revisão - angiotensina e endotelina

12
Review Article Vascular smooth muscle cell signaling mechanisms for contraction to angiotensin II and endothelin-1 Brandi M. Wynne, MS * , Chin-Wei Chiao, PhD, and R. Clinton Webb, PhD Department of Physiology, Medical College of Georgia, Augusta, Georgia, USA Manuscript received June 28, 2008 and accepted September 25, 2008 Abstract Vasoactive peptides, such as endothelin-1 and angiotensin II, are recognized by specific receptor proteins located in the cell membrane of target cells. After receptor recognition, the specificity of the cellular response is achieved by G-protein coupling of ligand binding to the regulation of intracellular effectors. These intracellular effectors will be the subject of this brief re- view on contractile activity initiated by endothelin-1 and angiotensin II. Activation of receptors by endothelin-1 and angio- tensin II in smooth muscle cells results in phospholipase C activation leading to the generation of the second messengers inositol trisphosphate (IP 3 ) and diacylglycerol (DAG). IP 3 stimulates intracellular Ca 2þ release from the sarcoplasmic retic- ulum and DAG causes protein kinase C activation. Additionally, different Ca 2þ entry channels, such as voltage-operated, re- ceptor-operated, and store-operated Ca 2þ channels, as well as Ca 2þ -permeable nonselective cation channels, are involved in the elevation of intracellular Ca 2þ concentration. The elevation in intracellular Ca 2þ is transient and initiates contractile ac- tivity by a Ca 2þ -calmodulin interaction, stimulating myosin light chain (MLC) phosphorylation. When the Ca 2þ concentra- tion begins to decline, Ca 2þ sensitization of the contractile proteins is signaled by the RhoA/Rho-kinase pathway to inhibit the dephosphorylation of MLC phosphatase (MLCP), thereby maintaining force generation. Removal of Ca 2þ from the cy- tosol and stimulation of MLCP initiates the process of smooth muscle relaxation. In pathologic conditions such as hyperten- sion, alterations in these cellular signaling components can lead to an overstimulated state causing maintained vasoconstriction and blood pressure elevation. J Am Soc Hypertens 2009;3(2):84–95. Ó 2009 American Society of Hypertension. All rights reserved. Keywords: Calcium channel; Rho-kinase; vasoactive peptides; phosphoinositides. Introduction In the vasculature, the small arteries and arterioles regulate the majority of blood flow resistance in the circu- lation. Circulating neurotransmitters, hormones, endothe- lium-derived factors, and even shear stress itself plays a role in modulating smooth muscle tone, and consequently, lumen diameter. 1–3 The final product is the control of blood pressure (BP) and organ blood flow. Arteries are composed of three layers: 1) the tunica ad- ventitia, 2) tunica media, and 3) tunica intima. Although the outer and inner layers are composed mainly of connec- tive tissue and endothelial cells, respectively, the tunica me- dia is composed of smooth muscle cells. 3 All smooth muscle cells, regardless of the stimulus, produce force or contraction through cross-bridge cycling between actin and myosin filaments. Although vascular smooth muscle cells are capable of dynamic changes in gene expression to either contractile or differentiation proteins, the contrac- tile phenotype in vascular smooth muscle predominates. 4 The contractile response of vascular smooth muscle is the product of myosin light chain kinase (MLCK) and myosin light chain phosphatase (MLCP) activation. In smooth mus- cle, this process is initiated by a calcium (Ca 2þ )-mediated change in the thick (myosin) filaments 2 (Figure 1). In this review, we will discuss the molecular mechanisms by which two common vasoactive peptides: angiotensin II and endothelin-1 produce smooth muscle contraction. This study was supported by the National Institutes of Health (Grant Nos. HL71138 and HL74167). Conflict of interest: none. *Corresponding author: Brandi M. Wynne, MS, Department of Physiology, Medical College of Georgia, Augusta, Georgia 30912. Tel: 706-721-3547; fax: 706-721-7299. E-mail: [email protected] 1933-1711/09/$ – see front matter Ó 2009 American Society of Hypertension. All rights reserved. doi:10.1016/j.jash.2008.09.002 Journal of the American Society of Hypertension 3(2) (2009) 84–95

Upload: carol-carvalho-correia

Post on 22-Jun-2015

6 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Revisão - Angiotensina e Endotelina

Journal of the American Society of Hypertension 3(2) (2009) 84–95

Review Article

Vascular smooth muscle cell signaling mechanisms for contractionto angiotensin II and endothelin-1

Brandi M. Wynne, MS*, Chin-Wei Chiao, PhD, and R. Clinton Webb, PhDDepartment of Physiology, Medical College of Georgia, Augusta, Georgia, USA

Manuscript received June 28, 2008 and accepted September 25, 2008

Abstract

Vasoactive peptides, such as endothelin-1 and angiotensin II, are recognized by specific receptor proteins located in the cellmembrane of target cells. After receptor recognition, the specificity of the cellular response is achieved by G-protein couplingof ligand binding to the regulation of intracellular effectors. These intracellular effectors will be the subject of this brief re-view on contractile activity initiated by endothelin-1 and angiotensin II. Activation of receptors by endothelin-1 and angio-tensin II in smooth muscle cells results in phospholipase C activation leading to the generation of the second messengersinositol trisphosphate (IP3) and diacylglycerol (DAG). IP3 stimulates intracellular Ca2þ release from the sarcoplasmic retic-ulum and DAG causes protein kinase C activation. Additionally, different Ca2þ entry channels, such as voltage-operated, re-ceptor-operated, and store-operated Ca2þ channels, as well as Ca2þ-permeable nonselective cation channels, are involved inthe elevation of intracellular Ca2þ concentration. The elevation in intracellular Ca2þ is transient and initiates contractile ac-tivity by a Ca2þ-calmodulin interaction, stimulating myosin light chain (MLC) phosphorylation. When the Ca2þ concentra-tion begins to decline, Ca2þ sensitization of the contractile proteins is signaled by the RhoA/Rho-kinase pathway to inhibitthe dephosphorylation of MLC phosphatase (MLCP), thereby maintaining force generation. Removal of Ca2þ from the cy-tosol and stimulation of MLCP initiates the process of smooth muscle relaxation. In pathologic conditions such as hyperten-sion, alterations in these cellular signaling components can lead to an overstimulated state causing maintainedvasoconstriction and blood pressure elevation. J Am Soc Hypertens 2009;3(2):84–95. � 2009 American Society ofHypertension. All rights reserved.Keywords: Calcium channel; Rho-kinase; vasoactive peptides; phosphoinositides.

Introduction

In the vasculature, the small arteries and arteriolesregulate the majority of blood flow resistance in the circu-lation. Circulating neurotransmitters, hormones, endothe-lium-derived factors, and even shear stress itself playsa role in modulating smooth muscle tone, and consequently,lumen diameter.1–3 The final product is the control of bloodpressure (BP) and organ blood flow.

This study was supported by the National Institutes of Health(Grant Nos. HL71138 and HL74167).

Conflict of interest: none.*Corresponding author: Brandi M. Wynne, MS, Department of

Physiology, Medical College of Georgia, Augusta, Georgia 30912.Tel: 706-721-3547; fax: 706-721-7299.

E-mail: [email protected]

1933-1711/09/$ – see front matter � 2009 American Society of Hyperdoi:10.1016/j.jash.2008.09.002

Arteries are composed of three layers: 1) the tunica ad-ventitia, 2) tunica media, and 3) tunica intima. Althoughthe outer and inner layers are composed mainly of connec-tive tissue and endothelial cells, respectively, the tunica me-dia is composed of smooth muscle cells.3 All smoothmuscle cells, regardless of the stimulus, produce force orcontraction through cross-bridge cycling between actinand myosin filaments. Although vascular smooth musclecells are capable of dynamic changes in gene expressionto either contractile or differentiation proteins, the contrac-tile phenotype in vascular smooth muscle predominates.4

The contractile response of vascular smooth muscle is theproduct of myosin light chain kinase (MLCK) and myosinlight chain phosphatase (MLCP) activation. In smooth mus-cle, this process is initiated by a calcium (Ca2þ)-mediatedchange in the thick (myosin) filaments2 (Figure 1).

In this review, we will discuss the molecular mechanismsby which two common vasoactive peptides: angiotensin IIand endothelin-1 produce smooth muscle contraction.

tension. All rights reserved.

