics similar to potassium ions in the porcine renal interlobar artery. Br. J. Pharmacol. 2002;137:647–654. [Europe PMC free article] [Abstract] [Google Scholar]
  • BYCHKOV R., BURNHAM M.P., RICHARDS G.R., EDWARDS G., WESTON A.H., FELETOU M., VANHOUTTE P.M. Characterization of a charybdotoxin-sensitive intermediate conductance Ca2+-activated K+ channel in porcine coronary endothelium: relevance to EDHF. Br. J. Pharmacol. 2002;137:1346–1354. [Europe PMC free article] [Abstract] [Google Scholar]
  • CAMPBELL W.B., GEBREMEDHIN D., PRATT P.F., HARDER D.R. Identification of epoxyeicosatrienoic acids as endothelium-derived hyperpolarizing factors. Circ. Res. 1996;78:415–423. [Abstract] [Google Scholar]
  • CAMPBELL W.B., HARDER D.R. Endothelium-derived hyperpolarizing factors and vascular cytochrome P450 metabolites of arachidonic acid in the regulation of tone. Circ. Res. 1999;84:484–488. [Abstract] [Google Scholar]
  • CHANSON M., WHITE M.M., GARBER S.S. cAMP promotes gap junctional coupling in T84 cells. Am. J. Physiol. 1996;271:C533–C539. [Abstract] [Google Scholar]
  • CHATAIGNEAU T., FELETOU M., DUHAULT J., VANHOUTTE P.M. Epoxyeicosatrienoic acids, potassium channel blockers and endothelium-dependent hyperpolarization in the guinea-pig carotid artery. Br. J. Pharmacol. 1998a;123:574–580. [Europe PMC free article] [Abstract] [Google Scholar]
  • CHATAIGNEAU T., FELETOU M., THOLLON C., VILLENEUVE N., VILAINE J.P., DUHAULT J., VANHOUTTE P.M. Cannabinoid CB1 receptor and endothelium-dependent hyperpolarization in guinea-pig carotid, rat mesenteric and porcine coronary arteries. Br. J. Pharmacol. 1998b;123:968–974. [Europe PMC free article] [Abstract] [Google Scholar]
  • 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. 2003b;57:207–216. [Abstract] [Google Scholar]
  • CHAUHAN S.D., NILSSON H., AHLUWALIA A., HOBBS A.J. Release of C-type natriuretic peptide accounts for the biological activity of endothelium-derived hyperpolarizing factor. Proc. Natl. Acad. Sci. U.S.A. 2003a;100:1426–1431. [Europe PMC free article] [Abstract] [Google Scholar]
  • CHAYTOR A.T., EDWARDS D.H., BAKKER L.M., GRIFFITH T.M. Distinct hyperpolarizing and relaxant roles for gap junctions and endothelium-derived H2O2 in NO-independent relaxations of rabbit arteries. Proc. Natl. Acad. Sci. U.S.A. 2003;100:15212–15217. [Europe PMC free article] [Abstract] [Google Scholar]
  • CHAYTOR A.T., EVANS W.H., GRIFFITH T.M. Peptides homologous to extracellular loop motifs of connexin 43 reversibly abolish rhythmic contractile activity in rabbit arteries. J. Physiol. 1997;503:99–110. [Abstract] [Google Scholar]
  • CHAYTOR A.T., EVANS W.H., GRIFFITH T.M. Central role of heterocellular gap junctional communication in endothelium-dependent relaxations of rabbit arteries. J. Physiol. 1998;508:561–573. [Abstract] [Google Scholar]
  • CHAYTOR A.T., MARTIN P.E., EDWARDS D.H., GRIFFITH T.M. Gap junctional communication underpins EDHF-type relaxations evoked by ACh in the rat hepatic artery. Am. J. Physiol. 2001;280:H2441–H2450. [Abstract] [Google Scholar]
  • CHAYTOR A.T., MARSH W.L., HUTCHESON I.R., GRIFFITH T.M. Comparison of glycyrrhetinic acid isoforms and carbenoxolone as inhibitors of EDHF-type relaxations mediated via gap junctions. Endothelium. 2000;7:265–278. [Abstract] [Google Scholar]
  • CHAYTOR A.T., MARTIN P.E.M., EVANS W.H., RANDALL M.D., GRIFFITH T.M. The endothelial component of cannabinoid-induced relaxation in rabbit mesenteric artery depends on gap junctional communication. J. Physiol. 1999;520:539–550. [Abstract] [Google Scholar]
  • CHAYTOR A.T., TAYLOR H.J., GRIFFITH T.M. Gap junction-dependent and -independent EDHF-type relaxations may involve smooth muscle cAMP accumulation. Am. J. Physiol. 2002;282:H1548–H1555. [Abstract] [Google Scholar]
  • CHEN G., SUZUKI H., WESTON A.H. Acetylcholine releases endothelium-derived hyperpolarizing factor and EDRF from rat blood vessels. Br. J. Pharmacol. 1988;95:1165–1174. [Europe PMC free article] [Abstract] [Google Scholar]
  • CHEN G., YAMAMOTO Y., MIWA K., SUZUKI H. Hyperpolarization of arterial smooth muscle induced by endothelial humoral substances. Am. J. Physiol. 1991;260:H1888–H1892. [Abstract] [Google Scholar]
  • CHEN G.F., SUZUKI H. Calcium dependency of the endothelium-dependent hyperpolarization in smooth muscle cells of the rabbit carotid artery. J. Physiol. 1990;421:521–534. [Abstract] [Google Scholar]
  • COATS P., JOHNSTON F., MACDONALD J., MCMURRAY J.J., HILLIER C. Endothelium-derived hyperpolarizing factor: identification and mechanisms of action in human subcutaneous resistance arteries. Circulation. 2001;103:1702–1708. [Abstract] [Google Scholar]
  • COHEN R.A., PLANE F., NAJIBI S., HUK I., MALINSKI T., GARLAND C.J. Nitric oxide is the mediator of both endothelium-dependent relaxation and hyperpolarization of the rabbit carotid artery. Proc. Natl. Acad. Sci. U.S.A. 1997;94:4193–4198. [Europe PMC free article] [Abstract] [Google Scholar]
  • COLEMAN H.A., TARE M., PARKINGTON H.C. EDHF is not K+ but may be due to spread of current from the endothelium in guinea-pig arterioles. Am. J. Physiol. 2001a;280:H2478–H2483. [Abstract] [Google Scholar]
  • COLEMAN H.A., TARE M., PARKINGTON H.C. K+ currents underlying the action of endothelium-derived hyperpolarizing factor in guinea-pig, rat and human blood vessels. J. Physiol. 2001b;531:359–373. [Abstract] [Google Scholar]
  • COSENTINO F., PATTON S., D'USCIO L.V., WERNER E.R., WERNER-FELMAYER G., MOREAU P., MALINSKI T., LÜSCHER T.F. Tetrahydrobiopterin alters superoxide and nitric oxide release in prehypertensive rats. J. Clin. Invest. 1998;101:1530–1537. [Europe PMC free article] [Abstract] [Google Scholar]
  • DAVIDSON J.S., BAUMGARTEN I.M. Glycyrrhetinic acid derivatives: a novel class of inhibitors of gap-junctional intercellular communication. Structure–activity relationships. J. Pharmacol. Exp. Ther. 1988;246:1104–1107. [Abstract] [Google Scholar]
  • DAVIDSON J.S., BAUMGARTEN I.M., HARLEY E.H. Reversible inhibition of intercellular junctional communication by glycyrrhetinic acid. Biochem. Biophys. Res. Commun. 1986;134:29–36. [Abstract] [Google Scholar]
  • DAVIS M.J., SHARMA N.R. Calcium-release-activated calcium influx in endothelium. J. Vasc. Res. 1997;34:186–195. [Abstract] [Google Scholar]
  • DE VRIESE A.S., VAN DE VOORDE J., LAMEIRE N.H. Effects of connexin-mimetic peptides on nitric oxide synthase- and cyclooxygenase-independent renal vasodilation. Kidney Int. 2002;61:177–185. [Abstract] [Google Scholar]
  • DEMBINSKA-KIEC A., PALLAPIES D., SIMMET T., PESKAR B.M., PESKAR B.A. Effect of carbenoxolone on the biological activity of nitric oxide: relation to gastroprotection. Br. J. Pharmacol. 1991;104:811–816. [Europe PMC free article] [Abstract] [Google Scholar]
  • DEUTSCH D.G., GOLIGORSKY M.S., SCHMID P.C., KREBSBACH R.J., SCHMID H.H., DAS S.K., DEY S.K., ARREAZA G., THORUP C., STEFANO G., MOORE L.C. Production and physiological actions of anandamide in the vasculature of the rat kidney. J. Clin. Invest. 1997;100:1538–1546. [Europe PMC free article] [Abstract] [Google Scholar]
  • DORA K.A., DOYLE M.P., DULING B.R. Elevation of intracellular calcium in smooth muscle causes endothelial cell generation of NO in arterioles. Proc. Natl. Acad. Sci. U.S.A. 1997;94:6529–6534. [Europe PMC free article] [Abstract] [Google Scholar]
  • DORA K.A., GARLAND C.J. Properties of smooth muscle hyperpolarization and relaxation to K+ in the rat isolated mesenteric artery. Am. J. Physiol. 2001;280:H2424–H2429. [Abstract] [Google Scholar]