Page 2: Revisão - Angiotensina e Endotelina

85B.M. Wynne et al. / Journal of the American Society of Hypertension 3(2) (2009) 84–95

Because intracellular Ca2þ is fundamental to the contractileprocess, we will also provide a brief review of the cellularmechanisms that regulate this cation.

Angiotensin II Signaling

Angiotensin II, which is the predominate bioactive pep-tide in the renin-angiotensin system (RAS), promotes themaintenance of systemic pressure through various mecha-nisms in the cardiovascular and renal systems.5 Althoughangiotensin II is crucial to salt and water homeostasis,this peptide is also implicated in several cardiovascularconditions, such as hypertension, atherosclerosis and heartfailure, and more specifically in vascular smooth musclecontraction; vascular remodeling including the inductionof hypertrophy and hyperplasia. Canonical angiotensin IIsignaling occurs through a membrane bound heterotrimericG-protein coupled receptor (GPCR). To date, there are twoGPCRs known to mediate angiotensin II function: the AT1

and AT2 receptors.5,6

The AT1 receptor is expressed in a variety of tissues, in-cluding the kidney, heart, adrenal gland, brain, lung, andadipose; however, the focus will be on AT1 receptor activa-tion in arterial smooth muscle. The AT2 receptor is highlyexpressed in fetal tissues, then declines quickly after birth.In adults, the AT2 receptor has a varied tissue distribution aswell, and is detectable in the kidney and adrenals, pancreas,ovary, brain, heart, and vasculature. In vessels, the AT2 re-ceptor is not localized on the smooth muscle, but is mainlyexpressed in the adventitia. After injury, levels of this re-ceptor have been observed to increase, which may explaincurrent dogma suggesting that the AT2 receptor is involvedin cellular growth. In contrast to the vasoconstrictive effectsof the AT1, the AT2 receptor has been shown to modulatevasodilation.7 Less is known about AT2 receptor mecha-nisms than the AT1 receptor; however, it is known that itcouples to Gi and activates tyrosine and serine/threonine

Figure 1. Overview of major contributors to vascular smooth muscle cmediated change in the thick filaments, or myosin. With myosin light cof interacting. ATP hydrolysis is the source for force generation in smyosin head. Relaxation occurs via the action of myosin light chain phing it. ATP, adenosine-5‘-triphosphate; Ca2þ, calcium.

phosphatases. This receptor isoform has also been docu-mented to trigger the kinin-NO-cGMP pathway.8 However,it is through the AT1 receptor with which the majority ofphysiologic and pathologic actions of angiotensin II is me-diated, and will be the focus of the signaling in this review.

The angiotensin II signaling cascade via the AT1 receptoris a consequence of the particular G-protein or signalingcascade that is activated: Gq/11, Gi, G12, and G13.5,6 TheAT1 receptor signals through three main pathways: classicalphospholipase C (PLC) signaling leading to inositol tri-sphosphate (IP3) and diacylglycerol (DAG) cleavage, phos-pholipase D, also culminating in DAG generation; andphosphatidylcholine, phosphatidic acid and phospholipaseA2, which is responsible for arachidonic acid and prosta-glandin production. Tyrosine phosphorylation, ie, mitogenactivated protein kinase (MAPK), which is distinctive forits role in growth factor and cytokine activation, has beenassociated with increased levels of AT1 receptor activation.Nonreceptor tyrosine kinase activity has also been associ-ated with angiotensin II, including but not limited to: theSrc family of kinases, proline-rich tyrosine kinases(Pyks), extracellular signal-regulated kinases (ERKs),paxillin, focal adhesion kinase (FAK) phosphoinositide 3kinase (PI3K) and an inflammatory cytokine pathway, januskinases/signal transducers and activators of transcription(JAK/STAT).7 Angiotensin II is a pleiotropic signaling mol-ecule, whose varied pathways is complex and specific, butyet also converges to bring forth multiple responses. Thisreview will focus on the PLC pathway, culminating in vas-cular smooth muscle contraction.

Recent literature suggests that the AT1 receptor couplesto Gq to mediate vasoconstriction, which is the pathwayleading to smooth muscle contraction via PLC.9 In addition,angiotensin II was found to elicit a contractile response viaactivation of G12/G13 G proteins and the regulator of G-pro-tein signaling-Rho-guanine nucleotide exchange factor(RGS-RhoGEF) signaling pathway. Recently, insights into

ontraction. In smooth muscle, contraction is initiated by a Ca2þ-hain phosphorylation, the actin and myosin filaments are capablemooth muscle; with contraction, inorganic phosphate leaves theosphatase, which dephosphorylates myosin light chain, inactivat-

Page 3: Revisão - Angiotensina e Endotelina

Figure 2. Molecular mechanisms of smooth muscle contraction. Vascular smooth muscle contraction is the summation of MLCK andMLCP activity. With receptor binding, an increase in intracellular Ca2þ occurs, both via channels located in the membrane and intracel-lular stores in the sarcoplasmic reticulum. The Ca2þ interacts with calmodulin, forming a Ca2þ-calmodulin complex, which activatesMLCK. MLCK can then p-MLC, allowing for the close interaction of the actin and myosin filaments for force generation. Relaxationoccurs with MLCP dephosphorylating MLC. In some instances, force generation can be tonic; this is mediated in a Ca2þ-independentmanner. Rho-kinase becomes activated via the small, activated RhoA protein, which subsequently phosphorylates MLCP, renderingthe enzyme inactive and incapable of de-phosphorylating MLC. In addition, PKC and Rho kinase work in concert to activate CPI-17,which inhibits MLCP. Thus, the vascular smooth muscle cannot relax and a tonix contraction occurs. Ca2þ, calcium; MLCK, myosin lightchain kinase; MLCP, myosin light chain phosphatase; p-MLC, phosphorylate myosin light chain.

86 B.M. Wynne et al. / Journal of the American Society of Hypertension 3(2) (2009) 84–95

this mechanism were discovered and will be discussed ina later section.10,11 After Gq/G11 GPCR activation, secondmessengers are generated, beginning with PLC activation.PLC is a membrane-bound enzyme responsible for cleavingmembrane lipid phosphoinositide 4, 5- bisphosphate, result-ing in the generation of IP3 and DAG.2,12 IP3 and DAG reg-ulate two distinct but parallel pathways leading to increasedintracellular Ca2þ and culminating in MLCphosphorylation.2,13,14

IP3 regulates calcium release into the cytosol from thesarcoplasmic reticulum (SR), mediated via the ryanodinereceptor (RyR). The RyR, which has been shown to be lo-calized centrally as well as peripherally on the SR, isthought to facilitate optimal binding of IP3.15,16 Activationof the RyR opens Ca2þ channels, causing a rapid increasein Ca2þ concentration. After cytosolic Ca2þ concentrationspeak and then decline, there is a sustained elevation ofCa2þ above that of basal levels.2 This sustained increasein Ca2þ is due to receptor-operated Ca2þ channels on

plasma membrane facilitating increased cytosolic Ca2þ

levels from the extracellular space.2,14 The EF handCa2þ-binding protein, calmodulin, is the primary targetfor elevated intracellular Ca2þ. The Ca2þ-calmodulin com-plex is then able to activate MLCK.2 It is also of importanceto note that the Ca2þ-tension relationship changes for stim-ulation type and during the time of the contraction. In gen-eral, studies show that the receptor-mediated stimulationsproduce a greater tension for a given Ca2þ concentration.Moreover, during the sustained phase of contraction, lowerlevels of Ca2þ are needed, which is referred to as Ca2þ sen-sitization. These are two differential points of regulation forvascular smooth muscle contraction17 (Figure 2).

DAG mediates vascular smooth muscle contraction viaactivation of protein kinase C (PKC). PKC is a serine/thre-onine kinase known to exist in several isoforms, all whichcontribute to various physiological activities and are acti-vated differentially. Of the three known vascular smoothmuscle isoforms, PKCa and PKCb are dependent on

Page 4: Revisão - Angiotensina e Endotelina

Figure 3. The concept of ROC channel, VOC channel, and SOC channel. ROC channels are activated via receptor stimulation of GPCRsdirectly or through the production of second messengers such as IP3 or DAG. IP3 can then bind to and activate the IP3 receptor on thesarcoplasmic reticulum membrane, causing discharge and release of the stored Ca2þ. The release of Ca2þ from the sarcoplasmic reticuluminduces Cl� efflux or the influx of Naþ and Caþ from the ROC channels, which can then activate VOC channels. The second messenger, IP3,activates the IP3 receptor on the sarcoplasmic reticulum membrane, causing discharge and release of the stored Ca2þ. After Ca2þ depletionfrom the sarcoplasmic reticulum, the SOC channels are activated. A, agonist; Ca2þ, calcium; DAG, diacylglycerol; GPCRs, G-protein–coupled receptors; IP3, inositol 1,4,5-trisphosphate; IP3R, IP3 receptor; PIP2, phosphatidylinositol 4,5-bisphosphate; PLC, phospholipaseC; R, receptor; ROC, receptor-operated channels; TK, tyrosine kinase. SOC, store-operated channels; VOC, voltage-operated channels.

87B.M. Wynne et al. / Journal of the American Society of Hypertension 3(2) (2009) 84–95

DAG activation and intracellular Ca2þ concentrations,whereas PKCe is DAG-dependent only. PKC modulatesvascular smooth muscle contraction by directly phosphory-lating MLCK, which subsequently phosphorylates MLC.Furthermore, PKC activation leads to the activation ofseveral other target proteins promoting smooth musclecontraction. PKC phosphorylates and activates ERK1/2,Rho-kinase (p160ROCK), and calmodulin-dependentprotein kinase II. In addition, intracellular Ca2þ elevationwas suggested to regulate angiotensin II-induced epidermalgrowth factor receptor and ERK activation.11 Various ionchannels and ion transporters have been demonstrated asdownstream targets of PKC. This phenomenon has beenillustrated with the use of the specific PKC agonists (ie,phorbol esters) which induce smooth muscle contraction.2

All the earlier events described culminating in MLCKactivation, resulting in contraction via the phosphorylationof Ser19 of the 20 kDa regulatory protein of MLC. Thephosphorylation of MLC allows for the interaction betweenactin and myosin filaments. Intrinsic adenosine-5‘-triphos-phatase (ATPase) activity of myosin is crucial for this pro-cess, because the hydrolysis and subsequent release of ATPresults in the cycling of the myosin cross-bridges withactin, causing force generation.3,18–21

Smooth muscle cells also contain Ca2þ-independentmechanisms to regulate contractility. The process of force

generation is mediated by MLCK activation, and subse-quent actin-myosin cross-bridging, but the process of forcemaintenance is mediated in a Ca2þ-independent man-ner.2,18,22 MLCP is the contender, regulating the inactiva-tion of MLC by removing the high-energy phosphategroup, promoting smooth muscle relaxation. MLCP is com-posed of two subunits; one is the catalytic 38 kDa subunitof type 1 protein phosphatase (PP1c, d isoform) and twoother noncatalytic subunits.12,17 This mechanism generatesforce maintenance by means of Ca2þ-sensitization of thecontractile proteins. This is signaled by the RhoA/Rho ki-nase pathway, which inhibits the dephosphorylation ofMLC by MLCP. RhoA is a small, monomeric G protein,which is regulated by the binding of GTP. This process isfacilitated by nucleotide exchange factors, RhoGEFs,which enable the exchange of guanosine diphosphate(GDP) for guanosine triphosphate (GTP), activatingRhoA. Upon RhoA activation, the RhoA-GTP complex isable to activate Rho kinase, a serine/threonine kinase.Rho kinase can then phosphorylate the myosin-bindingsubunit of MLCP, inhibiting it, and thus preventing the de-phosphorylation of MLC, causing maintenance of contrac-tion.2,23,24 In addition to the regulatory effects of Rhokinase, CPI-17 is another protein that regulates myosinphosphatase (MYPT) phosphorylation status by inhibitingPP1c, thus inactivating MLCP. CPI-17 phosphorylation

Page 5: Revisão - Angiotensina e Endotelina

Figure 4. New pathways in vascular smooth muscle signaling. Orai1 and STIM1 work together to function as Ca2þ sensors within thecell. Orai1 makes up the pore of the CRAC channel, allowing for Ca2þ entry, whereas STIM1 senses Ca2þ levels within the SR. Theirinteraction facilitates this process. The IKCa channels are intermediate conductance Ca2þ-activated Kþ channels which are of impor-tance in small resistance vessels. G12/13 signaling through LARG was recently found to be a mechanism for RhoA/Rho kinase activation,leading to MLCP inactivation. Ca2þ, calcium; CRAC, Ca2þ-release activated Ca2þ; IKCa, intermediate conductance Ca2þ-activated Kþ

channel; p-MLC, myosin light chain phosphatase; STIM, stromal-interacting molecule.

88 B.M. Wynne et al. / Journal of the American Society of Hypertension 3(2) (2009) 84–95

occurs in a Ca2þ-dependent and Ca2þ-independent mecha-nism. Ca2þ-dependent PKC was suggested to regulate therapid phosphorylation, whereas phosphorylation by PKCand Rho kinase occurs independently of Ca2þ in the laterphase. This has been demonstrated with the use of Rho ki-nase and PKC inhibitors; however, the effects were depen-dent on the type of agonist and tissue used. Its activity andexpression is a factor in the contractile state of vascularsmooth muscle and has also been shown to be of impor-tance in Ca2þ sensitization.12,17 Much needs to be exploredregarding MLCP regulation, but regardless, Rho kinase andCPI-17 are acknowledged to be of importance in vascularsmooth muscle contraction and force maintenance(Figure 2).

Endothelin-1 Signaling

Endothelin-1, which is a 21-amino acid peptide, isknown as one of the most potent endogenous vasoconstric-tors as described by Yanagisawa et al in 1988.25–27 Since itsdiscovery, the endothelin-1 system has become increasinglycomplex; several endothelin-1 isoforms and receptors havebeen identified in a variety of tissues, including neuronal,renal, and vascular tissues. Interestingly, although somedata show a correlation between endothelin-1 levels and hy-pertension, these data are not conclusive.4 Overall, endothe-lin-1 signaling in the vasculature is essentially the same asfor angiotensin II signaling. A brief overview will be dis-cussed, but the reader is directed to angiotensin II-GPCR

coupled signaling pathways for a more in depth explana-tion. Differing pathways regarding to endothelin-1 signal-ing itself will be discussed further.

Endothelin-1 is the main isoform secreted by the endo-thelium, and has been shown to act in a paracrine or auto-crine manner on its receptors in vascular smooth muscle.There are three known endothelin-1 receptors that havean assorted tissue distribution and functional role: ETA

and ETB are present in mammals and a putative ETC innon-mammals. The typical receptor found on vascularsmooth muscle cells is the ETA receptor, which mediatesthe vasoconstrictor effects of endothelin-1.4,25,28 Endothe-lin-1 receptor activation can lead to diverse responses inthe cell through interaction with pathways that are both per-tussis toxin-sensitive and toxin-insensitive, leading to theconclusion that endothelin-1 acts through several GPCRs.Generally, ETA receptors have been associated with vaso-constriction and cell growth, whereas ETB receptors are in-volved in the clearance of endothelin-1, inhibition ofendothelial cell apoptosis, the release of NO and prosta-glandins leading to vasorelaxation, and inhibition of the ex-pression of endothelin-1 converting enzyme; however, bothreceptors have been found to elicit vasoconstriction.29–31

ETA receptors have been shown to be functionallycoupled to the Gq/11 protein leading to PLC-b activation;Gs linked leading to increased cyclic adenosine monophos-phate (cAMP) and also to Gi, thus inhibiting adenylate cy-clase.30,32–34 Activation of Gq/11, ending in IP3 and DAGcleavage, can stimulate Ca2þ release intra- and