  • DORA K.A., GARLAND C.J., KWAN H.Y., YAO X. Endothelial cell protein kinase G inhibits release of EDHF through a PKG-sensitive cation channel. Am. J. Physiol. 2001;280:H1272–H1277. [Abstract] [Google Scholar]
  • DORA K.A., HINTON J.M., WALKER S.D., GARLAND C.J. An indirect influence of phenylephrine on the release of endothelium-derived vasodilators in rat small mesenteric artery. Br. J. Pharmacol. 2000;129:381–387. [Europe PMC free article] [Abstract] [Google Scholar]
  • DORA K.A., INGS N.T., GARLAND C.J. KCa channel blockers reveal hyperpolarization and relaxation to K+ in rat isolated mesenteric artery. Am. J. Physiol. 2002;283:H606–H614. [Abstract] [Google Scholar]
  • DORA K.A., MARTIN P.E.M., CHAYTOR A.T., EVANS W.H., GARLAND C., GRIFFITH T.M. Role of heterocellular gap junctional communication in endothelium-dependent smooth muscle hyperpolarization: inhibition by a connexin mimetic peptide. Biochem. Biophys. Res. Commun. 1999;254:27–31. [Abstract] [Google Scholar]
  • DOUGHTY J.M., BOYLE J.P., LANGTON P.D. Potassium does not mimic EDHF in rat mesenteric arteries. Br. J. Pharmacol. 2000;130:1174–1182. [Europe PMC free article] [Abstract] [Google Scholar]
  • DOUGHTY J.M., BOYLE J.P., LANGTON P.D. Blockade of chloride channels reveals relaxations of rat small mesenteric arteries to raised potassium. Br. J. Pharmacol. 2001;132:293–301. [Europe PMC free article] [Abstract] [Google Scholar]
  • DOUGHTY J.M., PLANE F., LANGTON P.D. Charybdotoxin and apamin block EDHF in rat mesenteric artery if selectively applied to the endothelium. Am. J. Physiol. 1999;276:H1107–H1112. [Abstract] [Google Scholar]
  • DRUMMOND G.R., SELEMIDIS S., COCKS T.M. Apamin-sensitive, non-nitric oxide (NO) endothelium-dependent relaxations to bradykinin in the bovine isolated coronary artery: no role for cytochrome P450 and K+ Br. J. Pharmacol. 2000;129:811–819. [Europe PMC free article] [Abstract] [Google Scholar]
  • DUBE S., CANTY J.M., JR Shear stress-induced vasodilation in porcine coronary conduit arteries is independent of nitric oxide release. Am. J. Physiol. 2001;280:H2581–H2590. [Abstract] [Google Scholar]
  • ECKMAN D.M., HOPKINS N., MCBRIDE C., KEEF K.D. Endothelium-dependent relaxation and hyperpolarization in guinea-pig coronary artery: role of epoxyeicosatrienoic acid. Br. J. Pharmacol. 1998;124:181–189. [Europe PMC free article] [Abstract] [Google Scholar]
  • EDWARDS G., DORA K.A., GARDENER M.J., GARLAND C.J., WESTON A. K+ is an endothelium-derived hyperpolarizing factor in rat arteries. Nature. 1998;96:269–272. [Abstract] [Google Scholar]
  • EDWARDS G., FELETOU M., GARDENER M.J., THOLLON C., VANHOUTTE P.M., WESTON A.H. Role of gap junctions in the responses to EDHF in rat and guinea-pig small arteries. Br. J. Pharmacol. 1999;128:1788–1794. [Europe PMC free article] [Abstract] [Google Scholar]
  • EDWARDS G., THOLLON C., GARDENER M.J., FELETOU M., VILAINE J., VANHOUTTE P.M., WESTON A.H. Role of gap junctions and EETs in endothelium-dependent hyperpolarization of porcine coronary artery. Br. J. Pharmacol. 2000;129:1145–1154. [Europe PMC free article] [Abstract] [Google Scholar]
  • EICHLER I., WIBAWA J., GRGIC I., KNORR A., BRAKEMEIER S., PRIES A.R., HOYER J., KOHLER R. Selective blockade of endothelial Ca2+-activated small- and intermediate-conductance K+-channels suppresses EDHF-mediated vasodilation. Br. J. Pharmacol. 2003;138:594–601. [Europe PMC free article] [Abstract] [Google Scholar]
  • EMERSON G.G., SEGAL S.S. Electrical coupling between endothelial cells and smooth muscle cells in hamster feed arteries: role in vasomotor control. Circ. Res. 2000;87:474–479. [Abstract] [Google Scholar]
  • FELETOU M., VANHOUTTE P.M. Endothelium-dependent hyperpolarization of canine coronary smooth muscle. Br. J. Pharmacol. 1988;93:515–524. [Europe PMC free article] [Abstract] [Google Scholar]
  • FISSLTHALER B., HINSCH N., CHATAIGNEAU T., POPP R., KISS L., BUSSE R., FLEMING I. Nifedipine increases cytochrome P450 2C expression and endothelium-derived hyperpolarizing factor-mediated responses in coronary arteries. Hypertension. 2000;36:270–275. [Abstract] [Google Scholar]
  • FISSLTHALER B., POPP R., KISS L., POTENTE M., HARDER D.R., FLEMING I., BUSSE R. Cytochrome P450 2C is an EDHF synthase in coronary arteries. Nature. 1999;401:493–497. [Abstract] [Google Scholar]
  • FLEMING I. Cytochrome P450 and vascular homeostasis. Circ. Res. 2001;89:753–762. [Abstract] [Google Scholar]
  • FLEMING I., FISSLTHALER B., BUSSE R. Interdependence of calcium signaling and protein tyrosine phosphorylation in human endothelial cells. J. Biol. Chem. 1996;271:11009–11015. [Abstract] [Google Scholar]
  • FRIEDEN M., SOLLINI M., BÉNY J. Substance P and bradykinin activate different types of KCa currents to hyperpolarize cultured porcine coronary artery endothelial cells. J. Physiol. 1999;519:361–371. [Abstract] [Google Scholar]
  • FUJIMOTO S., ASANO T., SAKAI M., SAKURAI K., TAKAGI D., YOSHIMOTO N., ITOH T. Mechanisms of hydrogen peroxide-induced relaxation in rabbit mesenteric small artery. Eur. J. Pharmacol. 2001;412:291–300. [Abstract] [Google Scholar]
  • FUKAO M., HATTORI Y., KANNO M., SAKUMA I., KITABATAKE A. Sources of Ca2+ in relation to generation of acetylcholine-induced endothelium-dependent hyperpolarization in rat mesenteric artery. Br. J. Pharmacol. 1997a;120:1328–1334. [Europe PMC free article] [Abstract] [Google Scholar]
  • FUKAO M., HATTORI Y., KANNO M., SAKUMA I., KITABATAKE A. Evidence against a role of cytochrome P450-derived arachidonic acid metabolites in endothelium-dependent hyperpolarization by acetylcholine in rat isolated mesenteric artery. Br. J. Pharmacol. 1997b;120:439–446. [Europe PMC free article] [Abstract] [Google Scholar]
  • FULTON D., QUILLEY J. Evidence against anandamide as the hyperpolarizing factor mediating the nitric oxide-independent coronary vasodilator effect of bradykinin in the rat. J. Pharmacol. Exp. Ther. 1998;286:1146–1151. [Abstract] [Google Scholar]
  • FULTON D., MCGIFF J.C., QUILLEY J. Role of phospholipase C and phospholipase A2 in the nitric oxide-independent vasodilator effect of bradykinin in the rat perfused heart. J. Pharmacol. Exp. Ther. 1996;278:518–526. [Abstract] [Google Scholar]
  • GAO Y.J., HIROTA S., ZHANG D.W., JANSSEN L.J., LEE R.M. Mechanisms of hydrogen-peroxide-induced biphasic response in rat mesenteric artery. Br. J. Pharmacol. 2003;138:1085–1092. [Europe PMC free article] [Abstract] [Google Scholar]
  • GAUTHIER K.M., DEETER C., KRISHNA U.M., REDDY Y.K., BONDLELA M., FALCK J.R., CAMPBELL W.B. 14,15-Epoxyeicosa-5(Z)-enoic acid: a selective epoxyeicosatrienoic acid antagonist that inhibits endothelium-dependent hyperpolarization and relaxation in coronary arteries. Circ. Res. 2002;90:1028–1036. [Abstract] [Google Scholar]
  • GEBREMEDHIN D., HARDER D.R., PRATT P.F., CAMPBELL W.B. Bioassay of an endothelium-derived hyperpolarizing factor from bovine coronary arteries: role of a cytochrome P450 metabolite. J. Vasc. Res. 1998;35:274–284. [Abstract] [Google Scholar]
  • GEORGE C.H., MARTIN P.E.M., EVANS W.H. Rapid determination of gap junction formation using HeLa cells microinjected with cDNAs encoding wild-type and chimeric connexins. Biochem. Biophys. Res. Commun. 1998;247:785–789. [Abstract] [Google Scholar]
  • GHISDAL P., MOREL N. Cellular target of voltage and calcium-dependent K+ channel blockers involved in EDHF-mediated responses in rat superior mesenteric artery. Br. J. Pharmacol. 2001;134:1021–1028. [Europe PMC free article] [Abstract] [Google Scholar]