Page 6: Revisão - Angiotensina e Endotelina

89B.M. Wynne et al. / Journal of the American Society of Hypertension 3(2) (2009) 84–95

extracellularly, as described for angiotensin II signaling. Inaddition to second messenger generation, endothelin-1 hasalso been demonstrated to activate Ca2þ channels on theplasma membrane and stimulate Ca2þ flux from the extra-cellular space.4,35 There is speculation that ETA receptoractivation can activate the Naþ/Hþ exchanger and activatethe enzymes phospholipase D generating DAG and phos-pholipase A2 releasing arachidonic acid.4 ETB receptorsin rat carotid were shown to mediate vasorelaxation viaa cyclic guanosine monophosphate-nitric oxide (cGMP-NO) pathway, the production of vasodilator cyclooxyge-nase products and the activation of voltage-activated Kþ

channels. Recently, endothelin-1 was shown to inhibit nic-otinamide adenine dinucleotide phosphate (NADPH) oxi-dase activity and superoxide generation via ETB1

receptors.36 The details of these mechanisms are still poorlyunderstood.31

Both agonists, angiotensin II and endothelin-1, mediatevasoconstriction via an increase in cytosolic Ca2þ althoughmechanisms of this process are different between the twoagonists. Endothelin-1 increases intracellular Ca2þ by stim-ulating influx through Ca2þ channels and is known to pro-duce a much more sustained contraction by the vascularsmooth muscle cells, whereas angiotensin II elicits a potentbiphasic response that is generated primarily by mobiliza-tion of Ca2þ from an intracellular store. Consequently,the actions of angiotensin II actions are relatively rapid incomparison.37–41

Given that both of these agonists mediate downstream sig-naling pathways via the same second messengers, it is easy toappreciate how cross-talk occurs between them. Both endo-thelin-1 and angiotensin II signal through distinct Gq-coupledGPCRs leading to phosphoinositide production and MAPKactivation. However, the mechanism by which this occursvaries at some points. It has been demonstrated that endothe-lin-1 and angiotensin II both stimulate this pathway via Ras-Raf; yet, angiotensin II produces phosphorylation of ERK1/2, SAPK/JNK, and p38MAPK via c-Src–dependent path-ways. In contrast, endothelin-1 was shown to induceMAPK phosphorylation through c-Src–independentpathways42 (Figure 3).

Calcium Mobilization

Given that Ca2þ is the trigger for smooth muscle contrac-tion, it is necessary to understand the mechanisms by whichintracellular Ca2þ levels increase. Hence, this is the focusof much research, and the subject of the remainder of thisreview. In vivo, arterial smooth muscle cells exist with anaverage intracellular Ca2þ concentration that is several or-ders of magnitude lower than that in the extracellular fluid.Intracellular Ca2þ concentration levels do not exist homo-genously throughout the cell, but exhibit dynamic changesin the cell, temporally and spatially, because of Ca2þ flux.The changes seen in Ca2þ flux are a component of several

different events, which are all dependent on intracellular ul-tra-structure as well as the spatial relationship of ion pumpsand channels in the plasma membrane.4

Ca2þ entry through channels in the plasma membraneand release from the SR are the major sources of intracel-lular Ca2þ.1,4,15 Of the different Ca2þ entry channels, thevoltage-operated Ca2þ (VOC), receptor-operated Ca2þ

(ROC), store-operated Ca2þ (SOC) channels, and a Ca2þ-permeable nonselective cation channels (NSCC) modulatemost Ca2þ mobilization within the cell. We will discussthese particular vascular smooth muscle Ca2þ entry chan-nels further in the following section (Figure 3).

Voltage-Operated Ca2D Channels

VOC channels represent a major route by which Ca2þ

enters vascular smooth cells.43 VOC channel function isregulated by membrane potential such that hyperpolariza-tion closes them and depolarization opens them; the latterleading to vasoconstriction.1

The majority of agonist-induced Ca2þ influx probablyoccurs through L-type VOC channels. Dihydropyridine-sensitive, L-type VOC channels are regulated by vasocon-strictors that are known to activate the PKC pathway. Vaso-dilators have also been shown to inactivate these channels,but through cAMP production.1,44 These gated channelsalso depolarize in response to membrane stretch, lendingsupport to the hypothesis that these channels are importantfor the myogenic response and vascular tone. Inhibition ofVOC channels can occur primarily because of increasingintracellular Ca2þ concentration and activation of cGMP-dependent protein kinase.1,2,44

Endothelin-1 has been shown to activate the VOC chan-nel in smooth muscle cells from porcine coronary arteries,and has also been shown to augment Ca2þ channel currentsin the smooth muscle cell membrane of guinea pig portalvein. In a model of cultured thoracic aorta vascular smoothmuscle cells, Kawanabe et al demonstrated that endothelin-1 produces a sustained increase in intracellular Ca2þ levelsfrom VOC channels, as well as others.41,45

Although endothelin-1 increases intracellular Ca2þ bystimulating influx through Ca2þ channels, the role ofVOC channels is limited, and current data suggest thatother channels are more important; namely the NSCC, asevidenced by experiments using nifedipine.41,46 Eventhough described more than 60 years ago, the exact mech-anisms of action of this peptide are not completelyunderstood.

Angiotensin II-GPCR stimulation leading to PLC is anaccepted mechanism acknowledged to enhance the L-typeCa2þ channel. Interestingly, intracellular angiotensin IIhas been demonstrated to stimulate VOC channels simi-larly, but yet independently of extracellular ligand bindingof angiotensin II to the AT1 receptor, possibly through theactions of PLC, PKC, and/or tyrosine kinase.47,48

Page 7: Revisão - Angiotensina e Endotelina

90 B.M. Wynne et al. / Journal of the American Society of Hypertension 3(2) (2009) 84–95

As shown in the smooth muscle cells of the anococcy-geus, there are two very distinct mechanisms for inwardCa2þ currents and Ca2þ store depletion.49 The increase inCa2þ concentration after the activation of VOC channelscan also lead to a phenomenon known as Ca2þ inducedCa2þ release, where the increase in Ca2þ concentrationleads to Ca2þ release from intracellular stores. The RyRon the SR mediates this process, as they are activated byCa2þ.50 This produces a membrane depolarization that pro-motes further VOC channel opening and Ca2þ entry.

Receptor-Operated Ca2D Channels

Only 20 years ago, the exact mechanism of Ca2þ channelactivation by extracellular ligands, such as hormones andneurotransmitters, was largely unknown. In addition, therewas still uncertainty concerning the mechanism by whichCa2þ entry was mediated until a model, proposed by Put-ney et al, detailing the mechanism by which activation ofsurface membrane receptors causes a sustained Ca2þ entryinto cells from the extracellular space.51 Furthermore, Ben-ham and Tsien suggested that agonists might activate Ca2þ

influx directly, by receptor interaction with Ca2þ-perme-able NSCC.52 Currently, ROC channels are defined by thefollowing: channels where molecules are separate fromthe ligand-binding protein are capable of activating a rangeof GPCRs via circulating agonists, and are neither VOC norSOC channels.28 After ligand binding, ROC channels aregenerally thought to be activated by GPCRs, which are cou-pled to PLC, leading to IP3 and DAG generation.53,54

Several members of the transient receptor potential chan-nel (TRPC) family are accepted to form ROC channels,including TRPC3, TRPC6, and TRPC7. Angiotensin IIhas been proposed to activate TRPC6 in rabbit mesentericarteries, whereas endothelin-1 activates TRPC3 andTRPC7.55–57

Store-Operated Ca2D Channels

SOC channels represent yet another mechanism of excita-tion-contraction coupling in smooth muscle. SOC channelsare activated by means of intracellular Ca2þ level depletion,and, as a result, are inhibited when Ca2þ stores are filled.This process was called capacitative Ca2þ entry, beforestore-operated Ca2þ entry (SOCE) was coined. It shouldbe noted that SOCE does not denote any one mechanismof Ca2þ entry, nor does it refer to any particular chan-nel.1,58–62 These channels are highly selective for Ca2þ,and have been found to be bradykinin and ATP sensitive.60