  • GLADWELL S.J., JEFFERYS J.G. Second messenger modulation of electrotonic coupling between region CA3 pyramidal cell axons in the rat hippocampus. Neurosci. Lett. 2001;300:1–4. [Abstract] [Google Scholar]
  • GOLDBERG G.S., MORENO A.P., BECHBERGER J.F., HEARN S.S., SHIVERS R.R., MACPHEE D.J., ZHANG Y.C., NAUS C.C. Evidence that disruption of connexon particle arrangements in gap junction plaques is associated with inhibition of gap junctional communication by a glycyrrhetinic acid derivative. Exp. Cell Res. 1996;222:48–53. [Abstract] [Google Scholar]
  • GOLDING E.M., KEPLER T.E. Role of estrogen in modulating EDHF-mediated dilations in the female rat middle cerebral artery. Am. J. Physiol. 2001;280:H2417–H2423. [Abstract] [Google Scholar]
  • GORDON J.L., MARTIN W. Endothelium-dependent relaxation of the pig aorta: relationship to stimulation of 86Rb efflux from isolated endothelial cells. Br. J. Pharmacol. 1983;79:531–541. [Europe PMC free article] [Abstract] [Google Scholar]
  • GRAIER W.F., HOLZMANN S., HOEBEL S.G., KUKOVETZ W.R., KOSTNER G.M. Mechanisms of L-NG nitroarginine/indomethacin-resistant relaxation in bovine and porcine coronary arteries. Br. J. Pharmacol. 1996;119:1177–1186. [Europe PMC free article] [Abstract] [Google Scholar]
  • GRAIER W.F., SIMECEK S., STUREK M. Cytochrome P450 mono-oxygenase-regulated signalling of Ca2+ entry in human and bovine endothelial cells. J. Physiol. 1995;482:259–274. [Abstract] [Google Scholar]
  • GRAZUL-BILSKA A.T., REYNOLDS L.P., BILSKI J.J., REDMER D.A. Effects of second messengers on gap junctional intercellular communication of ovine luteal cells throughout the estrous cycle. Biol. Reprod. 2001;65:777–783. [Abstract] [Google Scholar]
  • GRIFFITH T.M. Endothelial control of vascular tone by nitric oxide and gap junctions: a haemodynamic perspective. Biorheology. 2002;39:307–318. [Abstract] [Google Scholar]
  • GRIFFITH T.M., CHAYTOR A.T., TAYLOR H.J., GIDDINGS B.D., EDWARDS D.H. cAMP facilitates EDHF-type relaxations in conduit arteries by enhancing electrotonic conduction via gap junctions. Proc. Natl. Acad. Sci. U.S.A. 2002;99:6392–6397. [Europe PMC free article] [Abstract] [Google Scholar]
  • GRIFFITH T.M., TAYLOR H.J. Cyclic AMP mediates EDHF-type relaxations of rabbit jugular vein. Biochem. Biophys. Res. Commun. 1999;263:52–57. [Abstract] [Google Scholar]
  • GUAN X., WILSON S., SCHLENDER K.K., RUCH R.J. Gap junction disassembly and connexin 43 dephosphorylation induced by 18β-glycyrrhetinic acid. Mol. Carcinogen. 1996;16:157–164. [Abstract] [Google Scholar]
  • GUO Y., MARTINEZ-WILLIAMS C., GILBERT K.A., RANNELS D.E. Inhibition of gap junction communication in alveolar epithelial cells by 18α-glycyrrhetinic acid. Am. J. Physiol. 1999;276:L1018–L1026. [Abstract] [Google Scholar]
  • HAAS T.L., DULING B.R. Morphology favors an endothelial cell pathway for longitudinal conduction within arterioles. Microvasc. Res. 1997;53:113–120. [Abstract] [Google Scholar]
  • HAMILTON C.A., MCPHADEN A.R., BERG G., PATHI V., DOMINICZAK A.F. Is hydrogen peroxide an EDHF in human radial arteries. Am. J. Physiol. 2001;280:H2451–H2455. [Abstract] [Google Scholar]
  • HARKS E.G., DE ROOS A.D., PETERS P.H., DE HAAN L.H., BROUWER A., YPEY D.L., VAN ZOELEN E.J., THEUVENET A.P. Fenamates: a novel class of reversible gap junction blockers. J. Pharmacol. Exp. Ther. 2001;298:1033–1041. [Abstract] [Google Scholar]
  • HARRIS D., MARTIN P.E.M., EVANS W.H., KENDALL D.A., GRIFFITH T.M., RANDALL M.D. Role of gap junctions in endothelium-derived hyperpolarizing factor responses and mechanisms of K+-relaxation. Eur. J. Pharmacol. 2000;402:119–128. [Abstract] [Google Scholar]
  • HATTORI T., KAJIKURI J., KATSUYA H., ITOH T. Effects of H2O2 on membrane potential of smooth muscle cells in rabbit mesenteric resistance artery. Eur. J. Pharmacol. 2003;464:101–109. [Abstract] [Google Scholar]
  • HE D.S., JIANG J.X., TAFFET S.M., BURT J.M. Formation of heteromeric gap junction channels by connexins 40 and 43 in vascular smooth muscle cells. Proc. Natl. Acad. Sci. U.S.A. 1999;96:6495–6500. [Europe PMC free article] [Abstract] [Google Scholar]
  • HECKER M., BARA A.T., BAUERSACHS J., BUSSE R. Characterization of endothelium-derived hyperpolarizing factor as a cytochrome P450-derived arachidonic acid metabolite in mammals. J. Physiol. 1994;481:407–414. [Abstract] [Google Scholar]
  • HINTON J.M., LANGTON P.D. Inhibition of EDHF by two new combinations of K+-channel inhibitors in rat isolated mesenteric arteries. Br. J. Pharmacol. 2003;138:1031–1035. [Europe PMC free article] [Abstract] [Google Scholar]
  • HOEBEL B.G., KOSTNER G.M., GRAIER W.F. Activation of microsomal cytochrome P450 mono-oxygenase by Ca2+ store depletion and its contribution to Ca2+ entry in porcine aortic endothelial cells. Br. J. Pharmacol. 1997;121:1579–1588. [Europe PMC free article] [Abstract] [Google Scholar]
  • HONG T., HILL C.E. Restricted expression of the gap junctional protein connexin 43 in the arterial system of the rat. J. Anat. 1998;192:583–593. [Abstract] [Google Scholar]
  • HONIG M.L.H., SMITS P., MORRISON P.J., BURNETT J.C., JR, RABELINK T.J. C-type natriuretic peptide-induced vasodilation is dependent on hyperpolarization in human forearm resistance vessels. Hypertension. 2001;37:1179–1183. [Abstract] [Google Scholar]
  • HUANG A., SUN D., CARROLL M.A., JIANG H., SMITH C.J., CONNETTA J.A., FALCK J.R., SHESELY E.G., KOLLER A., KALEY G. EDHF mediates flow-induced dilation in skeletal muscle arterioles of female eNOS-KO mice. Am. J. Physiol. 2001;280:H2462–H2469. [Abstract] [Google Scholar]
  • HUANG A., SUN D., SMITH C.J., CONNETTA J.A., SHESELY E.G., KOLLER A., KALEY G. In eNOS knockout mice skeletal muscle arteriolar dilation to acetylcholine is mediated by EDHF. Am. J. Physiol. 2000;278:H762–H768. [Abstract] [Google Scholar]
  • HUTCHESON I.R., CHAYTOR A.T., EVANS W.H., GRIFFITH T.M. NO-independent relaxations to acetylcholine and A23187 involve different routes of heterocellular communication: role of gap junctions and phospholipase A2. Circ. Res. 1999;84:53–63. [Abstract] [Google Scholar]
  • HUTCHESON I.R., GRIFFITH T.M. Heterogeneous populations of K+ channels mediate EDRF release to flow but not agonists in rabbit aorta. Am. J. Physiol. 1994;266:H590–H596. [Abstract] [Google Scholar]
  • HUTCHESON I.R., GRIFFITH T.M. Role of phospholipase A2 and myoendothelial gap junctions in melittin-induced arterial relaxation. Eur. J. Pharmacol. 2000;406:239–245. [Abstract] [Google Scholar]
  • HWA J.J., GHIBAUDI L., WILLIAMS P., CHATERJEE M. Comparison of acetylcholine-dependent relaxation in large and small arteries of rat mesenteric vascular bed. Am. J. Physiol. 1994;266:H952–H958. [Abstract] [Google Scholar]
  • IESAKI T., OKADA T., SHIMADA I., YAMAGUCHI H., OCHI R. Decrease in Ca2+ sensitivity as a mechanism of hydrogen peroxide-induced relaxation of rabbit aorta. Cardiovasc. Res. 1996;31:820–825. [Abstract] [Google Scholar]
  • IMAEDA K., YAMAMOTO Y., FUKUTA H., KOSHITA M., SUZUKI H. Hyperpolarization-induced dilatation of submucosal arterioles in the guinea-pig ileum. Br. J. Pharmacol. 2000;131:1121–1128. [Europe PMC free article] [Abstract] [Google Scholar]
  • ITOH T., KAJIKURI J., HATTORI T., KUSAMA N., YAMAMOTO T. Involvement of H2O2 in superoxide-dismutase-induced enhancement of endothelium-dependent relaxation in rabbit mesenteric resistance artery. Br. J. Pharmacol. 2003;139:444–456. [Europe PMC free article] [Abstract] [Google Scholar]
  • IWASAKI H., MORI Y., HARA Y., UCHIDA K., ZHOU H., MIKOSHIBA K. 2-Aminoethoxydiphenyl borate (2-APB) inhibits capacitative calcium entry independently of the function of inositol 1,4,5-trisphosphate receptors. Receptors Channels. 2001;7:429–439. [Abstract] [Google Scholar]