RyR- or IP3-sensitive stores can induce Ca2þ release se-quentially, possibly amplifying the rise in intracellularCa2þ and further stimulating Ca2þ channel-dependent Cl�

channels. SOC channels were described in 1986, when Put-ney et al demonstrated that the same Ca2þ entry mechanismnormally activated as a result of Ca2þ-mobilizing agonists

can be triggered equally as well by depleting the intracellu-lar Ca2þ pool, even in the absence of receptor activation orelevated cellular levels of inositol polyphosphates.63–65 SOCchannel entry has been proposed to increase cytoplasmicCa2þ levels not only for contraction, but also cell prolifera-tion and apoptosis.4 SERCA pumps, which are found on themembrane of the sarcoplasmic reticulum, function by pump-ing the Ca2þ ions back into the SR after a contractile eventreplenishing Ca2þ concentrations.14,15 SERCA pumps canbe inhibited by cyclopiazonic acid or thapsigargin, whichdeplete intracellular Ca2þ stores and thereby activate SOCchannels.66,67 Ca2þ entering through the SOC channelscan then be pumped into the stores, replenishing them. Ac-cording to functional and pharmacologic experiments, SOCchannel entry elicited by specific inhibitors of SERCApumps induced a sustained elevation of intracellular Ca2þ,which is also dependent on extracellular Ca2þ entry and isattributed to SOCE.68 Thus, Ca2þ signals generated in re-sponse to receptors involve two coupled components: a tran-sient release of Ca2þ stored in the SR, followed by a slowlydeveloping extracellular Ca2þ entry.14,15,69,70 With depletedCa2þ stores, Ca2þ influx factor is generated and diffuses tothe plasma membrane where, through a series of reactions,Ca2þ influx factor activates Ca2þ-independent phospholi-pase A2. This generation of lysophospholipids, in turn, acti-vates the SOC channels.62

The family of TRPC is well known for their role inSOCE, although the mechanism allowing for TRPC activa-tion from store depletion is largely unknown. There isevidence for and against TRPCs actually being a SOCchannel, including TRPC1.56,58,62 Also, interesting datafrom recent high throughput RNAi screens of thapsigar-gin-activated Ca2þ entry revealed a stromal-interactingmolecule (STIM) in Drosophila S2 cells and in mammalianHeLa cells, which are now thought to play an essential rolein SOCE and conductance through Ca2þ-release activatedCa2þ (CRAC) channels.71–73 This single membrane span-ning protein is now thought to activate the SOC channelsby actually sensing Ca2þ within the stores. Pharmacologicexperiments showed that the actual contribution of SOCEto excitation/contraction coupling seems to depend on thesmooth muscle type, and may be more important in tonicsmooth muscle.62 A canonical TRP channel, TRPC7, whichis activated by AT1-coupled GPCR activation andDAG, was recently hypothesized to mediate angiotensinII–induced myocardial apoptosis. Moreover, angiotensinII has been postulated to activate TRPC1, as evidencedby data showing that angiotensin II stimulation increasedSOCE together with TRPC1 expression. These data wereobtained using a cell culture model, but given the heteroge-neity of TRPC channels in vascular smooth muscle, there isno reason to believe that this occurrence does not exist invivo.74 In a recent study in vascular smooth muscle cells,endothelin-1 activation of TRPC1 was found to not onlybe involved in a SOCE, but also in a ROC channel entry,

Page 8: Revisão - Angiotensina e Endotelina

91B.M. Wynne et al. / Journal of the American Society of Hypertension 3(2) (2009) 84–95

which requires IP3 receptor activation. A possible explana-tion for this exciting phenomenon is the report that TRPCsmay exist in heterotrimeric complexes, possibly accountingfor the varying pharmacologic properties seen.56

Nonselective Cation Channels

The NSCC, so called because it is ‘‘nonselective,’’ beingequally permeable to monovalent cations, such as Naþ andKþ in the extra- and intracellular compartments.25,28 Thischannel has been revealed to be activated by stimulationof native ETA receptors in vascular smooth muscle, andthe majority of endothelin-1 response being mediated byNSCCs; namely, NSCC-1 and NSCC-2.28,41,46,75 It waspreviously shown, pharmacologically and with electrophys-iology, that the response to endothelin-1 is dose-dependent.More specifically, with lower concentrations of endothelin-1, Ca2þ entry via IP3 formation from intracellular stores oc-curs; however, with higher endothelin-1 concentrationsboth the former in addition to extracellular Ca2þ entry oc-curs as well.25,28,38,76,77

The molecular mechanisms of NSCC activation are stillnot entirely clear. Recent evidence in rabbit internal carotidartery shows that endothelin-1 may induce an intracellularresponse after GPCR activation via Pyk2 and the ETA

receptor. Data from the same laboratory indicate thatPI3K also regulates the activation of Ca2þ entry after endo-thelin-1 GPCR activation and stimulation of NSCC-2.46,78

Hot Topics in Vascular Smooth Muscle Signaling

Within just the past year, exciting new insights into vas-cular smooth muscle signaling have been discovered. Wewill discuss several recent advancements in the field thathave been especially stimulating (Figure 4).

As described previously, GPCR signaling can elicit stim-ulatory or inhibitory signals in the cell. With regards to vas-cular smooth muscle, the Gq/G11 G proteins have beenaccepted as producing contraction via the Ca2þ-dependentPLC pathway; however, Ca2þ-independent mechanisms,not only for force maintenance, but for contraction haveemerged. By means of a novel murine knockout modelfor Gq/G11 and G12/G13 G proteins in smooth muscle cells,Wirth et al elaborated upon the knowledge of how angio-tensin II and endothelin-1 mediate contraction via G pro-teins. Using aortic segments, they demonstrated that inG12/G13 knockout mice, angiotensin II and endothelin-1were able to elicit a contraction; however, both potencyand efficacy were affected. In the Gq/G11 knockout mice,although endothelin-1 still produced a severely reducedcontraction, angiotensin II was not capable of elicitingany contraction. It was also demonstrated that G12/G13 cou-pled G proteins activate the RhoA/Rho kinase signalingpathway via interaction of RhoGEFs; in particular,

leukemia-associated RhoGEF (LARG). Using a LARG mu-rine knockout model, they showed that LARG is necessaryfor full contraction with angiotensin II and endothelin-1, asdemonstrated by the severely restricted constriction elicitedin aortic segments from LARG knockout mice.10,79

As described previously, Pyk2 is a Ca2þ-sensitive nonre-ceptor protein tyrosine kinase that is known to associatewith focal adhesion sites and is a downstream effector ofangiotensin II and endothelin-1 GPCR stimulation.7,15,80,81

Pyk2 phosphorylation and activation has been postulated inseveral different pathways, including ERK1/2 signaling incardiomyocytes, p38MAPK in mesangial cells and recentlyin RhoA/Rho kinase activation via PDZ-RhoGEF vascularsmooth muscle cell migration.15,80,82,83 It has even beensuggested that tyrosine kinase pathways such as thesemay even be important in angiotensin II signaling leadingto vasoconstriction.84 These alternative signaling pathwaysin angiotensin II signaling should be a new directive for in-vestigation into increased vascular smooth muscle reactiv-ity in conditions such as hypertension. This was a focusof a recent article from Giachini et al.80 It is known that in-creased vascular reactivity to contractile stimuli is presentin vessels from deoxycorticosterone acetate (DOCA)-salthypertensive mice. Using this model with a pharmacologi-cal approach, mechanistic studies were performed to deter-mine the role that Pyk2 plays in the hyperreactivityexhibited in DOCA-salt hypertension. In mesenteric ar-teries and aorta, Pyk2 inhibition attenuated the increasedvascular constriction to phenylephrine and Western blotdata showed increased Pyk2 and phospho-Pyk2 in vesselsfrom DOCA-salt treated mice vs. sham, thus confirminga role for Pyk2 in hypertension.80

A recent discovery of a transcription factor named therepressor element 1-silencing transcriptional (REST)factor, was shown to have a consensus sequence, KCNN4,which is known to encode for the intermediate conductanceCa2þ-activated Kþ channels (IKCa). These channels are ofthe utmost importance in small resistance vessels wherea primary vasodilating agent is endothelium-derived hyper-polarizing factor. Endothelium-derived hyperpolarizing fac-tor not only remains elusive as a factor, but the mechanism bywhich it produces vasodilation is only speculation. One suchhypothesis is that Kþ efflux from endothelial cells via theseIKCa channels in coordination with small-conductanceCa2þ-activated Kþ channels, activates inward rectifier Kþ

channels. This would lead to vascular smooth muscle relax-ation. Unpublished results from the laboratory of Webb andTostes show for the first time that REST expression is closelyassociated with IKCa expression in arteries from hypertensiveanimals. Giachini et al demonstrated that REST expressionwas downregulated, whereas IKCa channels were overex-pressed in arteries obtained from spontaneously hypertensivestroke-prone rats, when compared to their Wistar-Kyotocontrols. Protein levels of small-conductance Ca2þ-activated

Page 9: Revisão - Angiotensina e Endotelina

92 B.M. Wynne et al. / Journal of the American Society of Hypertension 3(2) (2009) 84–95

Kþ channels and IKCa were also assessed, and alterationswere observed in mesenteric arteries from hypertensiveanimals. It seems as though REST may be a negative modu-latory mechanism, which controls the levels of IKCa in thespontaneously hypertensive stroke-prone rat vasculature.