  • JIANG F., DUSTING G.J. Endothelium-dependent vasorelaxation independent of nitric oxide and K+ release in isolated renal arteries of rats. Br. J. Pharmacol. 2001;132:1558–1564. [Europe PMC free article] [Abstract] [Google Scholar]
  • JIANG Z.G., SI J.Q., LASAREV M.R., NUTTALL A.L. Two resting potential levels regulated by the inward-rectifier potassium channel in the guinea-pig spiral modiolar artery. J. Physiol. 2001;537:829–842. [Abstract] [Google Scholar]
  • KAGOTA S., YAMAGUCHI Y., NAKAMURA K., KUNITOMO M. Characterization of nitric oxide and prostaglandin-independent relaxation in response to acetylcholine in rabbit renal artery. Clin. Exp. Pharmacol. Physiol. 1999;26:790–796. [Abstract] [Google Scholar]
  • KAMATA K., UMEDA F., KASUYA Y. Possible existence of novel endothelium-derived relaxing factor in the endothelium of rat mesenteric arterial bed. J. Cardiovasc. Pharmacol. 1996;27:601–606. [Abstract] [Google Scholar]
  • KAMOUCHI M., DROOGMANS G., NILIUS B. Membrane potential as a modulator of the free intracellular Ca2+ concentration in agonist-activated endothelial cells. Gen. Physiol. Biophys. 1999;18:199–208. [Abstract] [Google Scholar]
  • KARASU C. Increased activity of H2O2 in aorta isolated from chronically streptozotocin-diabetic rats: effects of antioxidant enzymes and enzyme inhibitors. Free Radic. Biol. Med. 1999;27:16–27. [Abstract] [Google Scholar]
  • KATZ S.D., KRUM H. Acetylcholine-mediated vasodilation in the forearm circulation of patients with heart failure: indirect evidence for the role of endothelium-derived hyperpolarizing factor. Am. J. Cardiol. 2001;87:1089–1092. [Abstract] [Google Scholar]
  • KENNY L.C., BAKER P.N., KENDALL D.A., RANDALL M.D., DUNN W.R. The role of gap junctions in mediating endothelium-dependent responses to bradykinin in myometrial small arteries isolated from pregnant women. Br. J. Pharmacol. 2002;136:1058–1085. [Europe PMC free article] [Abstract] [Google Scholar]
  • KESSLER P., POPP R., BUSSE R., SCHINI-KERTH V.B. Proinflammatory mediators chronically downregulate the formation of the endothelium-derived hyperpolarizing factor in arteries via a nitric oxide/cyclic GMP dependent mechanism. Circulation. 1999;99:1878–1884. [Abstract] [Google Scholar]
  • KIMURA K., TSUDA K., MORIWAKI C., KAWABE T., HAMADA M., OBANA M., BABA A., HANO T., NISHIO I. Leukemia inhibitory factor relaxes arteries through endothelium-dependent mechanism. Biochem. Biophys. Res. Commun. 2002;294:359–362. [Abstract] [Google Scholar]
  • KISELYOV K., XU X., MOZHAYEVA G., KUO T., PESSAH I., MIGNERY G., ZHU X., BIRNBAUMER L., MUALLEM S. Functional interaction between InsP3 receptors and store-operated Htrp3 channels. Nature. 1998;396:478–482. [Abstract] [Google Scholar]
  • KRISTEK F., GEROVA M. Myoendothelial relations in the conduit coronary artery of the dog and rabbit. J. Vasc. Res. 1992;29:29–32. [Abstract] [Google Scholar]
  • KRISTEK F., GEROVA M. Dynamics of endothelium–muscle cell contacts in the coronary artery of the dog in ontogeny. Acta Anat. (Basel) 1997;158:166–171. [Abstract] [Google Scholar]
  • KRUGER O., BÉNY J.L., CHABAUD F., TRAUB O., THEIS M., BRIX K., KIRCHHOFF S., WILLECKE K. Altered dye diffusion and upregulation of connexin37 in mouse aortic endothelium deficient in connexin40. J. Vasc. Res. 2002;39:160–172. [Abstract] [Google Scholar]
  • KÜHBERGER E., GROSCHNER K., KUKOVETZ W.R., BRUNNER F. The role of myoendothelial cell contact in non-nitric oxide-, non-prostanoid-mediated endothelium-dependent relaxation of porcine coronary artery. Br. J. Pharmacol. 1994;113:1289–1294. [Europe PMC free article] [Abstract] [Google Scholar]
  • KUMAR N.M., GILULA N.B. The gap junction communication channel. Cell. 1996;84:381–388. [Abstract] [Google Scholar]
  • KWON S.C., PYUN W.B., PARK G.Y., CHOI H.K., PAIK K.S., KANG B.S. The involvement of K+ channels and the possible pathway of EDHF in the rabbit femoral artery. Yonsei Med. J. 1999;40:331–338. [Abstract] [Google Scholar]
  • LACY P.S., PILKINGTON G., HANVESAKUL R., FISH H.J., BOYLE J.P., THURSTON H. Evidence against potassium as an endothelium-derived hyperpolarizing factor in rat mesenteric small arteries. Br. J. Pharmacol. 2000;129:605–611. [Europe PMC free article] [Abstract] [Google Scholar]
  • LACZA Z., PUSKAR M., KIS B., PERCIACCANTE J.V., MILLER A.W., BUSIJA D.W. Hydrogen peroxide acts as an EDHF in the piglet pial vasculature in response to bradykinin. Am. J. Physiol. 2002;283:H406–H411. [Abstract] [Google Scholar]
  • LI P.L., CHEN C.L., BORTELL R., CAMPBELL W.B. 11,12-Epoxyeicosatrienoic acid stimulates endogenous mono-ADP-ribosylation in bovine coronary arterial smooth muscle. Circ. Res. 1999;85:349–356. [Abstract] [Google Scholar]
  • LI X., SIMARD J.M. Multiple connexins form gap junction channels in rat basilar artery smooth muscle cells. Circ. Res. 1999;11:1277–1284. [Abstract] [Google Scholar]
  • LI X., SIMARD J.M. Connexin45 gap junction channels in rat cerebral vascular smooth muscle cells. Am. J. Physiol. 2001;281:H1890–H1898. [Abstract] [Google Scholar]
  • LITTLE T.L., XIA J., DULING B.R. Dye tracers define differential endothelial and smooth muscle coupling patterns within the arteriolar wall. Circ. Res. 1995;76:498–504. [Abstract] [Google Scholar]
  • LIU M.Y., HATTORI Y., SATO A., ICHIKAWA R., ZHANG X.H., SAKUMA I. Ovariectomy attenuates hyperpolarization and relaxation mediated by endothelium-derived hyperpolarizing factor in female rat mesenteric artery: a concomitant decrease in connexin-43 expression. J. Cardiovasc. Pharmacol. 2002;40:938–948. [Abstract] [Google Scholar]
  • LIU Y., ZHAO H., LI H., KALYANARAMAN B., NICOLOSI A.C., GUTTERMAN D.D. Mitochondrial sources of H2O2 generation play a key role in flow-mediated dilation in human coronary resistance arteries. Circ. Res. 2003;93:573–580. [Abstract] [Google Scholar]
  • LU C., MCMAHON D.G. Modulation of hybrid bass retinal gap junctional channel gating by nitric oxide. J. Physiol. 1997;499:689–699. [Abstract] [Google Scholar]
  • LUCKHOFF A., BUSSE R. Calcium influx into endothelial cells and formation of endothelium-derived relaxing factor is controlled by the membrane potential. Pflugers Arch. 1990;416:305–311. [Abstract] [Google Scholar]
  • MA H.T., VENKATACHALAM K., LI H.S., MONTELL C., KUROSAKI T., PATTERSON R.L., GILL D.L. Assessment of the role of the inositol 1,4,5-trisphosphate receptor in the activation of transient receptor potential channels and store-operated Ca2+ entry channels. J. Biol. Chem. 2001;276:18888–18896. [Abstract] [Google Scholar]
  • MARCHENKO S.M., SAGE S.O. Electrical properties of resting and acetylcholine-stimulated endothelium in intact rat aorta. J. Physiol. 1993;462:735–751. [Abstract] [Google Scholar]
  • MARCHENKO S.M., SAGE S.O. Smooth muscle cells affect endothelial membrane potential in rat aorta. Am. J. Physiol. 1994;267:H804–H811. [Abstract] [Google Scholar]
  • MARCHETTI J., PRADDAUDE F., RAJERISON R., ADER J.L., ALHENC-GELAS F. Bradykinin attenuates the [Ca2+]i response to angiotensin II of renal juxtamedullary efferent arterioles via an EDHF. Br. J. Pharmacol. 2001;132:749–759. [Europe PMC free article] [Abstract] [Google Scholar]
  • MARRELLI S.P. Altered endothelial Ca2+ regulation after ischemia/reperfusion produces potentiated endothelium-derived hyperpolarizing factor-mediated dilations. Stroke. 2002;33:2285–2291. [Abstract] [Google Scholar]
  • MARRELLI S.P., ECKMANN M.S., HUNTE M.S. Role of endothelial intermediate conductance KCa channels in cerebral EDHF-mediated dilations. Am. J. Physiol. 2003;285:H1590–H1599. [Abstract] [Google Scholar]
  • MARTIN P.E.M., HILL N.S., KRISTENSEN B., ERRINGTON R.J., GRIFFITH T.M. Ouabain exerts biphasic effects on connexin functionality and expression in vascular smooth muscle cells. Br. J. Pharmacol. 2003;140:1261–1271. [Europe PMC free article] [Abstract] [Google Scholar]
  • MATOBA T., SHIMOKAWA H., KUBOTA H., MORIKAWA K., FUJIKI T., KUNIHIRO I., MUKAI Y., HIRAKAWA Y., TAKESHITA A. Hydrogen peroxide is an endothelium-derived hyperpolarizing factor in human mesenteric arteries. Biochem. Biophys. Res. Commun. 2002;290:909–913. [Abstract] [Google Scholar]
  • MATOBA T., SHIMOKAWA H., NAKASHIMA M., HIRAKAWA Y., MUKAI Y., HIRANO K., KANAIDE H., TAKESHITA A. Hydrogen peroxide is an endothelium-derived hyperpolarizing factor in mice. J. Clin. Invest. 2000;106:1521–1530. [Europe PMC free article] [Abstract] [Google Scholar]
  • MATSUMOTO T., KOBAYASHI T., KAMATA K. Alterations in EDHF-type relaxation and phosphodiesterase activity in mesenteric arteries from diabetic rats. Am. J. Physiol. 2003;285:H283–H291. [Abstract] [Google Scholar]
  • MCCULLOCH A.I., BOTTRILL F.E., RANDALL M.D., HILEY C.R. Characterization and modulation of EDHF-mediated relaxations in the rat isolated superior mesenteric arterial bed. Br. J. Pharmacol. 1997;120:1431–1438. [Europe PMC free article] [Abstract] [Google Scholar]
  • MCINTYRE C.A., BUCKLEY C.H., JONES G.C., SANDEEP T.C., ANDREWS R.C., ELLIOTT A.I., GRAY G.A., WILLIAMS B.C., MCKNIGHT J.A., WALKER B.R., HADOKE P.W. Endothelium-derived hyperpolarizing factor and potassium use different mechanisms to induce relaxation of human subcutaneous resistance arteries. Br. J. Pharmacol. 2001;133:902–908. [Europe PMC free article] [Abstract] [Google Scholar]
  • MCNALLY J.S., DAVIS M.E., GIDDENS D.P., SAHA A., HWANG J., DIKALOV S., JO H., HARRISON D.G. Role of xanthine oxidoreductase and NAD(P)H oxidase in endothelial superoxide production in response to oscillatory shear stress. Am. J. Physiol. 2003;285:H2290–H2297. [Abstract] [Google Scholar]
  • MCNEISH A.J., WILSON W.S., MARTIN W. Ascorbate blocks endothelium-derived hyperpolarizing factor (EDHF)-mediated vasodilation in the bovine ciliary vascular bed and rat mesentery. Br. J. Pharmacol. 2002;135:1801–1809. [Europe PMC free article] [Abstract] [Google Scholar]
  • MEHRKE G., DAUT J. The electrical response of cultured guinea-pig coronary endothelial cells to endothelium-dependent vasodilators. J. Physiol. 1990;430:251–272. [Abstract] [Google Scholar]
  • MILLANVOYE-VAN BRUSSEL E., DAVID-DUFILHO M., PHAM T.D., IOUZALEN L., AUDE DEVYNCK M. Regulation of arachidonic acid release by calcium influx in human endothelial cells. J. Vasc. Res. 1999;36:235–244. [Abstract] [Google Scholar]
  • MIURA H., BOSNJAK J.J., NING G., SAITO T., MIURA M., GUTTERMAN D.D. Role for hydrogen peroxide in flow-induced dilation of human coronary arterioles. Circ. Res. 2003;92:e31–e40. [Abstract] [Google Scholar]
  • MIURA H., LIU Y., GUTTERMAN D.D. Human coronary arteriolar dilation to bradykinin depends on membrane hyperpolarization: contribution of nitric oxide and Ca2+-activated K+ channels. Circulation. 1999;99:3132–3138. [Abstract] [Google Scholar]
  • MOMBOULI J.V., BISSIRIOU I., AGBOTON V.D., VANHOUTTE P.M. Bioassay of endothelium-derived hyperpolarizing factor. Biochem. Biophys. Res. Commun. 1996;221:484–488. [Abstract] [Google Scholar]
  • MOMBOULI J.V., SCHAEFFER G., HOLZMANN S., KOSTNER G.M., GRAIER W.F. Anandamide-induced mobilization of cytosolic Ca2+ in endothelial cells. Br. J. Pharmacol. 1999;126:1593–1600. [Europe PMC free article] [Abstract] [Google Scholar]
  • MORI Y., TAKAYASU M., SUZUKI Y., MASATO S., YOSHIDA J., HIDAKA H. Vasodilator effects of C-type natriuretic peptide on cerebral arterioles in rats. Eur. J. Pharmacol. 1997;320:183–186. [Abstract] [Google Scholar]
  • MUNDINA-WEILENMANN C., VITTONE L., RINALDI G., SAID M., DE CINGOLANI G.C., MATTIAZZI A. Endoplasmic reticulum contribution to the relaxant effect of cGMP- and cAMP-elevating agents in feline aorta. Am. J. Physiol. 2000;278:H1856–H1865. [Abstract] [Google Scholar]
  • MURAI T., MURAKI K., IMAIZUMI Y., WATANABE M. Levcromakalim causes indirect endothelial hyperpolarization via a myo-endothelial pathway. Br. J. Pharmacol. 1999;128:1491–1496. [Europe PMC free article] [Abstract] [Google Scholar]
  • MURPHY M.E., BRAYDEN J.E. Apamin-sensitive K+ channels mediate an endothelium-dependent hyperpolarization in rabbit mesenteric arteries. J. Physiol. 1995;489:723–734. [Abstract] [Google Scholar]
  • NELLI S., WILSON W.S., LAIDLAW H., LLANO A., MIDDLETON S., PRICE A.G., MARTIN W. Evaluation of potassium ion as the endothelium-derived hyperpolarizing factor (EDHF) in the bovine coronary artery. Br. J. Pharmacol. 2003;139:982–988. [Europe PMC free article] [Abstract] [Google Scholar]
  • NELSON M.T., PATLAK J.B., WORLEY J.F., STANDEN N.B. Calcium channels, potassium channels, and voltage dependence of arterial smooth muscle tone. Am. J. Physiol. 1990;259:C3–C18. [Abstract] [Google Scholar]
  • NEYLON C.B., LANG R.J., FU Y., BOBIK A., REINHART P.H. Molecular cloning and characterization of the intermediate-conductance Ca2+-activated K+ channel in vascular smooth muscle: relationship between KCa channel diversity and smooth muscle cell function. Circ. Res. 1999;85:e33–e43. [Abstract] [Google Scholar]
  • NIEDERHOFFER N., SZABO B. Involvement of CB1 cannabinoid receptors in the EDHF-dependent vasorelaxation in rabbits. Br. J. Pharmacol. 1999;126:1383–1386. [Europe PMC free article] [Abstract] [Google Scholar]
  • NILIUS B., DROOGMANS G. Ion channels and their functional role in vascular endothelium. Physiol. Rev. 2001;81:1415–1459. [Abstract] [Google Scholar]
  • NILIUS B., DROOGMANS G., WONDERGEM R. Transient receptor potential channels in endothelium: solving the calcium entry puzzle. Endothelium. 2003;10:5–15. [Abstract] [Google Scholar]
  • NISHIKAWA Y., STEPP D.W., CHILIAN W.M. Nitric oxide exerts feedback inhibition on EDHF-induced coronary arteriolar dilation in vivo. Am. J. Physiol. 2000;279:H459–H465. [Abstract] [Google Scholar]
  • NODE K., RUAN X.L., DAI J., YANG S.X., GRAHAM L., ZELDIN D.C., LIAO J.K. Activation of Gas mediates induction of tissue-type plasminogen activator gene transcription by epoxyeicosatrienoic acids. J. Biol. Chem. 2001;276:15983–15989. [Abstract] [Google Scholar]
  • NOWICKI P.T., FLAVAHAN S., HASSANAIN H., MITRA S., HOLLAND S., GOLDSCHMIDT-CLERMONT P.J., FLAVAHAN N.A. Redox signaling of the arteriolar myogenic response. Circ. Res. 2001;89:114–116. [Abstract] [Google Scholar]
  • OHASHI M., SATOH K., ITOH T. Acetylcholine-induced membrane potential changes in endothelial cells of rabbit aortic valve. Br. J. Pharmacol. 1999;126:19–26. [Europe PMC free article] [Abstract] [Google Scholar]
  • OHLMANN P., MARTINEZ M.C., SCHNEIDER F., STOCLET J.C., ANDRIANTSITOHAINA R. Characterization of endothelium-derived relaxing factors released by bradykinin in human resistance arteries. Br. J. Pharmacol. 1997;121:657–664. [Europe PMC free article] [Abstract] [Google Scholar]
  • OHNISHI Y., HIRANO K., NISHIMURA J., FURUE M., KANAIDE H. Inhibitory effects of brefeldin A, a membrane transport blocker, on the bradykinin-induced hyperpolarization-mediated relaxation in the porcine coronary artery. Br. J. Pharmacol. 2001;134:168–178. [Europe PMC free article] [Abstract] [Google Scholar]