As mentioned previously, the STIM molecule was shownto play an essential role for SOCE and conductance throughCRAC channels.71 The recently discovered subunit of theCRAC channel pore, termed Orai1, is essential for CRACchannel activation.62,72,85,86 However, the exact mechanismfor STIM/Orai1 signaling is not yet known. STIM can un-dergo phosphorylation at specific serine/threonine sites, aswell as undergo N-linked glycosylation.87 Sobolff et al de-termined that although STIM1 is expressed at the cell sur-face and within the endoplasmic reticulum (ER), theSTIM2 protein is expressed only intracellularly, whichlikely reflects an ER-retention signal that is present inSTIM2 but not STIM1.85–89 It was apparent that suppressedSTIM1 expression, but not STIM2, was able to preventSOCE and eliminate the store-dependent activation ofCRAC channels.71,73 Thus, the function of STIM1 is postu-lated to act as a Ca2þ sensor in the ER.72

On a final note, the question of ‘‘blame’’ for hyperten-sion is argued between two schools of thought: cardiovas-cular and renal physiologists; the former believing thathypertension is due to increased vascular resistance andan overall hyperreactivity of vascular tissue, the latter be-lieving that the kidney has the final say in BP control. Untilrecently, renal physiologists everywhere cited articles byGuyton, who was the first to clearly demonstrate the phe-nomenon of ‘‘pressure natriuresis’’ and argue for the centralrole for the kidney in BP control, and Coffman, who usesa model of AT1A receptor knockout to illustrate the neces-sity of the RAS in hypertension.90,91 These elegant studiesused the AT1 receptor knockout mice and kidney cross-transplantation to show that the AT1A receptor is crucialto basal BP regulation, as well as hypertension, and thatAT1A receptors in the kidney are paramount in hypertensionand the renal response to hypertension. However, thesestudies also demonstrated that BP regulation by AT1A re-ceptors in nonrenal tissues (ie, vasculature) were also a ma-jor contributor to systemic BP.92,93 An article recentlypublished in PNAS by Michael et al complements thesestudies, using a knocking mutant of the cGMP-dependentprotein kinase, PKGIa. PKGI is expressed in vascular andsmooth muscle cells and has been shown to regulate vascu-lar relaxation via endothelial-derived NO and other nitrova-sodilators. The most remarkable data from this study showthat the LZM-PKGIa mutant mice exhibit elevated BPs,even in the presence of normal renal function and normalrenal salt handling, suggesting an important mechanismfor vascular smooth muscle in the normal and pathophysi-ologic control of BP.94 Overall, these studies reiterate ourawareness of the complexity of hypertension, and thenecessity for systemic integration.

Conclusion

Arterial vascular smooth muscle cells constitute the ma-jority of the arterial wall, playing a foremost role in vascu-lar resistance and blood flow. Angiotensin II andendothelin-1 are potent agonists inducing contraction ofvascular smooth muscle. The contractile apparatus of vas-cular smooth muscle, actin, and myosin, can be activatedin a Ca2þ-dependent and Ca2þ-independent manner. Via li-gand binding to plasma membrane GPCRs, second messen-gers are generated and induce the release of Ca2þ throughchannels located on the plasma membrane or on the SRproducing a rapid and transient increase in intracellularCa2þ. Channels discussed in this review are activatedthrough ligand binding, store depletion, and membrane de-polarization. In the form of a Ca2þ-calmodulin complex,subsequent activation of MLCK occurs, inducing contrac-tion via actin-myosin cross-bridges. Contraction also oc-curs, Ca2þ independently, through the activation of theRhoA/Rho kinase pathway leading to MLCP inactivation,and the maintenance of contraction.

Given the varied mechanisms for smooth muscle contrac-tion, one can imagine the wide range of areas where dereg-ulation can occur, leading to increased BP or hypertension.On a molecular level, alterations in various cellular signal-ing components, can lead to over stimulation, causing anincreased and maintained vasoconstriction, decreased re-laxation, and consequently, elevation of systemic BP.

References

1. Jackson WF. Ion channels and vascular tone. Hyperten-sion 2000;35:173–78.

2. Hilgers RH, Webb RC. Molecular aspects of arterialsmooth muscle contraction: focus on Rho. Exp BiolMed (Maywood) 2005;230:829–35.

3. Woodrum DA, Brophy CM. The paradox of smoothmuscle physiology. Mol Cell Endocrinol 2001;177:135–43.

4. Wamhoff BR, Bowles DK, Owens GK. Excitation-transcription coupling in arterial smooth muscle. CircRes 2006;98:868–78.

5. Higuchi S, Ohtsu H, Suzuki H, Shirai H, Frank GD,Eguchi S. Angiotensin II signal transduction throughthe AT1 receptor: novel insights into mechanisms andpathophysiology. Clin Sci (Lond) 2007;112:417–28.

6. Ohtsu H, Suzuki H, Nakashima H, Dhobale S,Frank GD, Motley ED, et al. Angiotensin II signaltransduction through small GTP-binding proteins:mechanism and significance in vascular smooth musclecells. Hypertension 2006;48:534–40.

7. Touyz RM, Berry C. Recent advances in angiotensin IIsignaling. Braz J Med Biol Res 2002;35:1001–15.

Page 10: Revisão - Angiotensina e Endotelina

93B.M. Wynne et al. / Journal of the American Society of Hypertension 3(2) (2009) 84–95

8. Matsubara H. Pathophysiological role of angiotensin IItype 2 receptor in cardiovascular and renal diseases.Circ Res 1998;83:1182–91.

9. Harris DM, Cohn HI, Pesant S, Zhou RH, Eckhart AD.Vascular smooth muscle G(q) signaling is involved inhigh blood pressure in both induced renal and geneticvascular smooth muscle-derived models of hypertension.Am J Physiol Heart Circ Physiol 2007;293:H3072–79.

10. Wirth A, Benyo Z, Lukasova M, Leutgeb B,Wettschureck N, Gorbey S, et al. G12-G13-LARG-mediated signaling in vascular smooth muscle is re-quired for salt-induced hypertension. Nat Med 2008;14:64–8.

11. Ohtsu H, Higuchi S, Shirai H, Eguchi K, Suzuki H,Hinoki A, et al. Central role of Gq in the hypertrophicsignal transduction of angiotensin II in vascular smoothmuscle cells. Endocrinology 2008;149:3569–75.

12. Woodsome TP, Polzin A, Kitazawa K, Eto M,Kitazawa T. Agonist- and depolarization-induced sig-nals for myosin light chain phosphorylation and forcegeneration of cultured vascular smooth muscle cells.J Cell Sci 2006;119:1769–80.

13. Del Valle-Rodriguez A, Calderon E, Ruiz M, Ordonez A,Lopez-Barneo J, Urena J. Metabotropic Ca(2þ) channe-l-induced Ca(2þ) release and ATP-dependent facilitationof arterial myocyte contraction. Proc Natl Acad Sci U S A2006;103:4316–21.

14. Urena J, del Valle-Rodriguez A, Lopez-Barneo J. Me-tabotropic Ca2þ channel-induced calcium release invascular smooth muscle. Cell Calcium 2007;42:513–20.

15. Bouallegue A, Daou GB, Srivastava AK. Nitric oxideattenuates endothelin-1-induced activation of ERK1/2,PKB, and Pyk2 in vascular smooth muscle cells bya cGMP-dependent pathway. Am J Physiol Heart CircPhysiol 2007;293:H2072–79.

16. Lesh RE, Nixon GF, Fleischer S, Airey JA, Somlyo AP,Somlyo AV. Localization of ryanodine receptors insmooth muscle. Circ Res 1998;82:175–85.

17. Hirano K. Current topics in the regulatory mechanismunderlying the Ca2þ sensitization of the contractile ap-paratus in vascular smooth muscle. J Pharmacol Sci2007;104:109–15.

18. Hilgers RH, Todd J Jr, Webb RC. Increased PDZ-Rho-GEF/RhoA/Rho kinase signaling in small mesentericarteries of angiotensin II-induced hypertensive rats.J Hypertens 2007;25:1687–97.