  • OLESEN S.P., CLAPHAM D.E., DAVIES P.F. Haemodynamic shear stress activates a K+ current in vascular endothelial cells. Nature. 1988;331:168–170. [Abstract] [Google Scholar]
  • OLMOS L., MOMBOULI J.V., ILLIANO S., VANHOUTTE P.M. cGMP mediates the desensitization to bradykinin in isolated canine coronary arteries. Am. J. Physiol. 1995;268:H865–H870. [Abstract] [Google Scholar]
  • OLTMAN C.L., WEINTRAUB N.L., VANROLLINS M., DELLSPERGER K.C. Epoxyeicosatrienoic acids and dihydroxyeicosatrienoic acids are potent vasodilators in the canine coronary microcirculation. Circ. Res. 1998;83:932–939. [Abstract] [Google Scholar]
  • PACICCA C., SCHAAD O., BÉNY J.L. Electrotonic propagation of kinin-induced, endothelium-dependent hyperpolarizations in pig coronary smooth muscles. J. Vasc. Res. 1996;33:380–385. [Abstract] [Google Scholar]
  • PAPASSOTIRIOU J., KOHLER R., PRENEN J., KRAUSE H., AKBAR M., EGGERMONT J., PAUL M., DISTLER A., NILIUS B., HOYER J. Endothelial K+ channel lacks the Ca2+ sensitivity-regulating beta subunit. FASEB J. 2000;14:885–894. [Abstract] [Google Scholar]
  • PARKINGTON H.C., TONTA M.A., COLEMAN H.A., TARE M. Role of membrane potential in endothelium-dependent relaxation of guinea-pig coronary arterial smooth muscle. J. Physiol. 1995;484:469–480. [Abstract] [Google Scholar]
  • PASYK E., INAZU M., DANIEL E.E. CPA enhances Ca2+ entry in cultured bovine pulmonary arterial endothelial cells in an IP3-independent manner. Am. J. Physiol. 1995;268:H138–H146. [Abstract] [Google Scholar]
  • PAULSON A.F., LAMPE P.D., MEYER R.A., TENBROEK E., ATKINSON M.M., WALSETH T.F., JOHNSON R.G. Cyclic AMP and LDL trigger a rapid enhancement in gap junction assembly through a stimulation of connexin trafficking. J. Cell Sci. 2000;113:3037–3049. [Abstract] [Google Scholar]
  • PERKINS G.A., GOODENOUGH D.A., SOSINSKY G.E. Formation of the gap junction intercellular channel requires a 30 degree rotation for interdigitating two apposing connexons. J. Mol. Biol. 1998;277:171–177. [Abstract] [Google Scholar]
  • PETERSSON J., ZYGMUNT P.M., HOGESTATT E.D. Characterization of the potassium channels involved in EDHF-mediated relaxation in cerebral arteries. Br. J. Pharmacol. 1997;120:1344–1350. [Europe PMC free article] [Abstract] [Google Scholar]
  • PFISTER S.L., FALCK J.R., CAMPBELL W.B. Enhanced synthesis of epoxyeicosatrienoic acids by cholesterol-fed rabbit aorta. Am. J. Physiol. 1991;261:H843–H852. [Abstract] [Google Scholar]
  • PLANE F., HOLLAND M., WALDRON G.J., GARLAND C.J., BOYLE J.P. Evidence that anandamide and EDHF act via different mechanisms in rat isolated mesenteric arteries. Br. J. Pharmacol. 1997;121:1509–1511. [Europe PMC free article] [Abstract] [Google Scholar]
  • PLANE F., PEARSON T., GARLAND C.J. Multiple pathways underlying endothelium-dependent relaxation in the rabbit isolated femoral artery. Br. J. Pharmacol. 1995;115:31–38. [Europe PMC free article] [Abstract] [Google Scholar]
  • POMPOSIELLO S., RHALEB N.E., ALVA M., CARRETERO O.A. Reactive oxygen species: role in the relaxation induced by bradykinin or arachidonic acid via EDHF in isolated porcine coronary arteries. J. Cardiovasc. Pharmacol. 1999;34:567–574. [Abstract] [Google Scholar]
  • POPP R., BAUERSACHS J., HECKER M., FLEMING I., BUSSE R. A transferable, beta-naphthoflavone-inducible, hyperpolarizing factor is synthesized by native and cultured porcine coronary endothelial cells. J. Physiol. 1996;497:699–709. [Abstract] [Google Scholar]
  • POPP R., BRANDES R.P., OTT G., BUSSE R., FLEMING I. Dynamic modulation of interendothelial gap junctional communication by 11,12-epoxyeicosatrienoic acid. Circ. Res. 2002;90:800–806. [Abstract] [Google Scholar]
  • PRATT P.F., HILLARD C.J., EDGEMOND W.S., CAMPBELL W.B. N arachidonylethanolamide relaxation of bovine coronary artery is not mediated by CB1 cannabinoid receptor. Am. J. Physiol. 1998;274:H375–H381. [Abstract] [Google Scholar]
  • PRATT P.F., LI P., HILLARD C.J., KURIAN J., CAMPBELL W.B. Endothelium-independent, ouabain-sensitive relaxation of bovine coronary arteries by EETs. Am. J. Physiol. 2001;280:H1113–H1121. [Abstract] [Google Scholar]
  • QUIGNARD J.F., CHATAIGNEAU T., CORRIU C., EDWARDS G., WESTON A., FELETOU M., VANHOUTTE P.M. Endothelium-dependent hyperpolarization to acetylcholine in carotid artery of guinea-pig: role of lipoxygenase. J. Cardiovasc. Pharmacol. 2002;40:467–477. [Abstract] [Google Scholar]
  • QUIGNARD J.F., FELETOU M., EDWARDS G., DUHAULT J., WESTON A.H., VANHOUTTE P.M. Role of endothelial cell hyperpolarization in EDHF-mediated responses in the guinea-pig carotid artery. Br. J. Pharmacol. 2000;129:1103–1112. [Europe PMC free article] [Abstract] [Google Scholar]
  • QUIGNARD J.F., FELETOU M., THOLLON C., VILAINE J.P., DUHAULT J., VANHOUTTE P.M. Potassium ions and endothelium-derived hyperpolarizing factor in guinea-pig carotid and porcine coronary arteries. Br. J. Pharmacol. 1999;127:27–34. [Europe PMC free article] [Abstract] [Google Scholar]
  • RABELO L.A., CORTES S.F., ALVAREZ-LEITE J.I., LEMOS V.S. Endothelium dysfunction in LDL receptor knockout mice: a role for H2O2. Br. J. Pharmacol. 2003;138:1215–1220. [Europe PMC free article] [Abstract] [Google Scholar]
  • RANDALL M.D., ALEXANDER S.P., BENNETT T., BOYD E.A., FRY J.R., GARDINER S.M., KEMP P.A., MCCULLOCH A.I., KENDALL D.A. An endogenous cannabinoid as an endothelium-derived vasorelaxant. Biochem. Biophys. Res. Commun. 1996;229:114–120. [Abstract] [Google Scholar]
  • RAPACON M., MIEYAL P., MCGIFF J.C., FULTON D., QUILLEY J. Contribution of calcium-activated potassium channels to the vasodilator effect of bradykinin in the isolated, perfused kidney of the rat. Br. J. Pharmacol. 1996;118:1504–1508. [Europe PMC free article] [Abstract] [Google Scholar]
  • RICHARDS G.R., WESTON A.H., BURNHAM M.P., FELETOU M., VANHOUTTE P.M., EDWARDS G. Suppression of K+-induced hyperpolarization by phenylephrine in rat mesenteric artery: relevance to studies of endothelium-derived hyperpolarizing factor. Br. J. Pharmacol. 2001;134:1–5. [Europe PMC free article] [Abstract] [Google Scholar]
  • RUMMERY N.M., HICKEY H., MCGURK G., HILL C.E. Connexin37 is the major connexin expressed in the media of caudal artery. Arterioscler. Thromb. Vasc. Biol. 2002;22:1427–1432. [Abstract] [Google Scholar]
  • RUSKO J., TANZI F., VAN BREEMEN C., ADAMS D.J. Calcium-activated potassium channels in native endothelial cells from rabbit aorta: conductance, Ca2+ sensitivity and block. J. Physiol. 1992;455:601–621. [Abstract] [Google Scholar]
  • RZIGALINSKI B.A., WILLOUGHBY K.A., HOFFMAN S.W., FALCK J.R., ELLIS E.F. Calcium influx factor, further evidence it is 5,6-epoxyeicosatrienoic acid. J. Biol. Chem. 1999;274:175–182. [Abstract] [Google Scholar]
  • SAITO T., ITOH H., CHUN T.H., FUKUNAGA Y., YAMASHITA J., DOI K., TANAKA T., INOUE M., MASATSUGU K., SAWADA N., SAKAGUCHI S., ARAI H., MUKOYAMA M., TOJO K., HOSOYA T., NAKAO K. Coordinate regulation of endothelin and adrenomedullin secretion by oxidative stress in endothelial cells. Am. J. Physiol. 2001;281:H1364–H1371. [Abstract] [Google Scholar]
  • SANDOW S.L., BRAMICH N.J., BANDI H.P., RUMMERY N.M., HILL C.E. Structure, function, and endothelium-derived hyperpolarizing factor in the caudal artery of the SHR and WKY rat. Arterioscler. Thromb. Vasc. Biol. 2003;23:822–828. [Abstract] [Google Scholar]
  • SANDOW S.L., HILL C.E. Incidence of myoendothelial gap junctions in the proximal and distal mesenteric arteries of the rat is suggestive of a role in endothelium-derived hyperpolarizing factor-mediated responses. Circ. Res. 2000;86:341–346. [Abstract] [Google Scholar]
  • SANDOW S.L., TARE M., COLEMAN H.A., HILL C.E., PARKINGTON H.C. Involvement of myoendothelial gap junctions in the actions of endothelium-derived hyperpolarizing factor. Circ. Res. 2002;90:1108–1113. [Abstract] [Google Scholar]
  • SAVAGE D., PERKINS J., HONG LIM C., BUND S.J. Functional evidence that K+ is the non-nitric oxide, non-prostanoid endothelium-derived relaxing factor in rat femoral arteries. Vasc. Pharmacol. 2003;40:23–28. [Abstract] [Google Scholar]
  • SCHIRRMACHER K., NONHOFF D., WIEMANN M., PETERSONGRINE E., BRINK P.R., BINGMANN D. Effects of calcium on gap junctions between osteoblast-like cells in culture. Calcif. Tis. Internat. 1996;59:259–264. [Abstract] [Google Scholar]
  • SEDOVA M., KLISHIN A., HUSER J., BLATTER L.A. Capacitative Ca2+ entry is graded with degree of intracellular Ca2+ store depletion in bovine vascular endothelial cells. J. Physiol. 2000;523:549–559. [Abstract] [Google Scholar]
  • SEGAL S.S., BÉNY J.L. Intracellular recording and dye transfer in arterioles during blood flow control. Am. J. Physiol. 1992;263:H1–H7. [Abstract] [Google Scholar]
  • SHIMOKAWA H., YASUTAKE H., FUIJI K., OWADA M.K., NAKAIKE R., FUKUMOTO Y., TAKAYANAGI T., NAGAO T., EGASHIRA K., FUJISHIMA M., TAKESHITA A. The importance of the hyperpolarizing mechanism increases as the vessel size decreases in endothelium-dependent relaxations in rat mesenteric circulation. J. Cardiovasc. Pharmacol. 1996;28:703–711. [Abstract] [Google Scholar]
  • SMITH K.E., GU C., FAGAN K.A., HU B., COOPER D.M. Residence of adenylyl cyclase type 8 in caveolae is necessary but not sufficient for regulation by capacitative Ca2+ entry. J. Biol. Chem. 2002;277:6025–6031. [Abstract] [Google Scholar]
  • SNYDER G.D., MURALI KRISHNA U., FALCK J.R., SPECTOR A.A. Evidence for a membrane site of action for 14,15-EET on expression of aromatase in vascular smooth muscle. Am. J. Physiol. 2002;283:H1936–H1942. [Abstract] [Google Scholar]
  • SOFOLA O.A., KNILL A., HAINSWORTH R., DRINKHILL M. Change in endothelial function in mesenteric arteries of Sprague–Dawley rats fed a high salt diet. J. Physiol. 2002;543:255–260. [Abstract] [Google Scholar]
  • SPAGNOLI L.G., VILLASCHI S., NERI L., PALMIERI G. Gap junctions in myo-endothelial bridges of rabbit carotid arteries. Experientia. 1982;38:124–125. [Abstract] [Google Scholar]
  • SRINIVAS M., SPRAY D.C. Closure of gap junction channels by arylaminobenzoates. Mol. Pharmacol. 2003;63:1389–1397. [Abstract] [Google Scholar]
  • STRATA F., ATZORI M., MOLNAR M., UGOLINI G., BERRETTA N., CHERUBINI E. Nitric oxide sensitive depolarization-induced hyperpolarization: a possible role for gap junctions during development. Eur. J. Neurosci. 1998;10:397–403. [Abstract] [Google Scholar]
  • SUZUKI H. The electrogenic Na–K pump does not contribute to endothelium-dependent hyperpolarization in the rabbit ear artery. Eur. J. Pharmacol. 1988;156:295–297. [Abstract] [Google Scholar]
  • TAKAMURA Y., SHIMOKAWA H., ZHAO H., IGARASHI H., EGASHIRA K., TAKESHITA A. Important role of endothelium-derived hyperpolarizing factor in shear stress-induced endothelium-dependent relaxations in the rat mesenteric artery. J. Cardiovasc. Pharmacol. 1999;34:381–387. [Abstract] [Google Scholar]
  • TANAKA Y., OTSUKA A., TANAKA H., SHIGENOBU K. Glycyrrhetinic acid-sensitive mechanism does not make a major contribution to non-prostanoid, non-nitric oxide mediated endothelium-dependent relaxation of rat mesenteric artery in response to acetylcholine. Res. Commun. Mol. Path. Pharmacol. 1999;103:227–239. [Abstract] [Google Scholar]
  • TAO H., ZHANG L.M., CASTRESANA M.R., SHILLCUTT S.D., NEWMAN W.H. C-natriuretic peptide but not atrial natriuretic peptide increases cyclic GMP in cerebral arterial smooth muscle cells. Life Sci. 1995;56:2357–2365. [Abstract] [Google Scholar]
  • TARE M., COLEMAN H.A., PARKINGTON H.C. Glycyrrhetinic derivatives inhibit hyperpolarization in endothelial cells of guinea-pig and rat arteries. Am. J. Physiol. 2002;282:H335–H341. [Abstract] [Google Scholar]
  • TARE M., PARKINGTON H.C., COLEMAN H.A. EDHF, NO and a prostanoid: hyperpolarization-dependent and -independent relaxation in guinea-pig arteries. Br. J. Pharmacol. 2000;130:605–618. [Europe PMC free article] [Abstract] [Google Scholar]
  • TAYLOR H.J., CHAYTOR A.T., EDWARDS D.H., GRIFFITH T.M. Gap junction-dependent increases in smooth muscle cAMP underpin the EDHF phenomenon in rabbit arteries. Biochem. Biophys. Res. Commun. 2001;283:583–589. [Abstract] [Google Scholar]
  • TAYLOR H.J., CHAYTOR A.T., EVANS W.H., GRIFFITH T.M. Inhibition of the gap junctional component of endothelium-dependent relaxation in rabbit iliac artery by 18α-glycyrrhetinic acid. Br. J. Pharmacol. 1998;125:1–4. [Europe PMC free article] [Abstract] [Google Scholar]
  • TERASAWA T., OKADA T., HARA T., ITOH K. Glycyrrhetinic acid derivatives as potent inhibitors of Na+, K+-ATPase. Synthesis and structure–activity relationships. Eur. J. Med. Chem. 1992;27:345–351. [Google Scholar]
  • TOMIOKA H., HATTORI Y., FUKAO M., SATO A., LUI M., SAKUMA I., KITABATAKE A., KANNO M. Relaxation in different-sized rat blood vessels mediated by endothelium-derived hyperpolarizing factor: importance of processes mediating precontractions. J. Vasc. Res. 1999;36:311–320. [Abstract] [Google Scholar]
  • TREPAKOVA E.S., COHEN R.A., BOLOTINA V.M. Nitric oxide inhibits capacitative cation influx in human platelets by promoting sarcoplasmic/endoplasmic reticulum Ca2+-ATPase-dependent refilling of Ca2+ stores. Circ. Res. 1999;84:201–209. [Abstract] [Google Scholar]
  • TREPAKOVA E.S., CSUTORA P., HUNTON D.L., MARCHASE R.B., COHEN R.A., BOLOTINA V.M. Calcium influx factor directly activates store-operated cation channels in vascular smooth muscle cells. J. Biol. Chem. 2000;275:26158–26163. [Abstract] [Google Scholar]
  • UJIIE H., CHAYTOR A.T., BAKKER L.M., GRIFFITH T.M. Essential role of Gap junctions in NO- and prostanoid-independent relaxations evoked by acetylcholine in rabbit intracerebral arteries. Stroke. 2003;34:544–550. [Abstract] [Google Scholar]
  • UNGVARI Z., CSISZAR A., KOLLER A. Increases in endothelial Ca2+ activate KCa channels and elicit EDHF-type arteriolar dilation via gap junctions. Am. J. Physiol. 2002;282:H1760–H1767. [Abstract] [Google Scholar]
  • VAN DE VOORDE J., VANHEEL B. EDHF-mediated relaxation in rat gastric small arteries: influence of ouabain/Ba2+ and relation to potassium ions. J. Cardiovasc. Pharmacol. 2000;35:543–548. [Abstract] [Google Scholar]
  • VANHEEL B., VAN DE VOORDE J. Evidence against the involvement of cytochrome P450 metabolites in endothelium-dependent hyperpolarization of the rat main mesenteric artery. J. Physiol. 1997;501:331–341. [Abstract] [Google Scholar]
  • VAN RIJEN H.V., VAN VEEN T.A., HERMANS M.M., JONGSMA H.J. Human connexin40 gap junction channels are modulated by cAMP. Cardiovasc. Res. 2000;45:941–951. [Abstract] [Google Scholar]
  • VON DER WEID P.Y., BÉNY J.L. Simultaneous oscillations in the membrane potential of pig coronary artery endothelial and smooth muscle cells. J. Physiol. 1993;471:13–24. [Abstract] [Google Scholar]
  • WAGNER J.A., VARGA K., JARAI Z., KUNOS G. Mesenteric vasodilation mediated by endothelial anandamide receptors. Hypertension. 1999;33:429–434. [Abstract] [Google Scholar]
  • WALDRON G.J., DING H., LOVREN F., KUBES P., TRIGGLE C.R. Acetylcholine-induced relaxation of peripheral arteries isolated from mice lacking endothelial nitric oxide synthase. Br. J. Pharmacol. 1999;128:653–658. [Europe PMC free article] [Abstract] [Google Scholar]