19. Kamm KE, Murphy RA. Velocity and myosin phos-phorylation transients in arterial smooth muscle: effectsof agonist diffusion. Experientia 1985;41:1010–7.

20. Kamm KE, Stull JT. The function of myosin and myo-sin light chain kinase phosphorylation in smooth mus-cle. Ann Rev Pharmacol Toxicol 1985;25:593–620.

21. Gallagher PJ, Herring BP, Stull JT. Myosin light chainkinases. J Muscle Res Cell Motil 1997;18:1–16.

22. Chitaley K, Weber D, Webb RC. RhoA/Rho-kinase,vascular changes, and hypertension. Curr HypertensRep 2001;3:139–44.

23. Schmidt A, Hall A. Guanine nucleotide exchange fac-tors for Rho GTPases: turning on the switch. GenesDev 2002;16:1587–609.

24. Uehata M, Ishizaki T, Satoh H, Ono T, Kawahara T,Morishita T, et al. Calcium sensitization of smoothmuscle mediated by a Rho-associated protein kinasein hypertension. Nature 1997;389:990–94.

25. Miwa S, Iwamuro Y, Zhang XF, Inoki T, Okamoto Y,Okazawa M, et al. Ca2þ entry channels in rat thoracicaortic smooth muscle cells activated by endothelin-1.Jpn J Pharmacol 1999;80:281–88.

26. Yanagisawa M, Inoue A, Ishikawa T, Kasuya Y,Kimura S, Kumagaye S, et al. Primary structure, syn-thesis, and biological activity of rat endothelin, an en-dothelium-derived vasoconstrictor peptide. Proc NatlAcad Sci U S A 1988;85:6964–7.

27. Yanagisawa M, Kurihara H, Kimura S, Goto K,Masaki T. A novel peptide vasoconstrictor, endothelin,is produced by vascular endothelium and modulatessmooth muscle Ca2þ channels. J Hypertens Suppl1988;6:S188–91.

28. Miwa S, Kawanabe Y, Okamoto Y, Masaki T. Ca2þ en-try channels involved in endothelin-1-induced contrac-tions of vascular smooth muscle cells. J Smooth MuscleRes 2005;41:61–75.

29. Mohacsi A, Magyar J, Tamas B, Nanasi PP. Effects ofendothelins on cardiac and vascular cells: new thera-peutic target for the future? Curr Vasc Pharmacol2004;2:53–63.

30. Neylon CB. Vascular biology of endothelin signaltransduction. Clin Exp Pharmacol Physiol 1999;26:149–53.

31. Tirapelli CR, Casolari DA, Yogi A, Montezano AC,Tostes RC, Legros E, et al. Functional characterizationand expression of endothelin receptors in rat carotidartery: involvement of nitric oxide, a vasodilatorprostanoid and the opening of Kþ channels inETB-induced relaxation. Br J Pharmacol 2005;146:903–12.

32. Robin P, Boulven I, Desmyter C, Harbon S, Leiber D.ET-1 stimulates ERK signaling pathway through se-quential activation of PKC and Src in rat myometrialcells. Am J Physiol Cell Physiol 2002;283:C251–60.

33. Hilal-Dandan R, Ramirez MT, Villegas S, Gonzalez A,Endo-Mochizuki Y, Brown JH, et al. Endothelin ETAreceptor regulates signaling and ANF gene expressionvia multiple G-protein-linked pathways. Am J Physiol1997;272:H130–7.

34. Aramori I, Nakanishi S. Coupling of two endothelin re-ceptor subtypes to differing signal transduction intransfected Chinese hamster ovary cells. J Biol Chem1992;267:12468–74.

Page 11: Revisão - Angiotensina e Endotelina

94 B.M. Wynne et al. / Journal of the American Society of Hypertension 3(2) (2009) 84–95

35. Smith L, Payne JA, Sedeek MH, Granger JP,Khalil RA. Endothelin-induced increases in Ca2þ entrymechanisms of vascular contraction are enhanced dur-ing high-salt diet. Hypertension 2003;41:787–93.

36. Dammanahalli JK, Sun Z. Endothelin-1 InhibitsNADPH oxidase activity in human abdominal aorticendothelial cells: a novel function of ETB1 receptors.Endocrinology 2008;149:4979–87.

37. Douglas JT, Curiel DT. Strategies to accomplish tar-geted gene delivery to muscle cells employing tro-pism-modified adenoviral vectors. NeuromusculDisord 1997;7:284–98.

38. Rubanyi GM, Polokoff MA. Endothelins: molecular bi-ology, biochemistry, pharmacology, physiology, andpathophysiology. Pharmacol Rev 1994;46:325–415.

39. Komuro T, Miwa S, Zhang XF, Minowa T, Enoki T,Kobayashi S, et al. Physiological role of Ca2þ-permeablenonselective cation channel in endothelin-1-inducedcontraction of rabbit aorta. J Cardiovasc Pharmacol1997;30:504–9.

40. Dostal DE, Murahashi T, Peach MJ. Regulation of cy-tosolic calcium by angiotensins in vascular smoothmuscle. Hypertension 1990;15:815–22.

41. Kawanabe Y, Hashimoto N, Masaki T. Characterizationof Ca2þ channels involved in endothelin-1-inducedcontraction of rabbit basilar artery. J Cardiovasc Phar-macol 2002;40:438–47.

42. Yogi A, Callera GE, Montezano AC, Aranha AB,Tostes RC, Schiffrin EL, et al. Endothelin-1, but notAng II, activates MAP kinases through c-Src independentRas-Raf dependent pathways in vascular smooth musclecells. Arterioscler Thromb Vasc Biol 2007;27:1960–67.

43. Bolton TB. Mechanisms of action of transmitters andother substances on smooth muscle. Physiol Rev1979;59:606–718.

44. Hughes AD. Calcium channels in vascular smoothmuscle cells. J Vasc Res 1995;32:353–70.

45. Goto K, Kasuya Y, Matsuki N, Takuwa Y, Kurihara H,Ishikawa T, et al. Endothelin activates the dihydropyr-idine-sensitive, voltage-dependent Ca2þ channel in vas-cular smooth muscle. Proc Natl Acad Sci U S A 1989;86:3915–18.

46. Kawanabe Y, Hashimoto N, Masaki T. Involvements ofvoltage-independent Ca2þ channels and phosphoinosi-tide 3-kinase in endothelin-1-induced PYK2 tyrosinephosphorylation. Mol Pharmacol 2003;63:808–13.

47. Eto K, Ohya Y, Nakamura Y, Abe I, Iida M. Intracellu-lar angiotensin II stimulates voltage-operated Ca(2þ)

channels in arterial myocytes. Hypertension 2002;39:474–78.

48. Ohya Y, Sperelakis N. Involvement of a GTP-bindingprotein in stimulating action of angiotensin II on cal-cium channels in vascular smooth muscle cells. CircRes 1991;68:763–71.

49. Wayman CP, McFadzean I, Gibson A, Tucker JF. Twodistinct membrane currents activated by cyclopiazonicacid-induced calcium store depletion in single smoothmuscle cells of the mouse anococcygeus. Br J Pharma-col 1996;117:566–72.

50. Zhang AY, Li PL. Vascular physiology of a Ca2þ mobi-lizing second messenger - cyclic ADP-ribose. J CellMol Med 2006;10:407–22.

51. Putney JW Jr. A model for receptor-regulated calciumentry. Cell Calcium 1986;7:1–12.

52. Benham CD, Hess P, Tsien RW. Two types of calciumchannels in single smooth muscle cells from rabbit earartery studied with whole-cell and single-channelrecordings. Circ Res 1987;61:I10–6.

53. Morris AP, Gallacher DV, Irvine RF, Petersen OH. Syn-ergism of inositol trisphosphate and tetrakisphosphatein activating Ca2þ-dependent Kþ channels. Nature1987;330:653–5.

54. Kuno M, Gardner P. Ion channels activated by inositol1,4,5-trisphosphate in plasma membrane of humanT-lymphocytes. Nature 1987;326:301–4.

55. Saleh SN, Albert AP, Peppiatt CM, Large WA. Angio-tensin II activates two cation conductances with distinctTRPC1 and TRPC6 channel properties in rabbit mesen-teric artery myocytes. J Physiol 2006;577:479–95.