  • WANG H.Z., DAY N., VALCIC M., HSIEH K., SERELS S., BRINK P.R., CHRIST G.J. Intercellular communication in cultured human vascular smooth muscle cells. Am. J. Physiol. 2001;281:C75–C88. [Abstract] [Google Scholar]
  • WATSON E.L., WU Z., JACOBSON K.L., STORM D.R., SINGH J.C., OTT S.M. Capacitative Ca2+ entry is involved in cAMP synthesis in mouse parotid acini. Am. J. Physiol. 1998;274:C557–C565. [Abstract] [Google Scholar]
  • WEINTRAUB N.L., STEPHENSON A.H., SPRAGUE R.S., MCMURDO L., LONIGRO A.J. Relationship of arachidonic acid release to porcine coronary artery relaxation. Hypertension. 1995;26:684–690. [Abstract] [Google Scholar]
  • WENNBERG P.W., MILLER V.M., RABELINK T., BURNETT J.C., JR Further attenuation of endothelium-dependent relaxation imparted by natriuretic peptide receptor antagonism. Am. J. Physiol. 1999;277:H1618–H1621. [Abstract] [Google Scholar]
  • WESTON A.H., RICHARDS G.R., BURNHAM M.P., FELETOU M., VANHOUTTE P.M., EDWARDS G. K+-induced hyperpolarization in rat mesenteric artery: identification, localization and role of Na+/K+-ATPases. Br. J. Pharmacol. 2002;136:918–926. [Europe PMC free article] [Abstract] [Google Scholar]
  • WHITE R., HILEY C.R. A comparison of EDHF-mediated and anandamide-induced relaxations in the rat isolated mesenteric artery. Br. J. Pharmacol. 1997;122:1573–1584. [Europe PMC free article] [Abstract] [Google Scholar]
  • WIGG S.J., TARE M., TONTA M.A., O'BRIEN R.C., MEREDITH I.T., PARKINGTON H.C. Comparison of effects of diabetes mellitus on an EDHF-dependent and an EDHF-independent artery. Am. J. Physiol. 2001;281:H232–H240. [Abstract] [Google Scholar]
  • WULFF H., GUTMAN G.A., CAHALAN M.D., CHANDY K.G. Delineation of the clotrimazole/TRAM-34 binding site on the intermediate conductance calcium-activated potassium channel, IKCa1. J. Biol. Chem. 2001;276:32040–32045. [Abstract] [Google Scholar]
  • WULFF H., MILLER M.J., HANSEL W., GRISSMER S., CAHALAN M.D., CHANDY K.G. Design of a potent and selective inhibitor of the intermediate-conductance Ca2+-activated K+ channel, IKCa1: a potential immunosuppressant. Proc. Natl. Acad. Sci. U.S.A. 2000;97:8151–8156. [Europe PMC free article] [Abstract] [Google Scholar]
  • XIA Y., TSAI A.L., BERKA V., ZWEIER J.L. Superoxide generation from endothelial nitric-oxide synthase. A Ca2+/calmodulin-dependent and tetrahydrobiopterin regulatory process. J. Biol. Chem. 1998;273:25804–25808. [Abstract] [Google Scholar]
  • XIE Q., ZHANG Y., ZHAI C., BONANNO J.A. Calcium influx factor from cytochrome P-450 metabolism and secretion-like coupling mechanisms for depletion-activated calcium entry in corneal endothelial cells. J. Biol. Chem. 2002;277:16559–16566. [Abstract] [Google Scholar]
  • XIN D., BLOOMFIELD S.A. Effects of nitric oxide on horizontal cells in the rabbit retina. Vis. Neurosci. 2000;17:799–811. [Abstract] [Google Scholar]
  • XU H.L., SANTIZO R.A., BAUGHMAN V.L., PELLIGRINO D.A. ADP-induced pial arteriolar dilation in ovariectomized rats involves gap junctional communication. Am. J. Physiol. 2002;283:H1082–H1091. [Abstract] [Google Scholar]
  • YADA T., SHIMOKAWA H., HIRAMATSU O., KAJITA T., SHIGETO F., GOTO M., OGASAWARA Y., KAJIYA F. Hydrogen peroxide, an endogenous endothelium-derived hyperpolarizing factor, plays an important role in coronary autoregulation in vivo. Circulation. 2003;107:1040–1045. [Abstract] [Google Scholar]
  • YAMAKAWA N., OHHASHI M., WAGA S., ITOH T. Role of endothelium in regulation of smooth muscle membrane potential and tone in the rabbit middle cerebral artery. Br. J. Pharmacol. 1997;121:1315–1322. [Europe PMC free article] [Abstract] [Google Scholar]
  • YAMANAKA A., ISHIKAWA T., GOTO K. Characterization of endothelium-dependent relaxation independent of NO and prostaglandins in guinea-pig coronary artery. J. Pharmacol. Exp. Ther. 1998;285:480–489. [Abstract] [Google Scholar]
  • YAMAZAKI J., KITAMURA K. Intercellular electrical coupling in vascular cells present in rat intact cerebral arterioles. J. Vasc. Res. 2003;40:11–27. [Abstract] [Google Scholar]
  • YAMAMOTO Y., FUKUTA H., NAKAHIRA Y., SUZUKI H. Blockade by 18β-glycyrrhetinic acid of intercellular electrical coupling in guinea-pig arterioles. J. Physiol. 1998;511:501–508. [Abstract] [Google Scholar]
  • YAMAMOTO Y., IMAEDA K., SUZUKI H. Endothelium-dependent hyperpolarization and intercellular electrical coupling in guinea-pig mesenteric arterioles. J. Physiol. 1999;514:505–513. [Abstract] [Google Scholar]
  • YAMAMOTO Y., KLEMM M.F., EDWARDS F.R., SUZUKI H. Intercellular electrical communication among smooth muscle and endothelial cells in guinea-pig mesenteric arterioles. J. Physiol. 2001;535:181–195. [Abstract] [Google Scholar]
  • YASHIRO Y., DULING B.R. Integrated Ca2+ signalling between smooth muscle and endothelium of resistance vessels. Circ. Res. 2000;87:1048–1054. [Abstract] [Google Scholar]
  • YEH H.I., ROTHERY S., DUPONT E., COPPEN S.R., SEVERS N.J. Individual gap junction plaques contain multiple connexins in arterial endothelium. Circ. Res. 1998;83:1248–1263. [Abstract] [Google Scholar]
  • YOU J., MARRELLI S.P., BRYAN R.M., JR Role of cytoplasmic phospholipase A2 in endothelium-derived hyperpolarizing factor dilations of rat middle cerebral arteries. J. Cereb. Blood. Flow. Metab. 2002;22:1239–1247. [Abstract] [Google Scholar]
  • YOUSIF M.H., CHERIAN A., RIOWO M.A. Endothelium-dependent relaxation in isolated renal arteries of diabetic rabbits. Auton. Autacoid. Pharmacol. 2002;22:73–82. [Abstract] [Google Scholar]
  • 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;87:160–166. [Abstract] [Google Scholar]
  • ZORATTI C., KIPMEN-KORGUN D., OSIBOW K., MALLI R., GRAIER W.F. Anandamide initiates Ca2+ signaling via CB2 receptor linked to phospholipase C in calf pulmonary endothelial cells. Br. J. Pharmacol. 2003;140:1351–1362. [Europe PMC free article] [Abstract] [Google Scholar]
  • ZYGMUNT P.M., EDWARDS G., WESTON A.H., DAVIS S.C., HOGESTATT E.D. Effects of cytochrome P450 inhibitors on EDHF-mediated relaxation in the rat hepatic artery. Br. J. Pharmacol. 1996;118:1147–1152. [Europe PMC free article] [Abstract] [Google Scholar]
  • ZYGMUNT P.M., EDWARDS G., WESTON A.H., LARSSON B., HOGESTATT E.D. Involvement of voltage-dependent potassium channels in the EDHF-mediated relaxation of rat hepatic artery. Br. J. Pharmacol. 1997a;121:141–149. [Europe PMC free article] [Abstract] [Google Scholar]
  • ZYGMUNT P.M., HOGESTATT E.D. Role of potassium channels in endothelium-dependent relaxation resistant to nitroarginine in the rat hepatic artery. Br. J. Pharmacol. 1996;117:1600–1606. [Europe PMC free article] [Abstract] [Google Scholar]
  • ZYGMUNT P.M., HOGESTATT E.D., WALDECK K., EDWARDS G., KIRKUP A.J., WESTON A.H. Studies on the effects of anandamide in rat hepatic artery. Br. J. Pharmacol. 1997b;122:1679–1686. [Europe PMC free article] [Abstract] [Google Scholar]
  • ZYGMUNT P.M., RYMAN T., HOGESTATT E.D. Regional differences in endothelium-dependent relaxation in the rat: contribution of nitric oxide and nitric oxide-independent mechanisms. Acta Physiol. Scand. 1995;155:257–266. [Abstract] [Google Scholar]

Articles from British Journal of Pharmacology are provided here courtesy of The British Pharmacological Society

Citations & impact 


Impact metrics

Jump to Citations

Citations of article over time

Alternative metrics

Altmetric item for https://www.altmetric.com/details/3343933
Altmetric
Discover the attention surrounding your research
https://www.altmetric.com/details/3343933

Article citations


Go to all (161) article citations

Other citations