56. Tai K, Hamaide MC, Debaix H, Gailly P, Wibo M,Morel N. Agonist-evoked calcium entry in vascularsmooth muscle cells requires IP3 receptor-mediated ac-tivation of TRPC1. Eur J Pharmacol 2008;583:135–47.

57. Peppiatt-Wildman CM, Albert AP, Saleh SN,Large WA. Endothelin-1 activates a Ca2þ-permeablecation channel with TRPC3 and TRPC7 properties inrabbit coronary artery myocytes. J Physiol 2007;580:755–64.

58. Smyth JT, Dehaven WI, Jones BF, Mercer JC,Trebak M, Vazquez G, et al. Emerging perspectivesin store-operated Ca2þ entry: roles of Orai, STIM andTRP. Biochim Biophys Acta 2006;1763:1147–60.

59. Putney JW Jr, McKay RR. Capacitative calcium entrychannels. Bioessays 1999;21:38–46.

60. Nilius B, Droogmans G. Ion channels and their func-tional role in vascular endothelium. Physiol Rev2001;81:1415–59.

61. Parekh AB, Putney JW Jr. Store-operated calciumchannels. Physiol Rev 2005;85:757–810.

62. Leung FP, Yung LM, Yao X, Laher I, Huang Y. Store--operated calcium entry in vascular smooth muscle. Br JPharmacol 2008;153:846–57.

63. Putney JW Jr. Capacitative calcium entry revisited. CellCalcium 1990;11:611–24.

64. Putney JW Jr. Receptor-regulated calcium entry. Phar-macol Ther 1990;48:427–34.

65. Putney JW Jr. The integration of receptor-regulated in-tracellular calcium release and calcium entry across the

Page 12: Revisão - Angiotensina e Endotelina

95B.M. Wynne et al. / Journal of the American Society of Hypertension 3(2) (2009) 84–95

plasma membrane. Curr Top Cell Regul 1990;31:111–27.

66. Kasuya Y, Ishikawa T, Yanagisawa M, Kimura S,Goto K, Masaki T. Mechanism of contraction to endo-thelin in isolated porcine coronary artery. Am J Physiol1989;257:H1828–35.

67. Takemura H, Thastrup O, Putney JW Jr. Calcium effluxacross the plasma membrane of rat parotid acinar cellsis unaffected by receptor activation or by the micro-somal calcium ATPase inhibitor, thapsigargin. CellCalcium 1990;11:11–7.

68. Putney JW Jr, Broad LM, Braun FJ, Lievremont JP,Bird GS. Mechanisms of capacitative calcium entry.J Cell Sci 2001;114:2223–29.

69. Bouallegue A, Yamaguchi N. Nitric oxide inhibits thebradykinin B2 receptor-mediated adrenomedullary cate-cholamine release but has no effect on adrenal bloodflow response in vivo. J Pharmacol Sci 2005;98:151–60.

70. Venkatachalam K, van Rossum DB, Patterson RL,Ma HT, Gill DL. The cellular and molecular basis ofstore-operated calcium entry. Nat Cell Biol 2002;4:E263–72.

71. Roos J, DiGregorio PJ, Yeromin AV, Ohlsen K,Lioudyno M, Zhang S, et al. STIM1, an essential andconserved component of store-operated Ca2þ channelfunction. J Cell Biol 2005;169:435–45.

72. Zhang SL, Yu Y, Roos J, Kozak JA, Deerinck TJ,Ellisman MH, et al. STIM1 is a Ca2þ sensor that acti-vates CRAC channels and migrates from the Ca2þ storeto the plasma membrane. Nature 2005;437:902–5.

73. Liou J, Kim ML, Heo WD, Jones JT, Myers JW,Ferrell JE Jr, et al. STIM is a Ca2þ sensor essentialfor Ca2þ-store-depletion-triggered Ca2þ influx. CurrBiol 2005;15:1235–41.

74. Takahashi Y, Watanabe H, Murakami M, Ohba T,Radovanovic M, Ono K, et al. Involvement of transientreceptor potential canonical 1 (TRPC1) in angiotensinII-induced vascular smooth muscle cell hypertrophy.Atherosclerosis 2007;195:287–96.

75. Chen C, Wagoner PK. Endothelin induces a nonselec-tive cation current in vascular smooth muscle cells.Circ Res 1991;69:447–54.

76. Enoki T, Miwa S, Sakamoto A, Minowa T, Komuro T,Kobayashi S, et al. Long-lasting activation of cationcurrent by low concentration of endothelin-1 in mousefibroblasts and smooth muscle cells of rabbit aorta. Br JPharmacol 1995;115:479–85.

77. Kasuya Y, Takuwa Y, Yanagisawa M, Kimura S,Goto K, Masaki T. Endothelin-1 induces vasoconstric-tion through two functionally distinct pathways in por-cine coronary artery: contribution of phosphoinositideturnover. Biochem Biophys Res Commun 1989;161:1049–55.

78. Kawanabe Y, Hashimoto N, Masaki T. Effects of phos-phoinositide 3-kinase on endothelin-1-induced

activation of voltage-independent Ca2þ channels andvasoconstriction. Biochem Pharmacol 2004;68:215–21.

79. Schoner W. Salt abuse: the path to hypertension. NatMed 2008;14:16–7.

80. Giachini FRC, Carneiro FS, Lima VV, Carneiro ZN,Carvalho MHC, Fortes ZB, et al. Pyk2 mediates in-creased adrenergic contractile responses in arteriesfrom DOCA-salt mice. J Am Soc Hypertens 2008;2:431–38.

81. Yin G, Yan C, Berk BC. Angiotensin II signaling path-ways mediated by tyrosine kinases. Int J Biochem CellBiol 2003;35:780–83.

82. Bouallegue A, Daou GB, Srivastava AK. Endotheli-n-1-induced signaling pathways in vascular smoothmuscle cells. Curr Vasc Pharmacol 2007;5:45–52.

83. Schaller MD. Calcium-dependent Pyk2 activation:a role for calmodulin? Biochem J 2008;410:e3–4.

84. Berk BC. Angiotensin II signal transduction in vascu-lar smooth muscle: pathways activated by specific ty-rosine kinases. J Am Soc Nephrol 1999;11:S62–8.

85. Soboloff J, Spassova MA, Tang XD, Hewavitharana T,Xu W, Gill DL. Orai1 and STIM reconstitute store-op-erated calcium channel function. J Biol Chem 2006;281:20661–5.

86. Spassova MA, Soboloff J, He LP, Xu W, Dziadek MA,Gill DL. STIM1 has a plasma membrane role in the ac-tivation of store-operated Ca(2þ) channels. Proc NatlAcad Sci U S A 2006;103:4040–45.

87. Manji SS, Parker NJ, Williams RT, van Stekelenburg L,Pearson RB, Dziadek M, et al. STIM1: a novel phos-phoprotein located at the cell surface. Biochim BiophysActa 2000;1481:147–55.

88. Soboloff J, Spassova MA, Dziadek MA, Gill DL. Cal-cium signals mediated by STIM and Orai proteins—anew paradigm in inter-organelle communication. Bio-chim Biophys Acta 2006;1763:1161–68.

89. Soboloff J, Spassova MA, Hewavitharana T, He LP,Xu W, Johnstone LS, et al. STIM2 is an inhibitor ofSTIM1-mediated store-operated Ca2þ Entry. CurrBiol 2006;16:1465–70.

90. Coffman TM, Crowley SD. Kidney in hypertension:Guyton redux. Hypertension 2008;51:811–16.

91. Guyton AC. Blood pressure control–special role of thekidneys and body fluids. Science 1991;252:1813–16.

92. Coffman TM, Himmelstein S, Best C, Klotman PE.Post-transplant hypertension in the rat: effects of capto-pril and native nephrectomy. Kidney Int 1989;36:35–40.

93. Crowley SD, Gurley SB, Herrera MJ, Ruiz P, Griffiths R,Kumar AP, et al. Angiotensin II causes hypertension andcardiac hypertrophy through its receptors in the kidney.Proc Natl Acad Sci U S A 2006;103:17985–90.

94. Michael SK, Surks HK, Wang Y, Zhu Y, Blanton R,Jamnongjit M, et al. High blood pressure arising froma defect in vascular function. Proc Natl Acad Sci U SA 2008;105:6702–7.